22.02.2013 Views

Download Complete Issue (12300kb) - Academic Journals

Download Complete Issue (12300kb) - Academic Journals

Download Complete Issue (12300kb) - Academic Journals

SHOW MORE
SHOW LESS
  • No tags were found...

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

African Journal of<br />

Microbiology Research<br />

Volume 5 Number 16 18 August, 2011<br />

ISSN 1996-0808


About AJMR<br />

The African Journal of Microbiology Research is published monthly (one volume per year) by <strong>Academic</strong><br />

<strong>Journals</strong>.<br />

The African Journal of Microbiology Research (ISSN 1996-0808, IMPACT FACTOR 0.533) is an open access<br />

journal that provides rapid publication (weekly) of articles in all areas of Microbiology such as: Environmental<br />

Microbiology, Clinical Microbiology, Immunology, Viriology, Bacteriology, Phycology, Mycology and<br />

Parasitology, Protozoology, Microbial Ecology, Probiotics and Prebiotics, Molecular Microbiology,<br />

Biotechnology, Food Microbiology, Industrial Microbiology, Cell Physiology, Environmental Biotechnology,<br />

Genetics, Enzymology, Molecular and Cellular Biology, Plant Pathology, Entomology, Biomedical Sciences,<br />

Botany and Plant Sciences, Soil and Environmental Sciences, Zoology, Endocrinology, Toxicology. The Journal<br />

welcomes the submission of manuscripts that meet the general criteria of significance and scientific excellence.<br />

Papers will be published shortly after acceptance. All articles are peer-reviewed.<br />

Submission of Manuscript<br />

Submit manuscripts as e-mail attachment to the Editorial Office at: ajmr@acadjournals.org. A manuscript<br />

number will be mailed to the corresponding author shortly after submission.<br />

The African Journal of Microbiology Research will only accept manuscripts submitted as e-mail attachments.<br />

Please read the Instructions for Authors before submitting your manuscript. The manuscript files should be<br />

given the last name of the first author.


Editors<br />

Prof. Dr. Stefan Schmidt<br />

Applied and Environmental Microbiology<br />

School of Biochemistry, Genetics and Microbiology<br />

University of KwaZulu-Natal<br />

Private Bag X01<br />

Scottsville, Pietermaritzburg 3209<br />

South Africa.<br />

E-mail: ajmr.acadjourn@gmail.com<br />

Prof. Veronica Chima Nwosu (nee Dike)<br />

Department of Microbiology and Immunology<br />

Kunming Medical University<br />

Kunming 650031,<br />

China.<br />

Dr. Jianfeng Wu<br />

Dept. of Environmental Health Sciences,<br />

School of Public Health,<br />

University of Michigan<br />

USA<br />

Dr. Ahmet Yilmaz Coban<br />

OMU Medical School,<br />

Department of Medical Microbiology,<br />

Samsun,<br />

Turkey.


Editorial Board<br />

Dr. Kwang Young Song<br />

Department of Biological Engineering,<br />

School of Biological and Chemical Engineering,<br />

Yanbian Universityof Science and Technology,<br />

Yanji,<br />

China.<br />

Dr. Kamel Belhamel<br />

Faculty of Technology,<br />

University of Bejaia<br />

Algeria.<br />

Dr. Sladjana Jevremovic<br />

Institute for Biological Research<br />

Sinisa Stankovic,<br />

Belgrade,<br />

Serbia.<br />

Dr. Tamer Edirne<br />

Dept. of Family Medicine, Univ. of Pamukkale<br />

Turkey.<br />

Dr. R. Balaji Raja M.Tech (Ph.D)<br />

Assistant Professor,<br />

Department of Biotechnology,<br />

School of Bioengineering,<br />

SRM University,<br />

Chennai.<br />

India<br />

Dr. Mohd Fuat ABD Razak<br />

Institute for Medical Research<br />

Malaysia.<br />

Dr. Minglei Wang<br />

University of Illinois at Urbana-Champaign<br />

USA.<br />

Dr. Davide Pacifico<br />

Istituto di Virologia Vegetale – CNR<br />

Italy.<br />

Prof. Branislava Kocic<br />

Specaialist of Microbiology and Parasitology<br />

University of Nis, School of Medicine Institute<br />

for Public Health Nis, Bul. Z. Djindjica 50, 18000 Nis<br />

Serbia.<br />

Dr. Ntobeko A. B. Ntusi<br />

Cardiac Clinic, Department of Medicine,<br />

University of Cape Town and<br />

Department of Cardiovascular Medicine,<br />

University of Oxford<br />

South Africa and<br />

United Kingdom.<br />

Prof. N. S. Alzoreky<br />

Food Science & Nutrition Department,<br />

College of Agricultural Sciences & Food,<br />

King Faisal University,<br />

Saudi Arabia.<br />

Dr. Sivakumar Swaminathan<br />

Department of Agronomy,<br />

College of Agriculture and Life Sciences,<br />

Iowa State University,<br />

Ames, Iowa 50011<br />

USA.<br />

Dr. Alfredo J. Anceno.<br />

School of Environment, Resources and Development (SERD),<br />

Asian Institute of Technology,<br />

Thailand.<br />

Dr. Okonko, Iheanyi Omezuruike<br />

Department of Virology,<br />

Faculty of Basic Medical Sciences,<br />

College of Medicine,<br />

University of Ibadan,<br />

University College Hospital,<br />

Ibadan,<br />

Nigeria.<br />

Dr. S. Meena Kumari<br />

Department of Biosciences<br />

Faculty of Science<br />

University of Mauritius<br />

Reduit<br />

Mauritius.<br />

Luki Subehi<br />

Parasitology & Mycology Dept,<br />

Baghaeei Lab.,<br />

Shams Abadi St.<br />

Isfahan<br />

Iran.


Electronic submission of manuscripts is strongly<br />

encouraged, provided that the text, tables, and figures are<br />

included in a single Microsoft Word file (preferably in Arial<br />

font).<br />

The cover letter should include the corresponding author's<br />

full address and telephone/fax numbers and should be in<br />

an e-mail message sent to the Editor, with the file, whose<br />

name should begin with the first author's surname, as an<br />

attachment.<br />

Article Types<br />

Three types of manuscripts may be submitted:<br />

Regular articles: These should describe new and carefully<br />

confirmed findings, and experimental procedures should<br />

be given in sufficient detail for others to verify the work.<br />

The length of a full paper should be the minimum required<br />

to describe and interpret the work clearly.<br />

Short Communications: A Short Communication is suitable<br />

for recording the results of complete small investigations<br />

or giving details of new models or hypotheses, innovative<br />

methods, techniques or apparatus. The style of main<br />

sections need not conform to that of full-length papers.<br />

Short communications are 2 to 4 printed pages (about 6 to<br />

12 manuscript pages) in length.<br />

Reviews: Submissions of reviews and perspectives covering<br />

topics of current interest are welcome and encouraged.<br />

Reviews should be concise and no longer than 4-6 printed<br />

pages (about 12 to 18 manuscript pages). Reviews are also<br />

peer-reviewed.<br />

Review Process<br />

Instructions for Author<br />

All manuscripts are reviewed by an editor and members of<br />

the Editorial Board or qualified outside reviewers. Authors<br />

cannot nominate reviewers. Only reviewers randomly<br />

selected from our database with specialization in the<br />

subject area will be contacted to evaluate the manuscripts.<br />

The process will be blind review.<br />

Decisions will be made as rapidly as possible, and the<br />

journal strives to return reviewers’ comments to authors as<br />

fast as possible. The editorial board will re-review<br />

manuscripts that are accepted pending revision. It is the<br />

goal of the AJMR to publish manuscripts within weeks<br />

after submission.<br />

Regular articles<br />

All portions of the manuscript must be typed doublespaced<br />

and all pages numbered starting from the title<br />

page.<br />

The Title should be a brief phrase describing the<br />

contents of the paper. The Title Page should include the<br />

authors' full names and affiliations, the name of the<br />

corresponding author along with phone, fax and E-mail<br />

information. Present addresses of authors should<br />

appear as a footnote.<br />

The Abstract should be informative and completely selfexplanatory,<br />

briefly present the topic, state the scope of<br />

the experiments, indicate significant data, and point out<br />

major findings and conclusions. The Abstract should be<br />

100 to 200 words in length.. <strong>Complete</strong> sentences, active<br />

verbs, and the third person should be used, and the<br />

abstract should be written in the past tense. Standard<br />

nomenclature should be used and abbreviations should<br />

be avoided. No literature should be cited.<br />

Following the abstract, about 3 to 10 key words that will<br />

provide indexing references should be listed.<br />

A list of non-standard Abbreviations should be added.<br />

In general, non-standard abbreviations should be used<br />

only when the full term is very long and used often.<br />

Each abbreviation should be spelled out and introduced<br />

in parentheses the first time it is used in the text. Only<br />

recommended SI units should be used. Authors should<br />

use the solidus presentation (mg/ml). Standard<br />

abbreviations (such as ATP and DNA) need not be<br />

defined.<br />

The Introduction should provide a clear statement of<br />

the problem, the relevant literature on the subject, and<br />

the proposed approach or solution. It should be<br />

understandable to colleagues from a broad range of<br />

scientific disciplines.<br />

Materials and methods should be complete enough<br />

to allow experiments to be reproduced. However, only<br />

truly new procedures should be described in detail;<br />

previously published procedures should be cited, and<br />

important modifications of published procedures should<br />

be mentioned briefly. Capitalize trade names and<br />

include the manufacturer's name and address.<br />

Subheadings should be used. Methods in general use<br />

need not be described in detail.


Results should be presented with clarity and precision.<br />

The results should be written in the past tense when<br />

describing findings in the authors' experiments.<br />

Previously published findings should be written in the<br />

present tense. Results should be explained, but largely<br />

without referring to the literature. Discussion,<br />

speculation and detailed interpretation of data should<br />

not be included in the Results but should be put into the<br />

Discussion section.<br />

The Discussion should interpret the findings in view of<br />

the results obtained in this and in past studies on this<br />

topic. State the conclusions in a few sentences at the end<br />

of the paper. The Results and Discussion sections can<br />

include subheadings, and when appropriate, both<br />

sections can be combined.<br />

The Acknowledgments of people, grants, funds, etc<br />

should be brief.<br />

Tables should be kept to a minimum and be designed to<br />

be as simple as possible. Tables are to be typed doublespaced<br />

throughout, including headings and footnotes.<br />

Each table should be on a separate page, numbered<br />

consecutively in Arabic numerals and supplied with a<br />

heading and a legend. Tables should be self-explanatory<br />

without reference to the text. The details of the methods<br />

used in the experiments should preferably be described<br />

in the legend instead of in the text. The same data should<br />

not be presented in both table and graph form or<br />

repeated in the text.<br />

Figure legends should be typed in numerical order on a<br />

separate sheet. Graphics should be prepared using<br />

applications capable of generating high resolution GIF,<br />

TIFF, JPEG or Powerpoint before pasting in the Microsoft<br />

Word manuscript file. Tables should be prepared in<br />

Microsoft Word. Use Arabic numerals to designate<br />

figures and upper case letters for their parts (Figure 1).<br />

Begin each legend with a title and include sufficient<br />

description so that the figure is understandable without<br />

reading the text of the manuscript. Information given in<br />

legends should not be repeated in the text.<br />

References: In the text, a reference identified by means<br />

of an author‘s name should be followed by the date of<br />

the reference in parentheses. When there are more than<br />

two authors, only the first author‘s name should be<br />

mentioned, followed by ’et al‘. In the event that an<br />

author cited has had two or more works published during<br />

the same year, the reference, both in the text and in the<br />

reference list, should be identified by a lower case letter<br />

like ’a‘ and ’b‘ after the date to distinguish the works.<br />

Examples:<br />

Abayomi (2000), Agindotan et al. (2003), (Kelebeni,<br />

1983), (Usman and Smith, 1992), (Chege, 1998;<br />

1987a,b; Tijani, 1993,1995), (Kumasi et al., 2001)<br />

References should be listed at the end of the paper in<br />

alphabetical order. Articles in preparation or articles<br />

submitted for publication, unpublished observations,<br />

personal communications, etc. should not be included<br />

in the reference list but should only be mentioned in<br />

the article text (e.g., A. Kingori, University of Nairobi,<br />

Kenya, personal communication). Journal names are<br />

abbreviated according to Chemical Abstracts. Authors<br />

are fully responsible for the accuracy of the references.<br />

Examples:<br />

Chikere CB, Omoni VT and Chikere BO (2008).<br />

Distribution of potential nosocomial pathogens in a<br />

hospital environment. Afr. J. Biotechnol. 7: 3535-3539.<br />

Moran GJ, Amii RN, Abrahamian FM, Talan DA (2005).<br />

Methicillinresistant Staphylococcus aureus in<br />

community-acquired skin infections. Emerg. Infect. Dis.<br />

11: 928-930.<br />

Pitout JDD, Church DL, Gregson DB, Chow BL,<br />

McCracken M, Mulvey M, Laupland KB (2007).<br />

Molecular epidemiology of CTXM-producing<br />

Escherichia coli in the Calgary Health Region:<br />

emergence of CTX-M-15-producing isolates.<br />

Antimicrob. Agents Chemother. 51: 1281-1286.<br />

Pelczar JR, Harley JP, Klein DA (1993). Microbiology:<br />

Concepts and Applications. McGraw-Hill Inc., New York,<br />

pp. 591-603.<br />

Short Communications<br />

Short Communications are limited to a maximum of<br />

two figures and one table. They should present a<br />

complete study that is more limited in scope than is<br />

found in full-length papers. The items of manuscript<br />

preparation listed above apply to Short<br />

Communications with the following differences: (1)<br />

Abstracts are limited to 100 words; (2) instead of a<br />

separate Materials and Methods section, experimental<br />

procedures may be incorporated into Figure Legends<br />

and Table footnotes; (3) Results and Discussion should<br />

be combined into a single section.<br />

Proofs and Reprints: Electronic proofs will be sent (email<br />

attachment) to the corresponding author as a PDF<br />

file. Page proofs are considered to be the final version<br />

of the manuscript. With the exception of typographical<br />

or minor clerical errors, no changes will be made in the<br />

manuscript at the proof stage.


Fees and Charges: Authors are required to pay a $550 handling fee. Publication of an article in the African Journal of<br />

Microbiology Research is not contingent upon the author's ability to pay the charges. Neither is acceptance to pay the<br />

handling fee a guarantee that the paper will be accepted for publication. Authors may still request (in advance) that<br />

the editorial office waive some of the handling fee under special circumstances.<br />

Copyright: © 2012, <strong>Academic</strong> <strong>Journals</strong>.<br />

All rights Reserved. In accessing this journal, you agree that you will access the contents for your own personal use<br />

but not for any commercial use. Any use and or copies of this Journal in whole or in part must include the customary<br />

bibliographic citation, including author attribution, date and article title.<br />

Submission of a manuscript implies: that the work described has not been published before (except in the form of an<br />

abstract or as part of a published lecture, or thesis) that it is not under consideration for publication elsewhere; that if<br />

and when the manuscript is accepted for publication, the authors agree to automatic transfer of the copyright to the<br />

publisher.<br />

Disclaimer of Warranties<br />

In no event shall <strong>Academic</strong> <strong>Journals</strong> be liable for any special, incidental, indirect, or consequential damages of any<br />

kind arising out of or in connection with the use of the articles or other material derived from the AJMR whether or<br />

not advised of the possibility of damage, and on any theory of liability.<br />

This publication is provided "as is" without warranty of any kind, either expressed or implied, including, but not<br />

limited to, the implied warranties of merchantability, fitness for a particular purpose, or non-infringement.<br />

Descriptions of, or references to, products or publications does not imply endorsement of that product or publication.<br />

While every effort is made by <strong>Academic</strong> <strong>Journals</strong> to see that no inaccurate or misleading data, opinion or statements<br />

appear in this publication, they wish to make it clear that the data and opinions appearing in the articles and<br />

advertisements herein are the responsibility of the contributor or advertiser concerned. <strong>Academic</strong> <strong>Journals</strong> makes no<br />

warranty of any kind, either express or implied, regarding the quality, accuracy, availability, or validity of the data or<br />

information in this publication or of any other publication to which it may be linked.


International African Journal Journal of Medicine of Microbiology and Medical Research Sciences<br />

Table of Contents: Volume 5 Number 16 18 August, 2011<br />

nces<br />

Research Articles<br />

ARTICLES<br />

Isolation and characterization of a psychrophilic bacterium producing cold<br />

active lactose hydrolyzing enzyme from soil of Mt. Himalaya in Nepal 2198<br />

E. S. Nam, Y. H. Kim, K. H. Shon, and J. K. Ahn<br />

Nigerian phages: The first bacteriophages from Tropical Africa 2207<br />

Adeniran S. Koko, Hans W. Ackermann, Monisola A. Taiwo<br />

and Sunday A. Omilabu<br />

Evaluation of real-time PCR for Mycobacterium ulcerans in endemic region<br />

in Côte d’Ivoire 2211<br />

Elise Solange Ngazoa-Kakou, Euloge Ekaza, Nguetta Aka, David<br />

Coulibaly-N’Golo1, Bakary Coulibaly and Mireille Dosso<br />

Contamination of cattle carcasses by Escherichia coli shiga like toxin with<br />

high antimicrobials resistence 2217<br />

Everlon Cid Rigobelo, Renato Pariz Maluta, Clarissa Araújo Borges,<br />

Lívia Gerbasi Beraldo, Manoel Victor Franco, Lemos Sirlei<br />

Aparecida Maestá and Fernando Antonio de Ávila<br />

Determination of insecticidal toxicity of three species of entomopathogenic<br />

spore-forming bacterial isolates against Tenebrio molitor L. (Coleoptera:<br />

Tenebrionidae) 2222<br />

N. Du Rand and M. D. Laing<br />

What changed in necrotizing fasciitis in twenty-five years? 2229<br />

Baris Yildiz, Derya Karakoc, Erhan Hamaloglu, Arif Ozdemir<br />

and Ahmet Ozenc


Table of Contents: Volume 5 Number 16 18 August, 2011<br />

Table of Content: Volume 6 Number 23 21 June, 2012<br />

nces<br />

ARTICLES<br />

ARTICLES<br />

Aspects of bacterial colonization in newborn babies 2234<br />

Fariba Heshmati, Seyeed Amir Yazdanparast , Seyeed Akbar Moosavi ,<br />

Influence of ciprofloxacin on glioma cell line GL26: A new application for<br />

Hussein Dargahi, and Farnaz Tabibzadeh<br />

an old antibiotic<br />

Abdolreza Esmaeilzadeh, Massoumeh Ebtekar, Alireza Biglari and<br />

Zuhair Mohammad Hassan 4891<br />

Evaluation of anticarcinogenicity effect of Artemia urmiana by Salmonella<br />

typhimurium TA 100 strain 2241<br />

Masumeh Abbasi, Saman Mahdavi and Sedigheh Mehrabian<br />

Identification of microbial diversity in caecal content of broiler chicken<br />

S. Nathiya, G. Dhinakar Raj, A. Rajasekar, D. Vijayalakshmi and T. Devasena 4897<br />

mRNA expression of iron metabolism relation genes in macrophages by<br />

infection with Salmonella typhimurium 2245<br />

Microbial quality of some non-sterile pharmaceutical products sourced<br />

Pan Xin<br />

from some retail pharmacies in Lagos, Nigeria<br />

Adeola Anifowoshe R., Opara Morrison I. and Adeleye Isaac A. 4903<br />

Expanding drug resistance through integron acquisition in Salmonella spp.<br />

isolates obtained in Iran 2249<br />

Molecular detection of adhesins genes and biofilm formation in methicillin<br />

Bahareh Rajaei, Seyed Davar Siadat, Mohamad Reza Razavi,<br />

resistant Staphylococcus aureus<br />

Mohammad Reza Aghasadeghi, Nahid Sepehri Rad, Farzad Badmasti,<br />

Karima BEKIR, Omayma HADDAD, Mohammed GRISSA, Kamel CHAIEB,<br />

Somieh Khanjani Jafroodi, Taraneh Rajaei, Arfa Moshiri, Saifuddin Javadian<br />

Amina BAKHROUF and Salem IBRAHIM ELGARSSDI 4908<br />

The synthesis and role of hydroxyectoine in halophilic bacterium<br />

Amylase production by moderately halophilic Bacillus cereus in solid<br />

Halomonas ventosae DL7 2254<br />

state fermentation<br />

Daochen Zhu, Chenxiang Wang, Shoko Hosoi-Tanabe,<br />

P. Vijayabaskar, D. Jayalakshmi and T. Shankar 4918<br />

Weimin Zhang and Shinichi Nagata<br />

Networking clusters and sequence characteristics of clustered regularly<br />

Virulence and characteristics of a new nucleopolyhedrovirus strain of<br />

interspaced short palindromic repeats (CRISPR) direct repeats and their<br />

Dendrolimus kikuchii (Lepidoptera: Lasiocampidae) 2261<br />

evolutionary comparison with cas1 genes in lactic acid bacteria<br />

Miao Miao Yang, Meng Lou Li, Yu Zhu Wang, Liang Jian Qu, Ke Yue Huai,<br />

Kaibo Deng, Fei Liu, Chuntao Gu and Guicheng Huo 4927<br />

Xue Wen Nie, Lu Qin Qiao, Jin Ying Ding and Yong An Zhang<br />

Characterization of Staphylococcus spp strains isolated from hospital,<br />

Antibacterial screening of the root, stem and leaf extracts of Terminalia albida sc.<br />

community and environmental in Puebla city, Mexico 2266<br />

elliot on selected pathogenic bacteria<br />

Ana Marta de los Ángeles Lobo-Sánchez, Patricia Lozano-Zaraín,<br />

S. M. Ayodele, G. Alpheus and O. M. Iruaga 1457<br />

Ygnacio Martínez-Laguna, Zita Gutiérrez-Cázarez, Antonio Rivera-Tapia,<br />

Carmen Torres and Rosa del Carmen Rocha-Gracia


Table of Contents: Volume 5 Number 16 18 August, 2011<br />

nces<br />

ARTICLES<br />

Interaction of severe acute respiratory syndrome (SARS) nucleocapsid<br />

protein with macrophage migration inhibitory factor protein (MIF) 2273<br />

Yang Lin, Baohua Wang, Yijun Liu, Zhigang Yu, Manhua Cui and Haichun Ma<br />

Isolation and characterization of high caffeine-tolerant bacterium strains<br />

from the soil of tea garden 2278<br />

Fang-Yuan Fan, Yan Xu, Yue-Rong Liang, Xin-Qiang Zheng,<br />

Devajit Borthakur and Jian-Liang Lu<br />

Biopreservative potential of marine Lactobacillus spp 2282<br />

K. Indira, S. Jayalakshmi, A. Gopalakrishnan and M. Srinivasan<br />

Ephedra alata as biologically-based strategy inhibit aflatoxigenic<br />

seedborne mold 2297<br />

Al-Qarawi, A. A., Abd_Allah, E. F. and Hashem Abeer<br />

Primary resistance rates of Mycobacterium tuberculosis complex strains<br />

isolated from new tuberculosis cases: A 6-year observation 2304<br />

Servet Kayhan, Alper Akgüneş, Hikmet Tereci and Ümit Tutar<br />

Cytokine genes expression in mice hepatocytes during malaria infection 2311<br />

Saad Alkahtani, AL-Farraj S. A., Saud A. Alarifi, AL-Eissa Mohammed saad<br />

and Al-Dahmash B


Table of Contents: Volume 5 Number 16 18 August, 2011<br />

nces<br />

ARTICLES<br />

Phylogenetic analysis of the nematicidal actinobacteria from agricultural<br />

soil of China 2316<br />

Xu Chuan Kun, Lou Xiao Jun, Xi Jia Qin, Gu Lei, Duan Chang Qun, Mo Ming He,<br />

Zhang Ke Qin, Yang Fa Xiang and Fang Dun Huang<br />

A new recommended disinfectant for dental instruments 2325<br />

Jamileh bigom Taheri, Mahin Bakhshi, Sedigheh Bakhtiari, Bahare Nazemi,<br />

Fateme Fallah, Sahand Rezaie, Hamed Mortazavi and Somayyeh Azimi<br />

The functional roles of the residue tyrosine at position 26 in staphylococcal<br />

enterotoxin C2 2329<br />

Hongbo Wang, Junyi Zhou, Mingkai Xu, Huiwen Zhang and Chenggang Zhang<br />

Prevalence study of cytomegalovirus (CMV) infection among foreign<br />

manpower in Jeddah Saudi Arabia 2338<br />

N. A. Redwan, M. M. M. Ahmed and M. S. H. AL Awfi<br />

Antibacterial and cytotoxic activity of Eremurus persicus<br />

(Jaub and Spach) Boiss 2349<br />

Mojdeh Hakemi Vala, Jinous Asgarpanah, Mohammad Hossein Hedayati,<br />

Jeilan Shirali and Fatemeh Bagheri Bejestani<br />

Optimization of ultrasound-assisted extraction conditions using orthogonal<br />

matrix design to enhance the antimicrobial activity of extracts from<br />

Cichorium intybus root 2353<br />

Quanzhen Wang, Haitao Liu, Jinhong Du, Jian Cui, Guo Chen and Yuyan Liu


Table of Contents: Volume 5 Number 16 18 August, 2011<br />

nces<br />

ARTICLES<br />

Optimization of the production of exopolysaccharides by Bacillus thuringiensis<br />

27 in sand biological soil crusts and its bioflocculant activity 2359<br />

Z. R. Wang, J. P. Sheng, X. L. Tian, T. T. Wu, W. Z. Liu and L. Shen<br />

A simple and efficient method that uses low concentration fetal bovine<br />

serum to culture and purify Schwann cells 2367<br />

Han Feng, Qu Wei, Jiang Huajun, Fu Chongyang, Lu Ming, Zhang Weiguo and<br />

Lv Decheng<br />

First report on Enterobacter sakazakii from Sudanese patients 2374<br />

Humodi A. Saeed and Rania M. Musallam<br />

In search of enteroviruses in water media in Marrakech 2380<br />

Amina Hssaine, Jawhar Gharbi, Rafik Harrath, Rajae Harrak,<br />

Abderrahman Chait, Mahjoub Aouni and Jamal Hafid<br />

Laboratory analysis of a fatal meningococcal case due to serogroup B<br />

Neisseria meningitidis belonging to ST- 4821 complex 2385<br />

Zengguo Wang, Tiejun Hou, Zhijun Chen, Jinsong Li, Shouzhi Wu,<br />

Xiaoguang Wei, Yahui Sun and Quanli Du<br />

Genotyping of Hepatitis C virus (HCV) in infected patients from Saudi Arabia 2388<br />

Mohammed Ali M. Marie


African Journal of Microbiology Research Vol. 5(16), pp. 2198-2206, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR10.609<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Isolation and characterization of a psychrophilic<br />

bacterium producing cold active lactose hydrolyzing<br />

enzyme from soil of Mt. Himalaya in Nepal<br />

E. S. Nam 1 , Y. H. Kim 2 , K. H. Shon 2 , and J. K. Ahn 2 *<br />

1 Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 121-742, Republic of Korea.<br />

2 Department of Agricultural Sciences, Korea National Open University, Seoul 110-791, Republic of Korea.<br />

Accepted 18 February, 2011<br />

Psychrophilic bacteria, which grew on lactose as a carbon source, were isolated from soil of Island<br />

Peak area at Mt. Himalaya in Nepal, and identified. 148 bacterial isolates were isolated via X-gal<br />

hydrolysis on plate. Among these, three isolates, referred to as KNOUC 401, 402 and 403, displayed<br />

good cold-adapted growth and activity for hydrolysis of o-nitrophenyl- -D-galactopyranoside (ONPG)<br />

and lactose in cell free extracts. Strain KNOUC403 showing negative activity in -hemolysis was<br />

selected and identified. The strain was gram-positive bacilli, non-motile, catalase-positive and aerobic.<br />

Optimum growth was done at 20°C and pH 6.5~6.8. Anteiso-C15:0 (68.14%) was predominant in the fatty<br />

acid composition of cell. The physiological and biochemical properties, cellular fatty acid composition<br />

and sequence of 16S rDNA, suggest that strain KNOUC403 indicate a taxonomic affiliation to<br />

Arthrobacter psychrolactophilus. The optimal conditions for ONPG hydrolysis of cell free extracts from<br />

the strain were 15°C and pH 6.0, and the enzyme was stable at 4°C for 42 h. Zymogram assay of cell free<br />

extract from the strain in nondenaturing polyacrylamide gel showed a distinct band of X-gal hydrolysis.<br />

Key words: Psychrophilic bacteria, Arthrobacter psychrolactophilus, cold-active lactose hydrolyzing enzyme,<br />

Mt. Himalaya.<br />

INTRODUCTION<br />

Recently a wide diversity of cold adapted microorganisms<br />

have been found in various cold environments<br />

(Groudieva et al., 2004), and their enzymes attracted<br />

interests because of their biotechnological potential<br />

offering economical and ecological advantages such as<br />

energy saving, function in cold environment, minimizing<br />

undesirable chemical reactions that occur at high<br />

temperature, and inactivating easily by heating when<br />

required (Russell, 1988; Margesin and Schinner, 1999;<br />

Margesin et al., 2002; Cavicchioli et al., 2002). Owing to<br />

the fact that cold completely penetrates microorganisms,<br />

all components including enzymes of psychrophiles living<br />

in cold environment must be suitably adapted. Therefore<br />

psychrophiles can produce cold adapted enzymes which<br />

*Corresponding author. E-mail: ajk@knou.ac.kr. Tel: +82-2<br />

3668-4630. Fax: +82-2-3668-4187.<br />

are highly flexible in structure enabling increased<br />

complementarity between the active site and substrate at<br />

a low energy cost resulting in high catalytic activity at low<br />

temperature and labile at elevated temperature<br />

(Cavicchioli et al., 2002). A variety of useful cold active<br />

enzymes have been found in psychrophiles (Feller et al.,<br />

1997; Marshall, 1997; Gerday et al., 2000; Groudieva et<br />

al., 2004).<br />

Cold active enzymes are attractive in food industry, e.g.<br />

processing of fruit juices and milk, as there is an<br />

increasing industrial trend to treat foodstuffs under low<br />

temperature conditions in order to avoid changes in taste<br />

and nutritional value, and to save energy (Margesin and<br />

Schinner, 1994; Russel, 1998). Cold active enzyme<br />

hydrolyzing lactose to glucose and galactose at<br />

refrigerating temperature is an important food-industrial<br />

enzyme, because it can be used to produce lactose<br />

hydrolyzed milk for the lactose intolerant people, and can<br />

also be used to convert lactose in whey, a by-product of


cheese industry, to more readily fermentable glucose and<br />

galactose. Until now, some psychrophilic bacteria<br />

producing -galactosidase have been reported, but none<br />

of them have been hitherto used practically in the food<br />

industry, meaning that more study is required to find<br />

better microbial source. The aim of this study was to<br />

isolate a psychrophilic bacterium, from Mt. Himalaya of<br />

Nepal, producing the cold-active lactose hydrolyzing<br />

enzyme practically useful in food industry.<br />

MATERIALS AND METHODS<br />

Isolation and cultivation condition of microorganisms<br />

A soil sample was collected from each of the 39 regions of Island<br />

Peak area (27°55'N, 86°56'E) of 4,500 to 6,000 m height in Mt.<br />

Himalaya of Nepal by using sterilized awls, sterilized spoons and<br />

sterilized Falcon tubes. To cultivate the psychrophilic bacteria, 5 g<br />

of soil samples were added to 45 ml of Brain heart infusion (BHI;<br />

Difco Laboratories, Detroit, Mich) broth containing 1% (w/v) lactose,<br />

and incubated at 4°C aerobically by shaking (200 rpm) for 30 days.<br />

1 ml of cells grown in this enrichment were spread onto BHI agar<br />

containing 1% (w/v) lactose and 0.01% (w/v) 5-bromo-4-chloro-3indolyl-<br />

-D-galactopyranoside (X-gal; Duchefa Biochemei, Holland).<br />

After incubation at 4°C for 15 days, blue colonies were selected,<br />

then cultivated in brain heart infusion broth or agar for identification<br />

and determining properties of lactose hydrolyzing enzyme.<br />

Morphological, biochemical and physiological properties<br />

Isolates showing good growth and good X-gal hydrolysis on BHI<br />

agar, and high activity for hydrolysis of ONPG and lactose were<br />

investigated for Gram staining, morphological, biochemical, and<br />

physiological properties. Cell was grown in BHI broth to determine<br />

growth conditions of various temperatures (5 to 40°C) and pH (4.0<br />

to 8.0). pH of media was adjusted with HCl or NaOH. Acid<br />

production from carbohydrate and utilization of sole carbon sources<br />

were determined using API 20E and API 50CH test strips (Bio-<br />

Merieux Inc., France). For the test of hemolysis, a colony was<br />

streaked on sheep blood agar (KOMED. Co. Ltd). After incubation<br />

at 15°C for 3 days, -hemolytic activity was detected by lysis and<br />

complete digestion of red blood cell contents surrounding colony. All<br />

biochemical and physiological tests were done at 15°C for 3 days<br />

Composition of cellular fatty acid<br />

The cell biomass for cellular fatty acid composition analysis was<br />

collected from BHI agar plates after incubation at 15°C for 3 days.<br />

Cells were harvested, and the cellular fatty acids were saponified,<br />

methylated and extracted, following the instructions in the manual<br />

for Sherlock Microbial Identification System (MIDI, USA). The fatty<br />

acids were analyzed by gas chromatography (Hewlett Packard<br />

6890, USA) and identified using the microbial identification software<br />

package (Sasser, 1990).<br />

16S rDNA sequence determination and phylogenetic analysis<br />

Isolation of genomic DNA, PCR amplification of the 16S rDNA gene<br />

and sequencing of the purified PCR product were carried out as<br />

described by Rainey et al. (1996). Universal primers of fD1 (5’gagtttgatcctggctcag-3’)<br />

and rD1 (5’-agaaaggaggtgatccagcc-3’) were<br />

used for PCR. PCR product was purified by ethanol precipitation,<br />

Nam et al. 2199<br />

and sequenced with a model 377 Genetic Analyzer (Perkin-Elmer<br />

Co.). The 16S rDNA sequence obtained in this study was aligned<br />

against the previously determined sequences of the genus of<br />

Arthrobacter available from the Ribosomal Database Project<br />

(Maidak et al., 1996). The phylogenetic tree for the data set was<br />

inferred using the neighbor-joining method (Saitou and Nei, 1987).<br />

The PHYLIP package (Felsentein, 1993) was used for constructing<br />

the tree.<br />

Assay of lactose hydrolyzing enzyme activity<br />

Microorganism was cultivated at 4°C aerobically by shaking (200<br />

rpm) for 7 days to the late of log phase, and harvested by<br />

centrifugation at 8,000 X g for 10 min at 4°C, suspended in sodium<br />

phosphate buffer (0.01 M, pH 6.8), washed 2 times by the same<br />

buffer, suspended in the same buffer, and sonificated at 4°C. Cell<br />

debris was eliminated by centrifugation at 12,000 g and 4°C for 20<br />

min. The cell free extracts were used for assay of lactose<br />

hydrolyzing enzyme. Lactose hydrolyzing enzyme activity was<br />

determined by measuring the rate of hydrolysis of o-nitrophenyl β-<br />

D-galactopyranoside (ONPG) or lactose as substrate. The<br />

procedure of Miller (1972) for hydrolysis of ONPG was used. An<br />

aliquot of cell free extracts (0.5 ml) was added to 2.5 ml of ONPG<br />

(0.04 M) dissolved in sodium phosphate buffer (0.01 M, pH 6.8) and<br />

incubated at 4°C for 2 h. The reaction was stopped by addition of 3<br />

ml of 0.5 M Na2CO3 dissolved in H2O and the absorbance at 420<br />

nm was measured.<br />

One unit of enzyme activity for hydrolysis of ONPG was defined<br />

as the activity hydrolyzing 1 mol of ONPG per min by cell free<br />

extract from 1 ml of culture that was concentrated to 8 of A600. The<br />

hydrolysis of lactose was assayed by measuring the release of<br />

glucose from lactose. The reaction mixture consisted of 1.6 ml of<br />

skim milk (lactose conc., 4.7%) and 0.4 ml of cell free extracts. The<br />

mixture was incubated at 4°C for 5 days followed by heating in<br />

boiling water for 2 min. After centrifugation, glucose concentration in<br />

the supernatant was determined by the colorimetric method with a<br />

commercial Glucose C-II test kit (Wako Chemical Co., Japan) at<br />

505 nm. One unit of enzyme activity for hydrolysis of lactose is the<br />

activity of hydrolyzing 1 mol of lactose per day by cell free extracts<br />

from 1 ml of culture that was concentrated to 8 of A600 . Zymogram<br />

assay for the cell free extracts of a selected strain was performed.<br />

After native-PAGE of cell free extracts on 10% (w/v) polyacrylamide<br />

gel (Laemmli, 1970), the gel was stained with 0.25 mM X-gal<br />

(Trimbur et al., 1994) at 4°C for 2 h. Hydrolysis of X-Gal was<br />

confirmed by blue band within the polyacrylamide gel.<br />

Effect of temperature and pH on lactose hydrolyzing enzyme<br />

The effect of temperature on the activity of cell free extracts for<br />

ONPG hydrolysis was analyzed by measuring the enzyme activity<br />

at various temperatures (5 to 40°C) in sodium phosphate buffer<br />

(0.01 M, pH 6.8). Effect of pH on enzyme activity was evaluated at<br />

pH ranging from 4.3 to 7.8 at 4°C was used for the pH from 4.3 to<br />

6.0, and Na-phosphates buffer (0.01 M) was used for the pH from<br />

6.0 to 7.8. Stability of enzyme was investigated by residual activity<br />

during incubation of cell free extracts in sodium phosphate buffer<br />

(0.01 M, pH 6.8) for 42 h at 4 and 37°C.<br />

RESULTS<br />

Isolation of strain producing cold active lactose<br />

hydrolyzing enzyme<br />

Psychrophilic bacteria, which grow and hydrolyze lactose


2200 Afr. J. Microbiol. Res.<br />

Table 1. Physiological and biochemical properties of isolated strains.<br />

Characteristics<br />

Strains<br />

KNOUC401 KNOUC402 KNOUC403<br />

Gram staining - + +<br />

Shape Rod Rod Rod<br />

Motility - + -<br />

Spore formation - + -<br />

Optimum temp for growth 20°C 20°C 20°C<br />

Optimum pH for growth 6.8 6.5-6.8 6.5-6.8<br />

Growth at 4°C + + +<br />

Growth at 37°C + + -<br />

Catalase + + +<br />

Oxidase + + -<br />

ONPG hydrolysis + + +<br />

PNPG hydrolysis + + +<br />

Nitrate reduction - - -<br />

Gas production + - -<br />

H2S formation - - -<br />

Citrate utilization + - -<br />

Growth on MacConkey agar + - -<br />

Utilization of<br />

D-glucose + + +<br />

D-mannitol - - -<br />

D-maltose + + -<br />

D-rhamnose + + -<br />

D-mannose - - -<br />

D-galactose - - -<br />

D-lactose + + +<br />

D-fructose - - -<br />

D-adonitol - + -<br />

D-raffinose + + -<br />

D-sorbitol + + -<br />

Sucrose + - -<br />

Xylitol + + -<br />

Ducitol + + +<br />

Gelatin - - -<br />

Sorbose - - +<br />

Glycerol + + -<br />

Erythrol - - -<br />

Genus Aeromonas Bacillus Arthrobacter<br />

+, Positive reaction; -, Negative reaction.<br />

at low temperature, were isolated from the soil of Island<br />

Peak area at Mt. Himalaya in Nepal. 148 bacterial<br />

isolates were isolated via X-gal hydrolysis after soil<br />

samples were fortified in BHI broth, spread and incubated<br />

on BHI agar at 4°C. Among these, three isolates, referred<br />

to as KNOUC 401, KNOUC402 and KNOUC403, showed<br />

good growth at low temperature (4°C), formed distinct<br />

blue colony on BHI plate containing X-Gal, and their cell<br />

free extracts showed practically useful activity for<br />

hydrolysis of ONPG and lactose.<br />

Identification of strain KNOUC403<br />

The strains of KNOUC401, KNOUC402 and KNOUC403<br />

were characterized for their physiological and<br />

biochemical properties to get information on their<br />

suitability for use in the food industry. KNOUC401,<br />

KNOUC402 and KNOUC403 were presumably identified<br />

as Aeromonas, Bacillus and Athrobacter respectively by<br />

phenotypic features as in Table 1. Isolate KNOUC 403<br />

was negative for β-hemolysis on sheep blood agar, but


Table 2. Properties of preliminarily selected strains.<br />

Area (M) Strain No.<br />

Hydrolysis of<br />

ONPG (Unit 1) ) Lactose (Unit 2) )<br />

Nam et al. 2201<br />

Identification Hemolysis<br />

4700 KNOUC401 3.3394 11.54 Aeromonas +<br />

4950 KNOUC402 0.1467 1.04 Bacillus +<br />

5550 KNOUC403 0.23319 2.76 Arthrobacter -<br />

1)<br />

One unit of enzyme activity is defined as the activity hydrolyzing 1 mol of ONPG per min by cell free extract from 1 ml of culture whose<br />

A600 is 8.<br />

2)<br />

One unit of enzyme activity is the one hydrolyzing 1 mol of lactose per day at 4°C by cell free extracts from 1 ml of culture whose A600<br />

is 8.<br />

Table 3. Composition of major cellular fatty acids of<br />

strain KNOUC403.<br />

Fatty acid Contents (%)<br />

14:0 iso 5.73<br />

15:0 iso 3.14<br />

15:0 anteiso 68.14<br />

16:0 iso 8.08<br />

17:0 iso 4.21<br />

KNOUC401 and 402 were positive (Table 2). Therefore,<br />

we selected strain KNOUC403 for further study. For<br />

further identification of strain KNOUC403, the strain was<br />

tested for its cellular fatty acid composition and the<br />

sequence of 16S rDNA. The cellular fatty acids of<br />

KNOUC403 were composed of anteiso-C15:0 (68.14%),<br />

iso-C16:0 (8.08%), iso-C14:0 (5.73%), anteiso-C17:0 (4.21%),<br />

iso-C15:0 (3.14%), with additional fatty acids comprising<br />

less than 1% (Table 3).<br />

The sequence of 16S rDNA (GeneBank accession No.<br />

HQ610444) of strain KNOUC403 determined as 1,492 bp<br />

is shown in Figure 1, and the phylogenetic tree<br />

constructed by the neighbor-joining method (Felsenstein,<br />

1993) is shown in Figure 2. Comparing 16S rDNA<br />

sequence of strain KNOUC403 with the sequences in<br />

Ribosomal Data base Project (Maidak et al, 1996) and<br />

NCBI, demonstrated that strain KNOUC401 should be<br />

classified as an Arthrobacter sp., and its closest relative<br />

was Arthrobacter psychrolactophilus (98 to 100%).<br />

Therefore, strain KNOUC403 was identified as A.<br />

psychrolactophilus, and the strain was named as A.<br />

psychrolactophilus KNOUC403.<br />

Properties of lactose hydrolyzing enzyme<br />

Cell free extracts of strain KNOUC403 was tested for the<br />

properties of lactose hydrolyzing enzyme related to pH<br />

and temperature to confirm the possibility as a source of<br />

cold active lactose hydrolyzing enzyme. The optimal<br />

temperature for hydrolysis of ONPG was 15°C (Figure<br />

3a). The enzyme exhibited ca. 87% of maximal activity at<br />

4°C, and the enzyme activity decreased to less than 80%<br />

of maximal activity with a further increase in temperature<br />

to 40°C. The optimal pH was pH 6.0, it exhibited activities<br />

higher than 80% of optimum activity at broad pH from 4.3<br />

to 7.8 (Figure 3b), meaning that this strain produces cold<br />

active lactose hydrolyzing enzyme useful at wide range of<br />

pH. To determine the stability at 4 and 37°C, the cell free<br />

extracts in Na-phosphate buffer (0.01 M, pH 6.8) was<br />

incubated at those temperatures for 42 h and residual<br />

activity were tested during incubation. It was stable at<br />

4°C but lost its all activity at 37°C in 42 h (Figure 4). To<br />

confirm the existence of lactose hydrolyzing enzyme in<br />

crude cell free extracts of strain KNOUC403, zymogram<br />

assay for X-gal hydrolysis was performed. There was a<br />

distinct band of X-gal hydrolysis (Figure 5). In native<br />

electrophoresis, the enzyme moved to the spot of around<br />

130 kDa.<br />

DISCUSSION<br />

Cold-adapted lactose hydrolyzing enzymes of high<br />

activity at low temperature would be useful for hydrolysis<br />

of lactose in refrigerated milk; enabling it to be consumed<br />

by lactose-intolerant individuals, and for converting whey<br />

to a sweetener by hydrolyzing lactose to sweeter sugars<br />

of glucose and galactose. Psychrophilic bacteria living in<br />

cold environments, such as glacier silts, place of high<br />

altitude in Himalaya, Arctic soils and Antarctic environments,<br />

are potentially useful sources of cold-adapted<br />

enzymes. Among strains isolated from soil of perpetual<br />

snow area at Island peak region of Mt. Himalaya in<br />

Nepal, we selected the strain KNOUC403 showing the<br />

good growth and lactose hydrolyzing activity at 4°C, and<br />

no activity of β-hemolysis. The growth physiology of this<br />

strain, growing at 4~30°C with optimum growth<br />

temperature of 20~30°C and not growing over 37°C, is<br />

well fitted to define this strain as a psychrophile (Kocur et<br />

al., 1991). The strain KNOUC403 has phenotypic<br />

characters of Gram positive, rod, nonsporing, nonmotile ,<br />

and catalase negative, that are fitting to the genus<br />

Arthrobacter (Holt et al., 1994), as in Table 1. The


2202 Afr. J. Microbiol. Res.<br />

GATGAACGCTGGCGGCGTGCTTAACACATGCAAGTCGAACGATGAACCCCGCTTG<br />

CGGGGGGATTAGTGGCGAACGGGTGAGTAACACGTGAGTAACCTGCCCTTAACTC<br />

TGGGATAAGCCTTGGAAACGAGGTCTAATACTGGATATTGACTTTGCCTCGCATGG<br />

GGTTTGGTTGAAAGATTTATTGGTTTTGGATGGACTCGCGGCCTATCAGCTTGTTG<br />

GTGAGGTAATGGCTCACCAAGGCGACGACGGGTAGCCGGCCTGAGAGGGTGACC<br />

GGCCACACTGGGACTGAGACACGGCCCAGACTCCTACGGGAGGCAGCAGTGGGG<br />

AATATTGCACAATGGGCGAAAGCCTGATGCAGCGACGCCGCGTGAGGGATGACG<br />

GCCTTCGGGTTGTAAACCTCTTTCAGTAGGGAACAAGGCCAGCATTTTTGTTGGTT<br />

GAGGGTACTTGCAGAAGAAGCGCCGGCTAACTACGTGCCAGCAGCCGCGGTAAT<br />

ACGTAGGGCGCAAGCGTTATCCGGAATTATTGGGCGTAAAGAGCTCGTAGGCGGT<br />

TTGTCGCGTCTGCCGTGAAAGTCCGGGGCTCAACCCCGGATCTGCGGTGGGTAC<br />

GGGCAGACTAGAGTGATGTAGGGGAGACTGGAATTCCTGGTGTAGCGGTGAAATG<br />

CGCAGATATCAGGAGGAACACCGATGGCGAAGGCAGGTCTCTGGGCATTAACTGA<br />

CGCTGAGGAGCGAAAGCATGGGGAGCGAACAGGATTAGATACCCTGGTAGTCCAT<br />

GCCGTAAACGTTGGGCACTAGGTGTGGGGGACATTCCACGTTTTCCGCGCCGTAG<br />

CTAACGCATTAAGTGCCCCGCCTGGGGAGTACGGCCGCAAGGCTAAAACTCAAAG<br />

GAATTGACGGGGGCCCGCACAAGCGGCGGAGCATGCGGATTAATTCGATGCAAC<br />

GCGAAGAACCTTACCAAGGCTTGACATGAACTGGAAATACCTGGAAACAGGTGCC<br />

CCGCTTGCGGTCGGTTTACAGGTGGTGCATGGTTGTCGTCAGCTCGTGTCGTGAG<br />

ATGTTGGGTTAAGTCCCGCAACGAGCGCAACCCTCGTTCTATGTTGCCAGCACGTA<br />

ATGGTGGGGACTCATAGGAGACTGCCGGGGTCAACTCGGAGGAAGGTGAGGACG<br />

ACGTCAAATCATCATGCCCCTTATGTCTTGGGCTTCACGCATGCTACAATGGCCGG<br />

TACAATGGGTTGCGATACTGTGAGGTGGAGCTAATCCCAAAAAGCCGGTCTCAGTT<br />

CGGATTGGGGTCTGCAACTCGACCCCATGAAGTCGGAGTCGCTAGTAATCGCAGA<br />

TCAGCAACGCTGCGGTGAATACGTTCCCGGGCCTTGTACACACCGCCCGTCAAGT<br />

CACGAAAGTTGGTAACACCCGAAGCCGGTGGCCTAACCCCCTTGTGGGGAGGGA<br />

GCTGTCGAAGGTGGGACTGGCGATTGGGACTAAGTCGTAACAAGGTAGCCGTACC<br />

GGAAGGTGC<br />

Figure 1. 16S rDNA sequence of strain KNOUC403 (1,492 bp).<br />

cellular fatty acid composition profile of KNOUC403 with<br />

anteiso-C15:0 as a predominant fatty acid is typical for a<br />

member of genus Arthrobacter (Lee et al., 2003; Storms<br />

et al., 2003; Kotouckova et al., 2004; Margesin et al.,<br />

2004). Heyrman et al. (2005) reported that predominant<br />

fatty acid of the genus Arthrobacter was anteiso-C15:0 and<br />

iso-C15:0. The predominant fatty acids of A.<br />

psychrolactophilus (sp. nov.) was also anteiso-C15:0 that<br />

comprised 73% of total fatty acids (Loveland-Curtze et<br />

al., 1999). Therefore “Loveland-Curtze et al. (1999) it has<br />

been reported that several Arthrobacter strains produced<br />

cold-active -galactosidase (Coker et al., 2003;<br />

Nakagawa et al., 2003). Arthrobacter sp. 20B -<br />

galactosidase was optimally active at pH 6.0 to 8.0 and


Nam et al. 2203<br />

Figure 2. Phylogenetic tree based on 16S rDNA sequence showing the position of strain KNOUC403 as the type strain of Arthrobacter<br />

species and the representative of some other related taxa. Scale bar represents 0.01 substitutions per nucleotide.<br />

Relative Relative activity(%) activity (%)<br />

100<br />

90<br />

80<br />

70<br />

60<br />

0 10 20 30 40<br />

Temperature Temp.<br />

A


2204 Afr. J. Microbiol. Res.<br />

Relative actitvity(%)<br />

100<br />

90<br />

80<br />

70<br />

60<br />

Na-acetate buffer (0.01M)<br />

4 5 6 7 8<br />

pH<br />

B<br />

Na-phosphate buffer (0.01 M)<br />

Figure 3. Effects of temperature (A) and pH (B) on the activity of lactose hydrolyzing enzyme in cell free<br />

extracts of Arthrobacter psychrolactophilus KNOUC403 for the hydrolysis of ONPG. *Effect of temperature on<br />

enzyme activity was analyzed in Na-phosphate buffer (0.01 M, pH 6.8). *Values are means of triplicates ± S.D.<br />

*Effect of pH on enzyme activity was evaluated at 4°C in Na-acetate buffer (0.01 M, pH 4.3 to 6.3) and Naphosphate<br />

buffer (0.01 M, pH 5.8 to 7.8), *values are means of triplicates ± S.D.<br />

Residual activity(%)<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

0 6 12 18 24 30 36 42 48<br />

Hours<br />

Figure 4. Stability of lactose hydrolyzing enzyme in cell free extracts of Arthrobacter<br />

psychrolactophilus KNOUC403 at 4°C ( ) and 37°C ( ) in Na-phosphate buffer (0.01 M, pH<br />

6.8). *Values are means of triplicates ±S.D.<br />

25°C (Bialkowska et al., 2009).<br />

A -galactosidase of Arthrobacter sp. obtained from<br />

Antarctic dry valley soil was cold optimal at 18°C, showed<br />

50% activity of optimum activity at 0°C, and was an<br />

unstable one, inactivated within 10 min at 37°C (Coker et<br />

al., 2003). Arthrobacter reported by Nakagawa et al.<br />

(2006) produced cold active -galactosidase with<br />

optimum temperature of 10°C and optimum pH of 8.0.<br />

However, two -galactosidases of Pseudoalteromonas<br />

sp., a psychrophile, were not cold active enzymes whose<br />

optimum temperature were 26°C (Fernandes et al., 2002)<br />

and 45°C (Hoyoux et al., 2001) showing that all


KDa Ma 1 2<br />

X-gal hydrolyzing enzyme<br />

Figure 5. Native polyacylamide electrophoresis and zymogram of cell free extracts of<br />

strain Arthrobacter psychrolactophilus KNOUC 403. The protein was analyzed on a<br />

10% polyacrylamide gel. Ma: Kaleidoscope polypeptide standards marker. 1: The gel<br />

was stained with Coomassie Brilliant Blue R-250. 2: The gel was soaked in 0.1 M Xgal<br />

solution (in Z-buffer) at 4°C for 2 h.<br />

psychrophiles do not always produce cold active enzyme.<br />

The other -galactosidases of Athrobacter sp. B7<br />

(Trimbur et al., 1994) and Arthrobacter sp. C2-2<br />

(Karasova-Lipovova et al., 2003) were characterized also<br />

by high optimum temperature of 40°C. The lactose<br />

hydrolyzing enzyme in cell free extracts of strain<br />

KNOUC403 shows high activity at 4°C and in wide range<br />

of pH 4.3 to 7.8, is stable at 4°C, and has a moderate but<br />

valuable stability at 37°C, meaning that this strain has<br />

high possibility of producing cold active lactose<br />

hydrolyzing enzyme that is useful for hydrolysis of lactose<br />

in foods of wide range of pH at refrigerating temperature.<br />

Judging from these findings of physiological and<br />

enzymatic properties, A. psychrolactophilus KNOUC403<br />

is a psychrophile of useful source, for a cold active<br />

lactose hydrolyzing enzyme that would have advantages<br />

for application in the food industry.<br />

ACKNOWLEDGEMENTS<br />

This research was performed by the support of the Korea<br />

Institute of Planning and Evaluation for Technology of<br />

Food, Agriculture and Fishery. We thank the College of<br />

Agriculture and Life Science Alpine Association, Seoul<br />

National University for sampling soils at the area of Island<br />

Peak in Himalaya.<br />

REFERENCES<br />

Bialkowska A, Cieslinski H, Nowakowska KM, Kur J, Turkiexicz M<br />

(2009). A new -galactosidase with a low temperature optimum<br />

Nam et al. 2205<br />

isolated from the Antarctic Arthrobacter sp. 20B: gene cloning,<br />

purification and characterization. Arch. Microbiol., 191: 825-835.<br />

Cavicchioli R, Siddiqui KS, Andrews D, Sowers KR (2002). Lowtemperature<br />

extremophiles and their applications. Curr. Opin.<br />

Biotechnol., 13: 253-261.<br />

Coker J, Peter P, Loveland J, Brenchley JE (2003). Biochemical<br />

characterization of a -galactosidase with a low temperature optimum<br />

obtained from an Antarctic Arthrobacter isolates. J. Bacteriol., 185:<br />

5473-5482.<br />

Feller G, Zekhnini Z, Lamotte-Brasseur J, Gerday C (1997). Enzymes<br />

from cold-adapted microorganisms. The class C -lactamase from<br />

the Antarctic psychrophile Psychrobacter immobilis A5. Eur. J.<br />

Biochem., 244: 186-191.<br />

Felsenstein J (1993). PHYLIP: Phylogenetic Inference Package, version<br />

3.5. Department of Genetics, University of Washington, Seattle, WA,<br />

USA.<br />

Fernandes S, Geueke B, Delgado O (2002). -galactosidase from a<br />

cold-adapted bacterium; purification, characterization and application<br />

for lactose hydrolysis. Appl. Microbiol. Biotechnol., 58: 313-321.<br />

Gerday C, Aittaleb M, Bentahir M, Chessa JP, Claverie P, Collins T,<br />

D’Amico SM, Dumont J, Garsoux G, Georlette D, Hoyoux A,<br />

Lonhienne T, Meuwiw MA, Feller G (2000). Cold-adapted enzymes:<br />

from fundamentals to biotechnology. Trends Biotechnol., 18: 103-107.<br />

Groudieva T, Kambourova C, Yusef C, Royter C, Grote C, Trinks C,<br />

Antranikian C (2004). Diversity and cold-active hydrolytic enzymes of<br />

culturable bacteria associated with Arctic sea ice, Spitzbergen.<br />

Extremophiles, 8(6): 475-488.<br />

Heyrman J, Verbeeren J, Schumann P, Swings J, De Vos P (2005). Six<br />

novel Arthrobacter species isolated from deteriorated mural paintings.<br />

In. J. Syst. Evol. Microbiol., 55: 1457-1464.<br />

Holt JG, Krieg NR, Sneath PH, Staley JT, Williams ST (1994). Bergey’s<br />

manual of determinative bacteriology. 9 th Ed. Williams & Wilkins,<br />

Baltimore.<br />

Hoyoux A, Jennes I, Dubiose P, Genicot S, Dubaik F, Francoi JM<br />

(2001). Cold-adapted -galactosidase from the Antarctic psychrophile<br />

Pseudoalteromonas haloplanktis. Appl. Environ. Microbiol., 67: 1529-<br />

1535.<br />

Karasova-Lipovova P, Strnad H, Spiwok VMS, Kralova B, Russell NJ<br />

(2003). The cloning, purification and characterization of a cold-active


2206 Afr. J. Microbiol. Res.<br />

-galactosidase from the psychrotolerant Antarctic bacterium<br />

Arthrobacter sp. C2-2. Enzyme Microb. Technol., 33: 836-844.<br />

Kocur M, Kloos WE, Schleifer KH (1991). The genus Micrococcus. In:<br />

Balows A., Truper H.G., Dworkin M., Harder W., Scheifer K.H.(Eds.)<br />

The prokaryotes, Springer, Berlin Heidelberg New York, 2: 1300-<br />

1311.<br />

Kotouckova L, Schumann P, Durnova E, Sproer C, Sedlacek I, Neca J,<br />

Zdrahal Nemec M (2004). Arthrobacter nitroguajacolicus sp. nov., a<br />

novel 4-nitroguaiacol-degrading Actinobacterium. Int. J. Syst. Evol.<br />

Microbiol., 54: 773-777.<br />

Laemmli UK (1970). Cleavage of structural proteins during the<br />

assembly of the head of bacteriophage T4. Nature., 227: 680-685.<br />

Lee JS, Lee KC, Pyun YR, Bae KS (2003). Arthrobacter koreensis sp.<br />

nov., a novel alkalitolerant bacterium from soil. Int. J. Syst. Evol.<br />

Microbiol., 53: 1277-1280.<br />

Loveland-Curtze J, Sheridan PP, Gutshall KR, Brenchle JE (1999).<br />

Biochemical and phylogenetic analyses of psychrophilic isolated<br />

belong to the Arthrobacter subgroup and description of Arthrobacter<br />

psychrolactophilus sp. nov. Arch. Microbiol., 171: 355-363.<br />

Maidak BL, Olsen GJ, Larson N, Overbeck R, MaCoughey MJ, Woese<br />

CR (1996). The Ribosomal Database Project (RDP). Nucleic Acids<br />

Res., 24: 82-85.<br />

Margesin R, Schinner F (1994). Properties of cold-adapted<br />

microorganisms and their potential role in biotechnology. J.<br />

Biotechnol., 33: 1-14.<br />

Margesin R, Schinner F (1999). Biotechnological applications of coldadapted<br />

organisms. Berlin; Springer, pp. 1-16<br />

Margesin R, Feller G, Gerday C, Russel NJ (2002). Cold-adapted<br />

microorganisms; adaptation strategies and biotechnological potential.<br />

In The Encyclopedia of Environmental Microbiology, Edited by G.<br />

Bitton. New York: Wiley, 2: 871-885.<br />

Margesin R, Schumann P, Sproer C, Counot AM (2004). Arthrobacter<br />

psychrophenolicus sp. nov., isolated from an alpine ice cave. Int. J.<br />

Syst. Evol., 54: 2067-2072.<br />

Marshall CJ (1997). Cold-adapted enzymes. Trends Biotechnol., 15:<br />

359-364.<br />

Miller JH (1972). Experients in molecular genetics. Cold Spring Harbor<br />

Laboratory, Cold Spring Harber, N.Y.<br />

Nakagawa T, Fujimoto Y, Uchino M (2003). Isolation and<br />

characterization of psychrophiles producing cold-active -<br />

galactosidase. Lett. Appl. Microbiol., 37: 154-157.<br />

Nakagawa T, Fujimoto Y, Ikehata R, Miyaji T, Tomizuka N (2006).<br />

Purification and molecular characterization of cold-active -<br />

galactosidase from Arthrobacter psychrolactophilus strains F2. Appl.<br />

Microbiol. Biotechnol., 72: 720-725.<br />

Rainey FA, Ward-Rainey N, Kroppenstedt RM, Stackebrandt E (1996).<br />

The genus Nocardiopsis represents a phylogenetically coherent<br />

taxon and a distinct actinomycete lineage: proposal of<br />

Nocardiopsacease fam. nov. Int. Syst. Bacteriol., 46: 1088-1092.<br />

Russell NJ (1998). Molecular adaptations in psychrophilic bacteria:<br />

potential for biotechnological applications. Adv. Biochem. Engin.<br />

Biotechnol., 61: 1-21.<br />

Saitou N, Nei M (1987). The neighbor-joining method: a new method for<br />

reconstruction phylogenetic trees. Mol. Biol. Evol., 4: 406-424.<br />

Sasser M (1990). Identification of bacteria by gas chromatography of<br />

cellular fatty acids. MIDI Technical. Note Newark. DE: MIDI Inc., USA,<br />

p. 101.<br />

Storms V, Devries LA, Coopman R, Schumann P, Vyncke F, Gillis M<br />

(2003). Arthrobacter gandavensis sp. nov. for strains of veterinary<br />

origin. Int. J. Syst. Evol. Microbiol., 53: 1881-1884.<br />

Trimbur DE, Gutshall KR, Prema P, Brenchley JE (1994).<br />

Characterization of a psychrophilic Arthrobacter gene and its coldactive<br />

-galactosidase. Appl. Environ. Microbiol., 60: 4554-4552.


African Journal of Microbiology Research Vol. 5(16), pp. 2207-2210, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR10.647<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Nigerian phages: The first bacteriophages from<br />

Tropical Africa<br />

Adeniran S. Koko 1,3 , Hans W. Ackermann 2 *, Monisola A. Taiwo 1 and Sunday A. Omilabu 1<br />

1 Department of Botany and Microbiology, University of Lagos, Lagos, Nigeria.<br />

2 Félix d'Hérelle Reference Center for Bacterial Viruses, Department of Microbiology, Faculty of Medicine,<br />

Laval University, Québec, QC, G1K 7P4, Canada.<br />

3 Quest Diagnostics West Hills, CA 91304, CA, USA.<br />

Accepted 28 March, 2011<br />

Fifteen morphological bacteriophage types were observed in enrichment cultures of sewage, surface<br />

water and human feces collected in Lagos, Nigeria. All phages were tailed and belonged to the<br />

Myoviridae, Siphoviridae or Podoviridae families. Most phages could be assigned to well-known<br />

morphotypes. Ten of them corresponded to phages of enterobacteria or pseudomonads. Four<br />

morphotypes (φKZ, T4 and χ, Jersey) prevailed in most samples. Most others occurred in traces only.<br />

Key words: Bacteriophages, ecology, identification, morphotypes.<br />

INTRODUCTION<br />

Phage ecology has essentially been investigated in North<br />

America, Europe and the oceans. Research into phage<br />

ecology in other parts of the world is almost nonexistent.<br />

As a result the phage flora of vast geographic areas,<br />

namely most of South America, Africa, Australia, China,<br />

India, all of Middle America, the Middle East and Siberia<br />

remains essentially unknown. The only reports on phages<br />

from African environments are limited to tailed phages in<br />

the subsoil of the Moroccan and Tunisian Sahara (Prigent<br />

et al., 2005) and the Namibian desert (Prestel et al.,<br />

2008), RNA phages in sewage in South Africa (Grabow<br />

et al., 1993; Schaper et al., 2002), and unknown phages<br />

of nonidentified bacteria in Senegal (Bettarel et al., 2006).<br />

The Saharan phages were observed by electron<br />

microscopy, but not propagated or identified. The<br />

Namibian phages were observed by electron microscopy<br />

in bulk cultures, but they were not isolated and only one<br />

of the morphotypes present was identified. In addition,<br />

there are isolated reports of specific phages in various<br />

parts of the world, for example Vibrio phages from shrimp<br />

fisheries in Thailand (Pasharawipas et al., 2005), Bacillus<br />

*Corresponding author. E-mail: ackermann@mcb.ulaval.ca. Tel:<br />

+418 656-2131; 2558. Fax +418 656-7555.<br />

phages from deep sea wells in China (Liu et al., 2006),<br />

phages of particular actinomycetes in Australia (Kurtböke<br />

et al., 1993a, 1993b), and Thermus phages in Kamchatka<br />

hot springs (Yu et al., 2006).<br />

This situation is deeply insatisfactory from an ecological<br />

viewpoint. We intended to identify and diagnose our<br />

phages as precisely as possible. This is the first<br />

investigation of the bacteriophage flora in tropical Africa.<br />

MATERIALS AND METHODS<br />

We collected samples of sewage and surface water at the<br />

University of Lagos and in the Iddo area of the Lagos Lagoon, a<br />

natural lagoon of brackish water connected to the Atlantic Ocean<br />

through creeks. Nineteen water samples and one of human feces<br />

were enriched in Lagos, often three to four times in parallel, with<br />

common bacteria isolated from human wound and skin infections in<br />

Lagos hospitals (Bacillus spp., Escherichia coli, Klebsiella<br />

pneumoniae, Pseudomonas aeruginosa, Staphylococcus aureus<br />

and Xanthomonas sp.). Enrichment cultures were filtered, but single<br />

plaque purification was not attempted. Samples were examined in<br />

Canada by one of us (H.-W.A.). Approximately 75 enrichment<br />

cultures of 1 ml each were received. Although all vials were intact<br />

and none had leaked during transportation, all were heavily<br />

contaminated by viable enterobacteria and pseudomonads. This<br />

contamination had clearly occurred in Nigeria before shipment.<br />

Samples were too small for filtration and were clarified by simple<br />

decantation during storage for 1 month. Supernatants were<br />

centrifuged at 25,000 g using a Beckman J2-21 centrifuge and a


2208 Afr. J. Microbiol. Res.<br />

Table 1. Bacteriophage observations by sample.<br />

Sample Bacteriophage morphotype<br />

A41 φKZ Lu11 T4 χ χ (chi) Jersey N4 T7 Others<br />

1 * + + + + φ29, 44AJHD<br />

2 ++ + (+) * +<br />

3 +++ (+) *<br />

4 ++ * * *<br />

5 * ++ * * *<br />

6 + ++ *<br />

7 * ** * M1, S1<br />

8 ++ * ** * +<br />

9 * ++ + + ++ *<br />

10 ** + + ++<br />

11 * + * ++<br />

12 ** * ++ * * M1, S1<br />

13 * + * *<br />

14 + + * * ++ 121<br />

15 + * + +++<br />

Iddo (+) * + * φ29<br />

OPP1 * * * *<br />

Berger (+) +<br />

Unilag (+) * (+) S2<br />

Feces * + * * (+) * T5, φ29<br />

+++, ++, +, (+), *: Variable quantities of phages; +++, many; *, traces. M1, S1, S2: Novel morphotypes of Myoviridae (M) and Siphoviridae (S) families.<br />

Table 2. Main dimensions of bacteriophage phage morphtypes.<br />

Family Type Head, nm Tail, nm<br />

YerA41 130 235 x 20<br />

φKZ 121 190 x 21<br />

Myoviridae<br />

Lu11<br />

121<br />

121<br />

116<br />

190 x 21<br />

115 x 17<br />

M1 133 156 x 17<br />

T4 111 x 78 113 x 16<br />

Siphoviridae<br />

Podoviridae<br />

χ (chi) 63 227 x 10<br />

Jersey 68 160 x 8<br />

T5 78 183 x 8<br />

S1 99 664 x 8<br />

S2 90 237 x 8<br />

N4 70 10<br />

T7 60 17 x 8<br />

φ29 54 x 42 36 x 12<br />

44JHD 45 35 x 12<br />

JA-18.1 fixed-angle rotor and were washed twice with 0.1 M<br />

ammonium acetate (pH 7.0). Sediments were deposited on copper<br />

grids with carbon-coated Formvar films, stained with 2% potassium<br />

phosphotungstate (pH 7.0) and examined in a Philips EM 300<br />

electron microscope. Magnification was calibrated by means of T4<br />

phage tails (length 114 nm).<br />

RESULTS AND DISCUSSION<br />

Fifteen morphological bacteriophage types, listed roughly<br />

by family and size in Tables 1 and 2, were observed. All<br />

were tailed and had contractile, long and noncontractile<br />

or short tails. They were thus identified as members of<br />

the Myoviridae, Siphoviridae or Podoviridae families,<br />

respectively. Twelve morphotypes (shortly "types")<br />

corresponded to well-known phages described in earlier<br />

reviews or books to which the reader is referred<br />

(Ackerrmann and DuBow, 1987; Ackermann, 1987). Part<br />

of the corresponding phage groups (φKZ, T4, T7, N4, φ29<br />

and AJHD) relate to phage genera or subfamilies defined<br />

by the International Committee on Taxonomy of Viruses<br />

or ICTV (Hendrix and Casjens, 2005; Lavigne et al.,<br />

2008; 2009). Ten types corresponded to known<br />

enterobacterial and Pseudomonas bacteriophage species<br />

and only two corresponded to species of Bacillus or<br />

Staphylococcus phages (podovirus types φ29 and<br />

44AHJD, respectively). Most preparations were mixtures<br />

of 4 to 5, even 7 phage morphotypes (Table 1),<br />

suggesting that none of the bacteria used for enrichment<br />

had been pure. Most phage morphotypes could be<br />

related to well-known viruses with specific hosts. Four<br />

morphotypes were almost ubiquitous, namely : φKZ, T4,<br />

Jersey and χ (chi). Others occurred in traces only and<br />

were detected by intense scrutinizing only. This is not an


indication of their frequency or rarity in nature. No exact<br />

quantitation is possible here because the apparent<br />

frequency of phages in enrichment cultures depends on<br />

many variables (phage inoculum, host sensitivity, vitality<br />

of phages and nutrients), all of which may lead to<br />

overcrowding of some phages by others.<br />

Part of this may have occurred during transportation<br />

since, as noted earlier, no sample was sterile on arrival in<br />

Canada. Finally, the very procedure of negative staining<br />

often leads to uneven distribution of particles and<br />

gathering of different particles in different places of<br />

electron microscopical grids. Three particle types (M1,<br />

S1, S2; myovirus 1, siphovirus 1 and 2) could not be<br />

attributed to known bacteriophages and may represent<br />

novel species. Type S1 is particularly interesting. Types<br />

M1 and S2 are morphologically unremarkable and not<br />

illustrated for lack of space.<br />

Myoviridae<br />

Morphotype A41 corresponds to a giant phage of this<br />

name, observed in Yersinia enterocolitica in Canada<br />

(Stevenson and Airdrie, 1984). It has so far not been<br />

found elsewhere. Types φKZ and Lu11 correspond to<br />

giant Pseudomonas phages distinguished by the<br />

presence of long, curly tail fibers in the latter (Figure 2).<br />

Phages of type φKZ were previously isolated in Denmark,<br />

Germany, Georgia, Russia and the U.S.A. Type Lu11 is<br />

known from the Philippines only (Krylov et al., 2007). The<br />

relative frequency of the φKZ and Lu11 types is not<br />

surprising as representatives of these types multiply<br />

extremely well and may conceivably overcrowd other<br />

phages in mixed cultures. Enterobacterial phages of type<br />

121 were first observed in Romania (Nacesco et al.,<br />

1969) and later in Canada by one of us (Ackermann and<br />

Nguyen, 1983). Type 121 is characterized by a large<br />

head and a relatively short tail. Type T4, easily<br />

recognized by its elongated head and a tail with six long<br />

tail fibers, seems to be truly ubiquitous and has been<br />

reported in many countries (Ackermann and Krisch,<br />

1997) (Figures 1 to 4).<br />

Type M1 does not, to our knowledge, correspond to<br />

any myovirus known. As types A41, φKZ, Lu11 and 121,<br />

It has an unusually large head.<br />

Siphoviridae<br />

Phages of the χ (chi) type are characterized by a<br />

relatively small head and a thick tail equipped with a<br />

single long, wavy tail fiber. The original phage χ is<br />

specific to mobile bacteria of E. coli, Salmonella and<br />

Serratia (Meynell, 1961). The tail fiber is difficult to see<br />

and apparently easily removed by centrifugation. Despite<br />

their relatively thick tails, χ and its relatives are<br />

Koko et al. 2209<br />

Figure 1-4. Myoviridae, morphotypes A41,<br />

T4, φKZ, and LU11, respectively.<br />

Phosphotungstate, final magnification x<br />

297,000; bars represent 100 nm.<br />

siphoviruses. Type Jersey has a conspicuous base plate<br />

with six spikes. It was first found in Salmonella paratyphi<br />

B and seems to be restricted to Salmonella bacteria.<br />

However, the orginal phage Jersey has a tail of 116 nm in<br />

length only (Ackermann and Berthiaume, 1969) while the<br />

Jersey-like particles here have tails of much longer length<br />

(Figures 5 to 6). It may be speculated that they are tail<br />

length variants of the normal phage Jersey, phage Jersey<br />

in nature. Type T5 is indistinguishable from coliphage T5.<br />

Type S1 is morphologically unremarkable, but apparently<br />

has not been encountered elsewhere and may be novel.<br />

Type S2 is very interesting as it displays a single, rigid,<br />

thick, spiral tail fiber of approximately 200 nm in length.<br />

The spirals have a periodicity of 47 nm. Prestel et al.<br />

(2008) observed similar particles in the sand of the<br />

Namibian desert.<br />

Podoviridae<br />

The N4 morphotype shows bundles of short tail fibers<br />

laying at the sides of the tail. Phages of this type seem to<br />

be very frequent in enterobacteria. Some of them<br />

arespecific to capsulated bacteria, for example, Klebsiella<br />

and Vi antigen-containing Salmonella bacteria. However,<br />

the rather uncharacteristic morphology of these phages<br />

suggests that "type N4" is heterogeneous and comprises<br />

several genomically unrelated phages. The T7 type is<br />

very frequent in Gram-negative bacteria, especially<br />

enterics, pseudomonads and vibrios. It seems to be<br />

ubiquitous and to have a world-wide distribution. So far,


2210 Afr. J. Microbiol. Res.<br />

Figures 5-10: 5-6, Siphoviridae; 7-10, Podoviridae. 5,<br />

morphotypes χ (chi, left) and Jersey (right). 6, type Sx with<br />

long spiral tail fiber and an adventitious particle of type φKZ.<br />

7-10, Podoviridae. Types T7, N4, 44AJHD, and φ29,<br />

respectively. Phosphotungstate. Final magnification x<br />

148,500 in Figure. 6 and x 297,000 in all others; bars<br />

represent 100 nm.<br />

all sequenced T7-like phages have shown genomic<br />

relationships (Lavigne et al., 2008). The types<br />

represented by Bacillus phage φ29 and Staphylococcus<br />

phage 44AJHD occur in traces only. They differ in head<br />

shape (prolate in φ29 and isometric in 44AJHD), but<br />

share genomic relationships (Lavigne et al., 2008) and a<br />

particular tail structure with a collar and collar<br />

appendages. The phages represent two different genera<br />

of the same phage subfamily, the Nanovirinae (Lavigne<br />

et al., 2008). Type φ29 is ubiquitous and occurs not only<br />

in bacilli, but also in Kurthia and Streptococcus bacteria.<br />

Phages of types A41, φKZ, Lu11, 121, T4, χ, Jersey, T7,<br />

φ29 and 44AJHD are known to be virulent. This indicates<br />

that most phages here reported derive from lytic phage<br />

infections in the environment. At least eight types (φKZ,<br />

121, T4, χ, Jersey, N4, T7 and φ29) seem to have a<br />

global distribution (Figures 7 to 10). This is consistent<br />

with metagenomic studies reporting the occurrence of T4<br />

and T7 genes in the Arctic Ocean, the Gulf of Mexico and<br />

the Sargasso Sea (Angly et al., 2006) but remains to be<br />

ascertained by further ecological studies, especially in<br />

Third-World countries. It should be interesting to study<br />

the geographical distribution of phages and to investigate<br />

whether all phages of similar habitats occur world-wide or<br />

are limited to certain regions.<br />

Electron microscopy for its rapidity and wealth of<br />

information it generates appears as the technique of<br />

choice for these investigations. We believe that we are<br />

just at the threshold of phage ecology and we have<br />

experienced that each ecological investigation of phage<br />

habitats in any country is likely to yield new and<br />

interesting phages.<br />

REFERENCES<br />

Ackermann HW, Berthiaume L (1969). Ultrastructure des phages de lysotypie<br />

des Escherichia coli 0127:B8. Can. J. Microbiol., 15: 859-862.<br />

Ackermann HW, DuBow MS (1987). Viruses of Prokaryotes. Vol II.<br />

Natural Groups of Bacteriophages. CRC Press, Boca Raton, FL, pp.<br />

72-121.<br />

Ackermann HW, DuBow MS, Gershman M, Karska-Wysocki B,<br />

Kasatiya SS, Loessner MJ, Mamet-Bratley MD, Regué M (1997).<br />

Taxonomic changes in tailed phages of enterobacteria. Arch. Virol.<br />

142:1381-1390.<br />

Ackermann HW, Krisch H (1997). A catalogue of T4-type<br />

bacteriophages. Arch. Virol. 142:2329-2345.<br />

Ackermann HW, Nguyen TM (1983). Sewage coliphages investigated<br />

by electron microscopy. Appl. Environ. Microbiol. 45:1049-1059.<br />

Angly FE, Felts B, Breitbart M, Salamon P, Edwards RA, Carlson C,<br />

Chan AM, Haynes M, Kelley S, Liu H, Mahaffy JM, Mueller JE, Nulton<br />

J, Olson R, Parsons R, Rayhawk S, Suttle CA, Rohwer F (2006). The<br />

marine viromes of four oceanic regions. PloS Biol. 4:2121-2131 (e368).<br />

Bettarel Y, Bouvy M, Dumont C, Sime-Ngando T (2006). Virusbacterium<br />

interactions in water and sediment of West African inland<br />

water system. Appl. Environ. Microbiol. 72:5274-5282.<br />

Grabow W, Holtzhausen C, de Villiers C (1993). Research on<br />

bacteriophages as indicators of water quality. WRC Report No.<br />

321/1/93, Water Research Commission, Pretoria, South Africa.<br />

Hendrix RW, Casjens SR (2005). Caudovirales. In: Fauquet et al. (eds)<br />

Virus Taxonomy: VIIIth Report of the International Committee on<br />

Taxonomy of Viruses, Elsevier <strong>Academic</strong> Press. pp. 35-42.<br />

Kurtböke DI, Murphy NE, Sivasithamparam K (1993a). Use of<br />

bacteriophage for the selective isolation of thermophilic actinomycetes<br />

from composted eucalyptus bark. Can. J. Microbiol. 39:46-51.<br />

Kurtböke DI, Wilson CR, Sivasithamparam K (1993b). Occurrence of<br />

Actinomadura phage in organic mulches used for avocado<br />

plantations in Western Australia. Can. J. Microbiol. 39:389-394.<br />

Krylov VN, Dela Cruz DM, Hertveldt K, Ackermann HW (2007). "φKZlike<br />

viruses" - a proposed new genus of myovirus bacteriophages.<br />

Arch. Virol. 152:1955-1959.<br />

Lavigne R, Seto D, Mahadevan P, Ackermann HW, Kropinski AM<br />

(2008). Unifying classical and molecular taxonomic classification.<br />

Analysis of the Podoviridae using BLASTP-based tools. Res.<br />

Microbiol. 159:406-414.<br />

Lavigne R, Darius P, Summer EJ, Seto D, Mahadevan P, Nilsson<br />

AS, Ackermann HW, Kropinski AM (2009). Classification of<br />

Myoviridae bacteriophages using protein sequence similarity. BCM<br />

Microbiol. 9:224.<br />

Liu B, Wu SJ, Song Q, Zhang XB, Xie LH (2006) Two novel<br />

bacteriophages of thermophilic bacteria isolated from deep-sea<br />

hydrothermal fields. Curr. Microbiol. 53:163-166.<br />

Meynell EW (1961). A phage, χ, which attacks motile bacteria. J. Gen.<br />

Microbiol. 25:253-290.<br />

Nacesco N, Constantinesco SP, Petrovici A (1969). Aspects électronooptiques<br />

du phage convertisseur Proteus vulgaris 121. Arch. Roum.<br />

Pathol. Exp. Microbiol. 28:838-848.<br />

Pasharawipas T, Thaikua S, Sriurairatana S, Ruangpan L,<br />

Direkbusarakum S, Manopvisetcharean J, Flegel TW (2005). Partial<br />

characterization of a novel bacteriophage of Vibrio harveyi isolated<br />

from shrimp culture ponds in Thailand. Virus Res. 114:63-69.<br />

Prestel E, Salamitou S, Dubow MS (2008). An examination of the bacteriophages<br />

and bacteria of the Namib desert. J. Microbiol. 46:364-372.<br />

Prigent M, Leroy M, Confalonieri F, Dutertre M, DuBow MS (2005). A<br />

diversity of bacteriophage forms and genomes can be isolated from<br />

the surface sands of the Sahara Desert. Extremophiles 9:289-296.<br />

Schaper M, Jofre J, Uys M, Grabow WOK (2002). Distribution of<br />

genotypes of F-specific RNA bacteriophages in human and nonhuman<br />

sources of faecal pollution in South Africa and Spain. J. Appl.<br />

Microbiol. 92:657-667.<br />

Stevenson RMW, Airdrie DW (1984) Isolation of Yersinia ruckeri<br />

bacteriophages. Appl. Environ. Microbiol. 47:1201-1205.<br />

Yu MX, Slater MR, Ackermann HW (2006). Isolation and characterization<br />

of Thermus bacteriophages. Arch. Virol. 151:663-679.


African Journal of Microbiology Research Vol. 5(16), pp. 2211-2216, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR10.767<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Evaluation of real-time PCR for Mycobacterium<br />

ulcerans in endemic region in Côte d’Ivoire<br />

Elise Solange Ngazoa-Kakou 1,3 *, Euloge Ekaza 1 , Nguetta Aka 2 , David Coulibaly-N’Golo 1 ,<br />

Bakary Coulibaly 2 and Mireille Dosso 1,2<br />

1 Plateforme de Biologie Moléculaire, Institut Pasteur Cote d’Ivoire, BP 490 Abidjan, Cote d’Ivoire.<br />

2 Unité des Mycobactéries Tuberculeuses et Atypiques, Institut Pasteur Cote d’Ivoire, BP 490 Abidjan, Cote d’Ivoire.<br />

3 Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval, Qc, G1K 7P4, Canada.<br />

Accepted 19 May, 2011<br />

Buruli ulcer (BU) is caused by a mycobacterium called Mycobacterium ulcerans. The events of BU are<br />

the skin lesions. The lack of early diagnosis and treatment cause severe disability. Today the<br />

emergence to BU in Africa and particularly in Côte d’Ivoire needs faster diagnosis to control and to<br />

prevent the infection by M. ulcerans. The surveillance of BU is difficult, because the transmission of M.<br />

ulcerans occurs in rural regions where the transport of fresh collected sample is long, and the detection<br />

with culture technique needs several months. This study has allowed the application of polymerase<br />

chain reaction (PCR) technique in real time with two targets for molecular diagnosis of BU in Côte<br />

d'Ivoire. 63 samples (clinical, environmental, local strains and reference strains) were analyzed in realtime<br />

PCR by comparing the target of the Insertion Sequence (IS) 2404 and the sequence Ketoreductase-<br />

B (KR-B), located respectively on the chromosome and on the virulence plasmid. 49 samples (76%)<br />

were positive in real-time for both targets. The sensitivity of the PCR shows a detection limit of 0.25<br />

genome copy for both targets. The capacity, speed and sensitivity of real-time PCR assays improve the<br />

diagnosis and contribute to strengthening the eradication of BU in Côte d’Ivoire.<br />

Key words: Buruli ulcer, Mycobacterium ulcerans, real-time, insertion sequence, ketoreductase, Côte d’Ivoire.<br />

INTRODUCTION<br />

Mycobacterium ulcerans is the third mycobacteria<br />

infection after tuberculosis and Leprosy and it causes<br />

Buruli ulcer (BU), a severe skin disease that has<br />

irreversible consequences in rural populations<br />

(MacCallum et al., 1993). The WHO was esteemed that<br />

33 countries was endemic or semi-endemic for the Buruli<br />

ulcer (Asiedu et al., 2000; Janssens et al., 2005). The<br />

eradication of Buruli ulcer is difficult because, the lack of<br />

early diagnostic in rural endemic regions, and the<br />

unknown of the disease in medical national structures<br />

system. In the rural westlands and marshes of West<br />

Africa, children are most affected (Asiedu et al., 2000;<br />

WHO, 2008). The polymerase chain reaction (PCR) was<br />

become the gold method to confirm clinical and<br />

environmental samples (Ross et al., 1997a, b; Portaels et<br />

*Corresponding author. E-mail: ngazoa_solange@yahoo.fr.<br />

al., 2001; Marsollier et al., 2004; Portaels et al., 2008).<br />

The most using targets were the insertion sequence<br />

IS2404 or IS26406 and the Ketoreductase B (KR-B),<br />

these are respectively located on the chromosome and<br />

on the virulence plasmid pMUM001 (van Werf et al.,<br />

2003). The cytotoxic mycolactone of M. ulcerans is produced<br />

from of three large multienzymes complexes called<br />

polypeptide synthases that are encoded by the genes<br />

mlsA1, mlsA2, and mlsB located on the plasmid (Stinear<br />

et al. 2004; 2005). The detection of M. ulcerans in clinical<br />

and in environmental samples was improved for the both<br />

most targets IS2404 and KR such aquatic insects, snails<br />

or in plants (Ross et al. 1994; Stinear et al. 2000;<br />

Eddyani et al. 2004; Philipps et al., 2005). In West Africa,<br />

the endemic region of BU is located in Côte d’Ivoire that<br />

has more 2000 confirmatory cases yearly (WHO, 2010).<br />

Molecular detection was the best method to confirm new<br />

human cases because of the low-growth of M. ulcerans in<br />

vitro (Portaels et al., 2001). Recently, de Vandelannoote


2212 Afr. J. Microbiol. Res.<br />

et al. (2010) have detected M. ulcerans using real-time<br />

PCR in small animals and in water sample in a BUendemic<br />

region. However, the sensitivity, the specificity<br />

and the capacity of real-time PCR are the major step<br />

forward to detect M. ulcerans. The application of real-time<br />

PCR in the Institute Pasteur contributes to increase the<br />

capacities of the national surveillance to eradicate the<br />

BU, by elucidating of the ecology and the transmission in<br />

the environment. The main aim of this study is to<br />

evaluate the performance of real time PCR as molecular<br />

diagnosis method, by using two targets IS2404 and KR-B<br />

to identify M. ulcerans in several types’ samples in Buruli<br />

ulcer-endemic region in Côte d’Ivoire.<br />

MATERIALS AND METHODS<br />

Mycobacterial strains<br />

Local strains were gathered from patients with Buruli ulcerative form<br />

in Cote d’Ivoire endemic regions. Those strains were isolated and<br />

maintained within Abidjan Pasteur Institute Mycobacterial<br />

Laboratory. Fresh swab clinical specimens were spilt for DNA<br />

extraction and culture technique. Environmental samples were<br />

collected in 2009 from different endemic regions of BU and were<br />

inclusive with strains isolated from water insects Reference strains<br />

were provided from the Institute of Tropical Medicine, Antwerp,<br />

Belgium (ITM) and from Centre Pasteur of Yaoundé, Cameroon<br />

(Table 1).<br />

DNA extraction<br />

Clinical specimen<br />

DNA from clinical samples was extracted using Phenol extraction<br />

method as previously described (Ekaza et al., 2004; Coulibaly et<br />

al., 2010).<br />

Bacterial strains<br />

DNA from bacterial strains was extracted by a phenol/chloroform<br />

method. Briefly five to ten colonies were harvested from<br />

Löwenstein-Jensen slants and suspended in 250 µl of lysis buffer.<br />

After overnight incubation at 37°C, phenol chloroform extraction<br />

was applied. The DNA was washed in ethanol and the pellet was<br />

dried and eluted in 50 µl of sterile water.<br />

Environmental samples<br />

Aquatic insects were placed in sterile box and add to liquid<br />

nitrogen, the matrix was crushed and 2 ml of 50 mM NaOH were<br />

added. 1 ml of sample was added to 150 µl of 0.1 M Tris and<br />

heated 20 min, 95°C. The extraction protocol of Mo Bio kit was<br />

applied following the instructions of the manufacturer. The pellet<br />

was eluted in sterile water and stored at -20°C.<br />

Real-time TaqMan PCR assays<br />

Real-time PCR was performed using PCR Reagents Kit (Promega,<br />

Madison, WI, USA) and the 7300 real-time PCR (Applied<br />

Biosystems, Forster City, USA). The reaction took place in a final<br />

volume of 25 µl and contained 5 µl of DNA, 0.3 µM of each primer,<br />

namely IS2404R and IS2404F (Rondoni et al., 2003) or KR-B-F and<br />

KR-B-F (Fyfe et al., 2007), and 0.25 µM of labelled IS2404 probe or<br />

KR-B probe, 20 µl PCR-Mix containing 25 mM MgCl2, Rox Dye<br />

Pure (Invitrogen, Cergy Pontoise, France), 10 mM dNTPs, 5X<br />

Buffer, 0.2 µl Go-Taq Flexi DNA Polymerase (Promega, Madison,<br />

WI, USA).<br />

PCR consisted of 35 cycles of melting at 95°C for 5 s, annealing<br />

and extension at 60°C for 1 min. A DNA segment of 58 and 65 bp<br />

for IS2404 and KR respectively in length was thus amplified.<br />

Fluorescence of FAM liberated from the probe by TaqMan was<br />

measured to determine the amplification threshold cycle (Ct), which<br />

was the first cycle at which fluorescent emission was 10-fold higher<br />

than the standard deviation of the mean baseline emission.<br />

Negative controls were performed with 5 µl of sterile water. A serial<br />

10-fold dilution of 2x10 7 CFU/µl extracted DNA was used to<br />

establish the standard curve. Each sample was duplicated and<br />

tested in real time assays.<br />

Sensitivity test of real-time PCR assays<br />

To determine the detection limit of both assays, serial dilutions of<br />

extracted DNA from 2.5 x10 4 CFU/µl from 2MU II strain<br />

(unpublished) and 5 µl were tested duplicate in real-time assays.<br />

RESULTS<br />

Sensitivity of the real-time PCR of M. ulcerans<br />

The serial dilution of DNA solution containing 2.5x10 4<br />

CFU/µl of M. ulcerans (2MUII) shows the detection by<br />

dilution (10 -6 ) with the target IS2404 by Ct 39. 27 and with<br />

the Ketoreductase-B by Ct 35.29 (Table 2) The detection<br />

limit of both assays was corresponding of 0.25 genome<br />

copies from M. ulcerans. The IS2404 is more sensitive<br />

with similar detection limit than the assay using<br />

Ketoreductase-B gen.<br />

Evaluation of real-time PCR for Insertion sequence<br />

IS2404 and Ketoreductase-B by different samples of<br />

M. ulcerans<br />

From 63 samples tested in real-time assays, 47 samples<br />

(74%) were positive by IS2404 and 40 samples (77%) by<br />

KR. Both assays show identical performance by the<br />

detection of local strains and clinical samples (Table 3).<br />

References strains show similar results by both real-time<br />

assays. No environmental sample was detected in both<br />

assays. 31 clinical samples (96%) were detected in both<br />

assays and confirm the sensitivity of real-time assays. 14<br />

samples (26%) were detected negative for both detection<br />

targets and 3 samples (5%) were negative by IS 2404<br />

real-time or by KR-B real-time assay.<br />

DISCUSSION<br />

Buruli ulcer was endemic in Côte d’Ivoire. The infection


Table 1. Types samples of M. ulcerans tested in this study.<br />

Samples Source or strain References<br />

ITM 9540 References strain Stieger et al., 2006<br />

ITM 980912 References strain Stieger et al., 2006<br />

ITM 842 References strain Stieger et al., 2006<br />

ITM 7922 References strain Stieger et al., 2006<br />

ITM 940662 References strain Stieger et al., 2006<br />

ITM 990006 References strain Stieger et al. unpublished<br />

ITM 94821 References strain Stieger et al. unpublished<br />

ITM 5150 References strain Stieger et al. 2006<br />

ITM 970483 References strain Stieger et al. 2006<br />

ITM 051459 References strain Stieger et al. 2006<br />

CIV-B043 Local laboratory strain This study<br />

CIV-B058 Local laboratory strain This study<br />

CIV-01-0104 Local laboratory strain This study<br />

CIV-01-098 Local laboratory strain This study<br />

CIV-01-0119 Local laboratory strain This study<br />

CIV-01-044 Local laboratory strain This study<br />

CIV-06-046 Local laboratory strain This study<br />

CIV-06-0120 Local laboratory strain This study<br />

CIV-01-054a Local laboratory strain This study<br />

CIV-009 Local laboratory strain This study<br />

CIV-090 Local laboratory strain This study<br />

CIV-091 Local laboratory strain This study<br />

CIV-028 Local laboratory strain This study<br />

CIV-077 Local laboratory strain This study<br />

CIV-01-093 Local laboratory strain This study<br />

CIV-01-043 Local laboratory strain This study<br />

01-021b Local laboratory strain This study<br />

U064 Local laboratory strain This study<br />

U044 Local laboratory strain This study<br />

02-43 Local laboratory strain This study<br />

B12 Local laboratory strain This study<br />

06-108 Local laboratory strain This study<br />

06-117 Local laboratory strain This study<br />

01-078 Local laboratory strain This study<br />

B043 Local laboratory strain This study<br />

02-003 Local laboratory strain This study<br />

06-120 Local laboratory strain This study<br />

01-104 Local laboratory strain This study<br />

01-109 Local laboratory strain This study<br />

08U-045 Local laboratory strain This study<br />

U059 Local laboratory strain This study<br />

01-098 Local laboratory strain This study<br />

UB09-960 Clinical sample This study<br />

UB09-973 Clinical sample This study<br />

UB09-988 Clinical sample This study<br />

UB09-989 Clinical sample This study<br />

UB09-995 Clinical sample This study<br />

UB09-997 Clinical sample This study<br />

UB09-1009 Clinical sample This study<br />

UB09-1025 Clinical sample This study<br />

UB09-1027 Clinical sample This study<br />

Ngazoa-Kakou et al. 2213


2214 Afr. J. Microbiol. Res.<br />

Table 1 Contd.<br />

UB09-1039 Clinical sample This study<br />

UB09-1042 Clinical sample This study<br />

UB09-1043 Clinical sample This study<br />

UBE1 Environmental sample This study<br />

UBE2 Environmental sample This study<br />

UBES1 Environmental sample This study<br />

UBES2 Environmental sample This study<br />

UBES3 Environmental sample This study<br />

UBSE4 Environmental sample This study<br />

UBSE5 Environmental sample This study<br />

UBSE6 Environmental sample This study<br />

UBSE7 Environmental sample This study<br />

Table 2. Sensitivity of the both real-time TaqMan assays.<br />

Dilution of DNA CFU/µl IS2404 Ct KR-B Ct<br />

2.5.10 4 20.88 ±1.8 19.6 ±1.2<br />

2.5.10 3 24.85 ±1.7 22.03 ±0.7<br />

2.5.10 2 28.35 ±1.4 25.3 ±1.1<br />

2.5.10 1 31.82 ±1.3 28.46 ±1.4<br />

2.5.10 0 35.45 ±1.7 30.18 ±1.3<br />

0.25 39.27 ±1.8 35.29±1.2<br />

Table 3. Detection of different samples for M. ulcerans in real-time assays.<br />

Sample type<br />

IS2404<br />

KR-B<br />

Positive Negative Positive Negative<br />

Clinical 8 4 8 4<br />

Environmental 0 9 0 9<br />

Local strains 30 2 31 1<br />

References strains 9 1 10 0<br />

Total 47 16 49 14<br />

% 74 26 77 23<br />

can be eradicated if the national surveillance becomes<br />

effective in all regions. Early diagnostic is the only step to<br />

prevent ulceration that has irreversible consequences for<br />

the patients. The number of cases of M. ulcerans in Côte<br />

d’Ivoire continues to rise, with more 2000 new human<br />

cases reported in 2009 compared with 1440 cases in<br />

2008 and 1 case in 1978 (WHO, 2010). With increasing<br />

numbers of clinical specimens for diagnosis as well as<br />

the need to determine the mode of transmission and the<br />

natural reservoir of the mycobacteria, rapid, sensitive,<br />

and specific molecular tests are needed. Fyfe et al.<br />

(2007) have developed two multiplex, real-time TaqMan<br />

PCR assays for the detection of M. ulcerans DNA in<br />

clinical and environmental samples. Recently, de<br />

Vandelannoote et al. (2010) have detected M. ulcerans in<br />

small animals in Ghana. To ameliorate the capacity of<br />

molecular diagnostic, we apply the real-time PCR with<br />

two targets in monoplex reaction to detect M. ulcerans in<br />

clinical and environmental samples. The IS2404 is the<br />

most target for M. ulcerans PCR used in several labours<br />

(Stinear et al., 1999). The Ketoreductase B domain is<br />

present in 15 copies within the mycolactone synthase<br />

gen and the real-time assay has shown gut sensitivity in<br />

previous assay (Rondoni et al., 2003). Real-time PCR is<br />

more 10-10.000 sensitive than classic PCR, however the<br />

PCR is recommended by WHO as the confirmatory test<br />

for clinical diagnosis of M. ulcerans infection.<br />

The application in this study of real-time assays to


detect clinical samples of Buruli ulcer was chosen in<br />

rapidity and the capacity for the most endemic area of M.<br />

ulcerans in West Africa. Our results confirm the sensitivity<br />

of both assays and the detection limit was 0. 25 genome<br />

copies. However, 96% of clinical samples have been<br />

confirmed the infection of M. ulcerans in this study. This<br />

detection limit was consistent of the results of Rondoni et<br />

al. (2003) with the detection of 0.2 genome copy in realtime<br />

of IS2404. Fyfe et al. (2007) have demonstrated that<br />

IS2404 or KR real-time PCR assays are able to detect all<br />

geographic strains of the world including African strains.<br />

The combination of both assays has facilitated to detect<br />

simultaneous mycolactone producing strains by the target<br />

KR-B and non-mycolactone producing strains by the<br />

target IS2404.<br />

The application of several types of samples confirms<br />

the specificity of both real-time assays. Because of the<br />

long storage of references ITM strains DNA, several<br />

strains missed by real-time assays. No environmental<br />

sample shows the presence of DNA of M. ulcerans in<br />

both assays. The presence of inhibitors in environmental<br />

samples was investigated because of the correlation of<br />

the high incidence of BU in Côte d’Ivoire. Our results<br />

show gut performance of real-time assay for both targets<br />

to detect mycolactone producing strain and non<br />

producing strains. We will investigate in future studies to<br />

detect M. ulcerans in environmental samples.<br />

In view of the high number of the samples received in<br />

the case of the confirmation of the cases of Buruli ulcer in<br />

Côte d’Ivoire and the search for the environmental<br />

reservoir of M. ulcerans. This technology is going to allow<br />

the avoidance of not only the long manipulations, but also<br />

the contaminations after PCR, and also to increase the<br />

sensibility and the specificity of the detection. However,<br />

the application of real-time PCR for M. ulcerans in<br />

Institute Pasteur will permit us to elucidate the ecology<br />

and the transmission of BU in Côte d’Ivoire.<br />

ACKNOWLEDGEMENTS<br />

The authors thank the Centre Pasteur of Yaoundé,<br />

Cameroon for the grateful gift of DNA of the strain MU2II.<br />

We thank the Institute for Tropical Medicine of Antwerp,<br />

Belgium for the references DNA using in this study. We<br />

thank the national surveillance program of Buruli ulcer<br />

and the World Health Organization. We thank Dr S.<br />

Bouzid for critical support of all parts in this work. This<br />

work was supported by a grant of Institut Pasteur de Côte<br />

d’Ivoire and Réseau International des Instituts Pasteur<br />

(RIIP).<br />

REFERENCES<br />

Asiedu K, Scherpbier R, Raviglione M (2000). Buruli Ulcer,<br />

Mycobacterium ulcerans infection. World Health Organization,<br />

Geneva, Switzerland, WHO.<br />

Coulibaly B, Coulibaly-N’Golo MDG, Ekaza E, Aka N, N’Guessan KR,<br />

Ngazoa-Kakou et al. 2215<br />

Baudryard A, Assandé JM, Trébissou N, Guédé-Guina F, Dosso M<br />

(2010). Implementation of in vitro culture of Mycobacterium ulcerans<br />

from clinical samples versus detection of acid fast bacilli and bacterial<br />

genome in Abidjan, Cote d’Ivoire. Bull. Soc. Path. Exo., 103(12): 2-7.<br />

PMID: 20084485.<br />

Eddyani M, Ofori-Adjei D, Teugels G, De Weirdt D, Boakye D, Meyers<br />

W, Portaels F (2004). Potential role for fish in transmission of<br />

Mycobacterium ulcerans disease (Buruli Ulcer): an environmental<br />

study. Appl. Environ. Microbiol., 70(9): 5679-5681. PMID: 15345458.<br />

Ekaza E, Kacou-N'douba A, Oniangue NC, Ehuie P, N'Guessan KR, Aka<br />

N, Bouzid SA, Faye-Kette H, Dosso M (2004). Contribution of genic<br />

amplification in the detection of M. ulcerans in exudates and in<br />

cutaneous biopsies in Cote d’Ivoire. Bull. Soc. Path. Exo., 97(2): 95-<br />

96.<br />

Fyfe JA, Lavender CJ, Johnson PD, Globan M, Sievers A, Azuolas J,<br />

Stinear TP (2007). Development and application of two multiplex realtime<br />

PCR assays for the detection of Mycobacterium ulcerans in<br />

clinical and environmental samples. Appl. Environ. Microbiol., 73(15):<br />

4733-4740. PMID: 17526786.<br />

Janssens PG, Pattyn SR, Meyers WM, Portaels F (2005). Buruli Ulcer:<br />

Anhistorical overview with updating to 2005. Bull. Seances Acad. R.<br />

Sci. Outre Mer., 51: 165–199.<br />

MacCallum P, Tolhurst J, Buckle G, Sissons H (1993). A new<br />

mycobacterial infection in man. J. Pathol. Bacteriol., 60: 93–122.<br />

PMID: 18935206.<br />

Marsollier L, Stinear T, Aubry J, Saint Andre´ JP, Robert R, Legras P,<br />

Manceau AL, Audrain C, Bourdon S, Kouakou H, Carbonnelle B<br />

(2004). Aquatic plants stimulate the growth of and biofilm formation<br />

by Mycobacterium ulcerans in axenic culture and harbor these<br />

bacteria in the environment. Appl. Environ. Microbiol., 70(2): 1097–<br />

1103. PMID: 14766593.<br />

Phillips R, Horsfield C, Kuijper S, Lartey A, Tetteh I, Etuaful S,<br />

Nyamekye B, Awuah P, Nyarko KM, Osei-Sarpong F, Lucas S, Kolk<br />

AH, Wansbrough-Jones M (2005). Sensitivity of PCR targeting the<br />

IS2404 insertion sequence of Mycobacterium ulcerans in an assay<br />

using punch biopsy specimens for diagnosis of Buruli Ulcer. J. Clin.<br />

Microbiol., 43(8): 3650–3656. PMID: 16081892.<br />

Portaels F, Johnson P, Meyers WM (ed.) (2001). Buruli Ulcer; diagnosis<br />

of Mycobacterium ulcerans disease; a manual for health care<br />

providers. World Health Organization, Geneva, Switzerland.<br />

Portaels F, Meyers WM, Ablordey A, Castro AG, Chemlal K, de Rijk P,<br />

Elsen P, Fissette K, Fraga AG, Lee R, Mahrous E, Small PL, Stragier<br />

P, Torrado E, Van Aerde A, Silva MT, Pedrosa J (2008). First<br />

cultivation and characterization of Mycobacterium ulcerans from the<br />

environment. PLoS. Negl. Trop. Dis., 2(3):e178.<br />

Rondini S, Mensah-Quainoo E, Troll H, Bodmer T, Pluschke G (2003).<br />

Development and application of real-time PCR assay for<br />

quantification of Mycobacterium ulcerans DNA. J. Clin. Microbiol.,<br />

41(9): 4231–4237. PMID: 12958250.<br />

Ross B, Johnson P, Oppedisano F, Marino L, Sievers A, Stinear T,<br />

Hayman J, Veitch M, Robins-Browne R (1997). Detection of<br />

Mycobacterium ulcerans in environmental samples during an<br />

outbreak of ulcerative disease. Appl. Environ. Microbiol., 63(10):<br />

4135–4138. PMID: 9327583.<br />

Ross B, Marino L, Oppedisano F, Edwards R, Robins-Browne R,<br />

Johnson P (1997). Development of a PCR assay for rapid diagnosis<br />

of Mycobacterium ulcerans infection. J. Clin. Microbiol., 35(7): 1696–<br />

1700. PMID: 9196176.<br />

Stinear TP, Davis JK, Marino L, Robin-Browne RM, Oppedisano F,<br />

Sievers A, Johnson PDR (1999). Identification and characterization of<br />

IS2404 and IS2606: two distinct repeated sequences for detection of<br />

Mycobacterium ulcerans by PCR. J. Clin. Microbiol., 37(4): 1018–<br />

1023. PMID: 10074520.<br />

Stinear T, Davies JK, Jenkin GA, Portaels F, Ross BC, Oppedisano F,<br />

Purcell M, Hayman JA, Johnson PD (2000). A simple PCR method<br />

for rapid genotype analysis of Mycobacterium ulcerans. J. Clin.<br />

Microbiol., 38(4): 1482–1487. PMID: 10747130.<br />

Stinear TP, Mve-Obiang A, Small PL, Frigui W, Pryor MJ, Brosch R,<br />

Jenkin GA, Johnson PD, Davies JK, Lee RE, Adusumilli S, Garnier T,<br />

Haydock SF, Leadlay PF, Cole ST (2004). Giant plasmid-encoded<br />

polyketide synthases produce the macrolide toxin of Mycobacterium<br />

ulcerans. Proc. Natl. Acad. Sci., 101(5): 1345-1349.PMID: 14736915.


2216 Afr. J. Microbiol. Res.<br />

Stragier P, Ablordey A, Bayonne LM, Lugor YL, Sindani IS, Suykerbuyk<br />

P, Wabinga H, Meyers WM, Portaels F (2006). Heterogeneity among<br />

Mycobacterium ulcerans Isolates from Africa. Emerging Infectious<br />

Diseases, 12(5): 844-847. PMID: 16704851.<br />

van der Werf T, Stinear T, Stienstra Y, van der Graaf W, Small P<br />

(2003). Mycolactones and Mycobacterium ulcerans disease. Lancet,<br />

362(9389): 1062–1064. PMID: 14522538.<br />

de Vandelannoote K, Durnez L, Amissah D, Gryseels S, Dodoo A,<br />

Yeboah S, Addo P, Eddyani M, Leirs H, Ablordey A, Portaels F<br />

(2010). Application of real-time PCR in Ghana, a Buruli Ulcerendemic<br />

country, confirms the presence of Mycobacterium ulcerans<br />

in the environment. FEMS. Microbiol. Lett., 304(2): 191-194. PMID:<br />

20146745.<br />

World Health Organization (2008). Buruli Ulcer: progress report, 2004–<br />

2008. Wkly. Epidemiol. Rec., 83(17): 145–156. PMID: 18437758.<br />

World Health Organization (2010). Annual Meeting of Buruli Ulcer, 22-<br />

24 march Geneva, Switzerland


African Journal of Microbiology Research Vol. 5(16), pp. 2217-2221, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR10.782<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Contamination of cattle carcasses by Escherichia coli<br />

shiga like toxin with high antimicrobials resistence<br />

Everlon Cid Rigobelo 1 *, Renato Pariz Maluta 2 , Clarissa Araújo Borges 2 ,<br />

Lívia Gerbasi Beraldo 2 , Manoel Victor Franco 3 , Lemos Sirlei Aparecida Maestá 1 and<br />

Fernando Antonio de Ávila 4<br />

1 Campus Experimental de Dracena- Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Brazil.<br />

2 Programa de Pós-Graduação em Microbiologia Agropecuária Universidade Estadual Paulista Júlio de Mesquita Filho<br />

(UNESP) Jaboticabal Brazil.<br />

3 Faculdade de Ciências Agrárias e Veterinárias de Jaboticabal. Departamento de Biologia Aplicada a Agropecuária,<br />

Brazil.<br />

4 Faculdade de Ciências Agrárias e Veterinárias de Jaboticabal, Departamento de Patologia Veterinária, Brazil.<br />

Accepted 15 March, 2011<br />

During processing of cattle carcasses, contamination may occurs with the transfer of microbiota of<br />

animals feaces to carcasses. This contamination many times may be by Escherichia coli carriers of<br />

virulence factor as stx and eae genes being classified as Shiga like toxin. Shiga toxin-producing<br />

Escherichia coli (STEC) is recognized wordwide as human pathogen. A survey was performed to<br />

determine the sensibility profile to several antimicrobial drugs of STEC in carcasses obtained from an<br />

abattoir in Brazil between March 2008 and August at 2009. A total of 120 STEC were isolated. All<br />

isolates were confirmed as being E. coli by their biochemical analysis and submitted to polymerase<br />

chain reaction (PCR) for detection of stx, eae and ehly genes. No strains was isolated being carriers of<br />

ehly gene. The number of isolates carriers of eae gene were 48/120. The most frequent resistance was<br />

seen against cephalothin (84.0%), streptomycin (45.0%), nalidixic acid (42.0%) and tetracycline (20.0%).<br />

Multidrug resistance (MDR) to three or more antimicrobial agents was observed in 46 (38.3%) E. coli<br />

isolates. The findings of STEC and MRD show that cattle carcasses may be a reservoir of pathogenic<br />

bacterial for the consumer public.<br />

Key words: Multi-drug resistance, Escherichia coli, shiga toxin-producing Escherichia coli (STEC).<br />

INTRODUCTION<br />

Pathogenic Escherichia coli are classified at different<br />

groups of strains that cause a common disease using<br />

common and remarkable assortments of virulence factors<br />

(Kaper et al., 2004). One such pathotype, the STEC is<br />

the causative agent of severe clinical syndromes in<br />

humans such as haemolytic uremic syndrome (HUS) and<br />

haemorrhagic colitis. However the transmission of STEC<br />

*Corresponding author. E-mail: everlon@dracena.unesp.br. Tel:<br />

+ 55 (18) 3821- 8200.<br />

Abbreviations: STEC, Shiga toxin-producing Escherichia coli;<br />

PCR, polymerase chain reaction; MDR, multidrug resistance;<br />

HUS, haemolytic uremic syndrome; PR, Paraná.<br />

occurs by waterborne, from person to person and also<br />

may be transmitted by food borne (Nataro and Kaper,<br />

1998). E. coli is regarded as an indicator of fecal<br />

contamination when isolated from carcass processing.<br />

Levels of E. coli associated with cattle carcasses can<br />

increase or decrease during processing according to<br />

factors such as the levels of fecal contamination of live<br />

cattle, efficiency of evisceration and hygienic practices in<br />

the abattoir (Bell, 1997).<br />

Cattle, considered primary reservoirs of both O157 and<br />

non-O157 STEC bacteria (Bettelheim, 2000), frequently<br />

carry STEC without showing any pathological symptoms<br />

(Blanco et al., 1997). The full list of bacterial virulence<br />

determinants necessary for STEC's pathological effects is<br />

not known. Two types of Shiga like toxin, stx1 and stx2<br />

(encoded by stx1 and stx2 genes), are associated with


2218 Afr. J. Microbiol. Res.<br />

human disease. These toxins vary in their amino-acid<br />

sequence (Kaper et al., 1998) antigenicity, and in their<br />

activation and receptor specificity (Schmitt et al., 1999).<br />

E. coli acquire stx genes, and the subsequent ability to<br />

produce toxins, following infection with temperate<br />

bacteriophages (James et al., 2001). The ability of E. coli<br />

to adhere to intestinal epithe-lium is crucial in the<br />

colonization of the intestine, and therefore the<br />

progression of disease in humans.<br />

The protein intimin, encoded by the eae gene, enables<br />

intimate attachment of E. coli to intestinal cells<br />

(Donnenberg et al., 1992), causing characteristic<br />

attaching/effacing lesions (Paton et al., 1998). This<br />

attachment also enables Shiga toxins to be injected into<br />

the epithelial cytoplasm through a type III secretion<br />

system (Kaper, 2004). Other virulence factors such as<br />

intimin (eae) and hemolysin (hly A) are thought to<br />

enhance pathogenicity, but are not required for strains to<br />

produce severe disease, including HUS (Bonnet et al.,<br />

1998; Acheson, 2000). Antimicrobial therapy is an<br />

important tool for infection treatment, resistance to<br />

antimicrobials is a cause of great concern in veterinary<br />

medicine (Monro and Polk, 2000). Indeed, a close<br />

association between the use of antimicrobial agents for<br />

the treatment of infections in animals and the observed<br />

levels of resistance exists (Chaslus-Dancia, 2001). The<br />

use of antibiotics in animal agriculture has been a<br />

controversial issue due to the potential transfer of<br />

antibiotic resistance from animals to humans. This could<br />

have several public health implications that may cause<br />

treatment failure, including death and illness prolongation,<br />

as well as increase in the associated costs (Kelly et<br />

al., 2004).<br />

The direct impact of resistance evolved from the use of<br />

antimicrobials in treatment of animal infection, is not<br />

clear. Since the antimicrobials routinely used for the treatment<br />

of infections in humans are also used in animals for<br />

both therapy and prevention or as growth promotion<br />

factors, it is not easy to describe the relative contributions<br />

of animal derived resistant strains to human E. coli<br />

disease (Maynard et al., 2004). Outbreaks have been<br />

associated with consumption of STEC contaminated and<br />

undercooked hamburgers, subsequent to both animal<br />

and foods (Erickson and Doyle, 2007). This probably<br />

occurs because during the processing of the carcass,<br />

fecal contamination or transfer of bacteria from the<br />

animal's hide to the carcass can facilitate transmission of<br />

pathogenic E. coli to food supplies (Bell, 1997; Barkocy-<br />

Gallagher et al., 2001). Some studies found a high<br />

prevalence of STEC in feces of healthy cattle, in Brazil,<br />

(Irino et al., 2005), Rio de Janeiro (Cerqueira et al.,<br />

1999), Rio Grande do Sul (Moreira et al., 2003; Timm et<br />

al., 2007) and also in Paraná (PR), (Farah et al., 2007;<br />

Pigatto et al., 2008) and a prevalence of 1 to 2% of STEC<br />

in cases of diarrhea in humans was reported by Vaz et<br />

al. (2004), De Toni et al. (2009). The objective of this<br />

study was to determine the virulence profiles and the<br />

antimicrobial drug resistance of E. coli isolates from beef<br />

carcasses at an abattoir in Brazil.<br />

MATERIALS AND METHODS<br />

Carcass samples<br />

Six hundred carcass samples were collected an abattoir in São<br />

Paulo State, in southwestern Brazil, between March 2008 and<br />

August 2009. Samples studied were from carcasses cattle raised at<br />

pastures. Sampling of 150 feedlot cattle was done on four different<br />

occasions, two in the rain season and two in the dry season. Each<br />

sample was obtained using a Specie- Sponge (3M- Brazil)<br />

moistened with 25 ml of Brilliant Green (BBL/Becton Dickins on) in<br />

a stomacher bag. Sponges were wrung out as much as possible<br />

within the bag and used to swab each area. Each carcass was<br />

followed along the processing and sampled at three different stages<br />

always at the same site of the rump, near the anus over an area of<br />

10 × 30 cm, delineated by a sterile metal template, from the same<br />

half of each carcass. All samples were taken to the laboratory in an<br />

ice-cooled bag and kept for 12 h at room temperature.<br />

Bacterial isolates<br />

One hundred microliters of each sample was streaked on<br />

MacConkey agar plates (Oxoid Limited) and incubated at 37°C for<br />

24 h. Colonies showing E. coli characteristics were submitted to<br />

Gram staining and identified by standard biochemical tests; oxidase<br />

negative, indole positive, Simon's citrate negative, urease negative<br />

and hydrogen sulfide negative (Koneman et al., 1997). The isolates<br />

were serotyped for O157 using Latex Agglutination test kit (Oxoid,<br />

Basingstoke, UK). Negative strains were considered non-O157<br />

strains.<br />

PCR screening of samples<br />

Bacterial strains, grown overnight in nutrient broth (Sigma Chemical<br />

Company) at 37°C, were pelleted by centrifugation at 12, 000 g for<br />

1 min , resuspended in 200 µl of sterile distilled water and lyzed by<br />

boiling for 10 min. Lysate was centrifuged as described above and<br />

150 µl of the supernatants were used as DNA for the PCR (Wani et<br />

al., 2003). A total of 120 E. coli isolates were subjected to PCR. stx<br />

1, stx 2 and eae genes were detected using the primers and PCR<br />

conditions described by China et al. (1998) .<br />

Expression of E-Hly<br />

Expression of enterohemolysin was determined based on the<br />

method described by Beutin et al. (1989). Plates were incubated at<br />

37°C for 24 h and observed for hemolysis after 3 h (for expression<br />

of a -hemolysin) and 24 h (for E- Hly), respectively. The reference<br />

strains used in this assay were E. coli U4- 41 (positive control for a -<br />

hemolysin), E. coli 32511 (STEC O157: H7) (positive control for E-<br />

Hly), and E. coli K12 (negative control).<br />

Susceptibility testing<br />

In vitro susceptibility testing was performed by a standardized disk<br />

diffusion method (CLSI 2008). Staphylococcus aureus ATCC 29213<br />

and E. coli ATCC 25922 served as quality control strains. Four<br />

antimicrobial agents were selected for the tests: cephalothin,<br />

streptomycin, nalidixic acid and tetracycline. The antimicrobials<br />

used in this study were the same used by farmers in animal<br />

produce.


Table 1. Distribution of the Escherichia coli isolates at two different<br />

seasons collected between March 2008 and August 2009.<br />

Carcass<br />

Collection Season Stx genes eae<br />

1º Rainy 35/150 23/150<br />

2º Rainy 47/150 12/150<br />

3º Dry 17/150 5/150<br />

4º<br />

Total<br />

Dry 21/150<br />

120 Stx+<br />

8/150<br />

48 eae+<br />

Figure 1. Photograph of a 1.5% agarose gel stained with ethidium<br />

bromide. Columns: 1 = 100 bp DNA ladder; 2 = positive control; 3 =<br />

negative control; 4 = strain positive for eae; 4, 6 and 8 = strain<br />

negative for all studied genes; 7 = strain positive for stx1 and stx2.<br />

RESULTS<br />

All isolates, confirmed as being E. coli by their<br />

biochemical analysis, were submitted to PCR for the<br />

detection of sequences of virulence genes. From each<br />

MacConkey agar plate a loopful from a confluent<br />

bacterial growth was collected and analyzed. A total six<br />

hundred E. coli strains isolates the cattle carcasses were<br />

separate 120 isolates that carrying stx1, stx2 and eae<br />

genes. These isolates just 45 were carriers of eae gene<br />

(Table1 and Figure 1). All isolates were collected of<br />

preevisceration stage. There were not isolating of strains<br />

of pre-evisceration stages and neither of post-processing<br />

stage (data not show). The isolates number containing<br />

both stx and eae gene during rainy season were high<br />

than dry season (Table1) and also the isolates number<br />

that carried the eae genes were high than the number of<br />

isolates that carried eae genes. In no isolates was<br />

verified the expression of enterohemolisyn. All isolates<br />

Rigobelo et al. 2219<br />

were tested for this hemolytic toxin and also no isolated<br />

were isolates was serotyped as O157. E. coli strains<br />

were tested against ten antimicrobial agents. The<br />

resistance pattern observed was: cephalothin (84.0%),<br />

streptomycin (45.0%) and nalidixic acid (42.0%) and<br />

tetracycline (20.0%) (Figure 1), 24% of the isolates were<br />

resistant to all the antibiotics tested. Multidrug resistance<br />

was seen in 38.4% of the isolates and resistance to 2 or<br />

3 antibiotics was common (Figure 2 and 3).<br />

DISCUSSION<br />

Among 600 strains analyzed only three were<br />

enterohemolysin positive. These results were similar to<br />

Rigobelo et al. (2008) that analyzed 216 samples from<br />

bovine carcasses and all of the isolates were negative for<br />

ehly gene. During raining season were found a high<br />

prevalence of STEC than dry season; probably the<br />

presence of water increased the spread of bacteria<br />

STEC. Some authors as Rogerie et al. (2001) reported<br />

lower post processing of nonO157 STEC prevalence<br />

(1.9%) on carcasses sampled during the summer in<br />

plants in France. Similarly, the non-O157 STEC<br />

prevalence on carcasses processed in Hong Kong was<br />

reported to be 1.7% (Leung et al., 2001), however, Arthur<br />

et al. (2002) reported higher level (54.0%) of contamination<br />

with nonO157 STEC in carcasses processed in<br />

the United States. Major sources of pathogens in<br />

processing of carcasses have been the hide and hair<br />

(Barkocy-Gallagher et al., 2001). It is not clear what<br />

proportion of non-O157 STEC bacteria detected in cattle<br />

feces or on beef carcasses is able to cause disease in<br />

humans. Gyles et al. (1998) defend the idea that all<br />

STEC bacteria could be pathogenic under adequate<br />

circumstances. In the present study, the detected level of<br />

STEC strains (20%) did not match those reported by<br />

others (Rogerie et al., 2001; Leung et al., 2001; Mora et<br />

al., 2005). To the best of our knowledge, we could not<br />

find data from Brazil for comparison. Only Rigobelo et al.<br />

(2006) report of STEC (1.25%) and Rigobelo et al. (2008)<br />

report (1, 4%) of STEC. These differences were probably<br />

because of low hygienic conditions of abattoir where we<br />

collected the samples.<br />

Some authors have reported the detection of STEC<br />

strains in fecal samples of dairy cattle (Irino et al., 2005),<br />

from diarrheic (Leomil et al., 2003) and from mastitic<br />

cattle (Lira et al., 2004) but none from abattoir samples.<br />

In all of them, the stx 2 gene has been predominantly<br />

found, and the non-O 157 STEC strains detected. Only a<br />

small number of O157 strains have been detected among<br />

bovine fecal samples 0.6% as reported by Irino et al.<br />

(2005), they did not express the stx gene. Interestingly,<br />

the O157: H 7 strains isolated in São Paulo State from<br />

human infections, were all stx -producers (Vaz et al.,<br />

2004 ), predominantly presenting the stx 1 gene. For<br />

more than four decades it has been a common practice<br />

on farms to use antimicrobial agents for disease


2220 Afr. J. Microbiol. Res.<br />

Figure 2. Antimicrobial resistance pattern of Escherichia coli isolate. CFL-cephalothin;<br />

STR - streptomycin – NAL-nalidixic acid; TET-tetracycline.<br />

Figure 3. Distribution of multidrug resistance to four antimicrobial drugs among<br />

Escherichia coli strains (n=120).<br />

prevention and growth promotion of animals. Widespread<br />

use of antimicrobial agents, select for resistance<br />

enhancement and may have promoted the increasing<br />

frequency of STEC strain's multidrug resistance in<br />

bovines. This could result in STEC population increases<br />

and perhaps greater shedding which could lead to higher<br />

contamination of animal food products with STEC (Zhao<br />

et al., 2001).<br />

Khan et al. (2002) reported resistance to one or more<br />

antibiotics in 49.2% of STEC strains in India, with some<br />

strains exhibiting multidrug resistance. Antimicrobial<br />

resistant bacteria from animals may colonize human<br />

population via the food chain; it is possible that resistant<br />

bacteria may be readily transferred to humans from<br />

animals used as food sources (Van den Bogaard and<br />

Stobberingh, 2000). During processing at an abattoir in<br />

Brazil we report a high level (20%) of occurrence of<br />

STEC strains on beef carcasses and also high


antimicrobial resistance suggesting poor hygienic<br />

conditions of slaughter of animals.<br />

ACKNOWLEDGEMENT<br />

FAPESP Fundação de Amparo Pesquisa do Estado de<br />

São Paulo financial support.<br />

REFERENCES<br />

Acheson DW (2000). How does Escherichia coli O157:H7 testing in<br />

meat compare with what we are seeing clinically? J. Food Protect.,<br />

63: 819–821.<br />

Arthur TM, Barkocy-Gallagher GA, Rivera-Betancourt M, Koohmaraie M<br />

(2002). Prevalence and characterization of non O157 Shiga toxin<br />

producing Escherichia coli on carcasses in commercial beef cattle<br />

processing plants. Appl. Environ. Microbiol., 68: 4847–4852.<br />

Barkocy-Gallagher GA, Arthur GA, Siragusa GR, Keen JE, Elder RO,<br />

Laegreid WW, Koohmaraie M (2001). Genotype analyses of<br />

Escherichia coli O157: H7 and O157 nonmotile isolates recovered<br />

from beef cattle and carcasses at processing plants in the<br />

Midwestern states of the United States. Appl. Environ. Microbiol., 67:<br />

3810– 3818.<br />

Bell RG (1997). Distribution and sources of microbial contamination of<br />

beef carcasses. J. Appl. Microbiol., 82: 292–300.<br />

Bettelheim KA (2000). Role of non - O15 7 VTEC. J. Appl. Microbiol.,<br />

88: 385–505.<br />

Beutin L, Geier D, Zimmermann S, Aleksic S, Gillespie HA, Whittam TS<br />

(1989). Epidemiological relatedness and clonal types of natural<br />

populations of Escherichia coli strains producing Shiga toxins in<br />

separate populations of cattle and sheep. Appl. Environ. Microbiol.,<br />

63: 2175–2180.<br />

Blanco J, Blanco M, Blanco JE (1993). Enterotoxigenic, verotoxigenic<br />

and necrotoxigenic Escherichia coli isolated from cattle in Spain. Am.<br />

J. Vet. Res., 54: 1446– 1451.<br />

Blanco M, Blanco JE, Blanco J, Mora A, Prado C, Alonso MP, Mourino<br />

M, Madrid C, Balsalobre C, Juarez A (1997). Distribuition and<br />

characterization of faecal verotoxin producing Escherichia coli<br />

(VTEC) isolated from healthy cattle. Vet. Microbiol., 54: 309–319.<br />

Bonnet R, Souweine B, Gauthier G, Rich C, Livrelli V, Sirot J, Joly B,<br />

Forestier C (1998). Non-O157:H7 Stx2 producing Escherichia coli<br />

strains associated with sporadic cases of hemolytic uremic syndrome<br />

in adults. J. Clin. Microbiol., 36: 1777– 1780.<br />

Cerqueira AMF, Guth BEC, Joaquim RM, Andrade JRC (1999). High<br />

occurrence of Shiga toxin-producing Escherichia coli (STEC) in<br />

healthy cattle in Rio de Janeiro state, Brazil. Vet. Microbiol., 70: 111-<br />

121.<br />

China B, Pirson V, Mainil J (1998). Prevalence and molecular typing of<br />

attaching and effacing Escherichia coli among calf population in<br />

Belgium. Vet. Microbiol., 63: 249 –259.<br />

De Toni F, Souza EM, Pedrosa FO, Klassen K, Irino K, Rigo LU,<br />

Steffens MBR, Fialho OB, Farah SMSS, Fadel-Picheth CMT (2009).<br />

A prospective study on Shiga toxin-producing Escherichia coli in<br />

children with diarrhoea in Paraná state, Brazil. Lett. Appl. Microbiol.,<br />

48: 645-647.<br />

Donnenberg MS, Kaper JB (1992). Enteropathogenic Escherichia coli.<br />

Infect. Immun., 60: 3953– 3961.<br />

Erickson MC, Doyle MP (2007). Food as a vehicle for transmission of<br />

Shiga toxin-producing Escherichia coli. J. Food Prot., 70: 2426-2449.<br />

Farah SMSS, Souza EM, Pedrosa FO, Irino K, Silva LR, Rigo LU,<br />

Steffens MBR, Pigatto CP, Fadel-Picheth, CMT (2007). Phenotypic<br />

and genotypic traits of Shiga toxin-producing Escherichia coli strains<br />

isolated from beef cattle from Paraná state, Southern Brazil. Lett.<br />

Appl. Microbiol., 44: 607-612.<br />

Gyles C, Johnson R, Gao, A, Ziebell K, Pierard D, Aleksic S, Boerlis, P<br />

(1998). Association of enterohemorrhagic Escherichia coli hemolysin<br />

with serotypes of Shiga toxin producing E. coli of humans and bovine<br />

origins. Appl. Environm. Microbiol., 64: 4134 –4141.<br />

Irino K, Kato MAMF, Vaz TMI, Ramos, II, Souza MAC, Cruz AS, Gomes<br />

Rigobelo et al. 2221<br />

TAT, Vieira, MAM, Guth BEC (2005). Serotypes and virulence markers<br />

of Shiga toxin-producing Escherichia coli (STEC) isolated from dairy<br />

cattle in São Paulo State, Brazil. Vet. Microbiol., 105: 29 –36.<br />

James CE (2001). Lytic and lysogenic infection of diverse Escherichia<br />

coli and Shigella strains with a verocytotoxigenic bacteriophage.<br />

Appl. Environ. Microbiol., 67: 4335–4337.<br />

Kaper JB, Nataro JP (2004). Mobley LT. Pathogenic Escherichia coli.<br />

Nat. Rev. Microbiol., 2: 123– 140.<br />

Kaper JB, O’brian AD (1998). Escherichia coli O157:H7 and other Shiga<br />

Toxin-producing E. coli Strains. Washington DC: ASM Press.<br />

Khan A, Das SC, Ramamurthy T, Sikdar A, Khanam J, Yamasaki S,<br />

Takeda Y, Nair, GB (2002). Antibiotic resistance, virulence gene, and<br />

molecular profiles of Shiga toxin producing Escherichia coli isolates<br />

from diverse source in Calcutta India. J. Clin, Microbiol., 40: 2009–<br />

2015.<br />

Koneman EW, Allen SD, Schrekenberger PC, Janda WM, Winn WC<br />

(1997). Color Atlas and Textbook Microbiology, 5 ed. Lippincott<br />

Company, Philadelphia.<br />

Leomil L, Aidar-Ugrinovich L, Guth BEC, Irino K, Vettorato MP, Onuma<br />

DL, De Castro AFP (2003). Frequency of Shiga toxin-producing<br />

Escherichia coli (STEC) isolates among diarrheic and non-diarrheic<br />

calves in Brazil. Vet. Microbiol., 97: 103– 109.<br />

Leung PHM, Yam WC, Ng WW, Peiris JS (2001). The prevalence and<br />

characterization of verotoxin-producing Escherichia coli isolated from<br />

cattle and pigs in an abattoir in Hong Kong. Epidemiol. Infect., 126:<br />

173–179.<br />

Lira WM, Macedo C, Marin JM (2004). The incidence of Shiga<br />

toxinproducing Escherichia coli in cattle with mastitis in Brazil. J.<br />

Appl. Microbiol., 97: 861–866.<br />

Maynard C, Bekal S, Sanschagrin F, Levesque RC, Brousseau R,<br />

Masson L, Lariviere S, Harel J (2004). Heterogeneity among<br />

virulence and antimicrobial resistance gene profiles of extraintestinal<br />

Escherichia coli isolates of animal and human origin. J. Clin.<br />

Microbiol., 42: 5444–5452.<br />

Mora A, Blanco JE, Blanco M, Alonso MP, Dhabi G, Echeita A,<br />

Gonzalez EA, Bernardez MI, Blanco J (2005). Antimicrobial<br />

resistance of Shiga toxin (verotoxin)-producing Escherichia coli<br />

O157:H7 and non-O157 strains isolated from humans, cattle, sheep<br />

and food in Spain. Res. Microbiol., 156: 793-806.<br />

Moreira CN, Pereira MA, Brod CS, Rodrigues DP, Carvalhal JB, Aleixo<br />

JAG (2003). Shiga toxin-producing Escherichia coli (STEC) isolated<br />

from healthy dairy cattle in Southern Brazil. Vet. Microbiol., 93: 179-<br />

183.<br />

Nataro JP, Kaper JB (1998). Diarrheagenic Escherichia coli. Clin.<br />

Microbiol. Rev., 11: 142–201.<br />

Paton JC, Paton AW (1998). Pathogenesis and diagnosis of Shiga-<br />

toxin producing Escherichia coli infections. Clin. Microbiol. Rev., 11:<br />

450–479.<br />

Pigatto CP, Schocken-Iturrino RP, Souza EM, Pedrosa FO, Comarella<br />

L, Irino K, Kato MAMF, Farah SMSS, Warth JF, Fadel-Picheth CMT<br />

(2008). Virulence properties and antimicrobial susceptibility of Shiga<br />

toxin-producing Escherichia coli strains isolated from healthy cattle<br />

from Paraná state, Brazil. Can. J. Microbiol., 54: 588-593.<br />

Rigobelo EC, Santo E, Marin JM (2008). Beef carcass contamination by<br />

Shiga toxin-producing Escherichia coli strains in an abattoir in Brazil:<br />

Characterization and Resistance to antimicrobial drugs. Foodborne<br />

and diseases., 5: 6.<br />

Rigobelo EC, Stella AE, Ávila FA, Macedo C, Marin JM (2006).<br />

Characterization of Escherichia coli isolated from carcasses of beef<br />

cattle during their processing at an abattoir in Brazil. Inter. J. Food<br />

Microbiol., 110: 194-198.<br />

Schmitt CK, Meysick KC, O’Brien AD (1999). Bacterial toxins: friends or<br />

foes? Emerging Infect. Dis., 5: 224–234.<br />

Timm CD, Irino K, Gomes TAT, Vieira MM, Guth BEC, Vaz TMI, Moreira<br />

CN, Aleixo JAG (2007). Virulence markers and serotypes of Shiga<br />

toxin-producing Escherichia coli, isolated from cattle in Rio Grande<br />

do Sul, Brazil. Lett. Appl. Microbiol., 44: 419-425.<br />

Vaz TMI, Irino K, Kato MAMF, Dias AMG, Gomes TAT, Medeiros MIC,<br />

Rocha MMM, Guth BEC (2004). Virulence properties and<br />

characteristics of Shiga toxin-producing Escherichia coli in São<br />

Paulo, Brazil, from 1976 through 1999. J. Clin. Microbiol., 42: 903-<br />

905.


African Journal of Microbiology Research Vol. 5(16), pp. 2222-2228, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.069<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Determination of insecticidal toxicity of three species of<br />

entomopathogenic spore-forming bacterial isolates<br />

against Tenebrio molitor L. (Coleoptera: Tenebrionidae)<br />

N. Du Rand and M. D. Laing<br />

Department of Plant Pathology, University of KwaZulu-Natal, Private Bag X01, Scottsville 3209, Pietermaritzburg,<br />

Republic of South Africa.<br />

Accepted 30 July, 2011<br />

Bioassays were conducted using larvae of mealworms, Tenebrio molitor, to determine lethal<br />

concentration for five entomopathogenic strains of spore-forming bacteria. Lethal concentration was<br />

determined by feeding T. molitor larvae cabbage discs dipped in whole cell cultures of these five strains<br />

of bacteria. The strains of bacteria were isolates of Bacillus cereus, Bacillus thuringiensis and<br />

Brevibacillus laterosporus. An isolate of B. cereus required the highest concentration of viable spores<br />

(8.531 x 10 7 spores ml -1 ) to achieve its LC50, whereas an isolate of B. laterosporus required the lowest<br />

concentration of viable spores (3.388 x 10 6 spores ml -1 ) to achieve LC50.<br />

Key words: Mealworms, spore-forming bacteria, bioassays.<br />

INTRODUCTION<br />

The first isolates of B. thuringiensis Berliner subspecies<br />

effective against coleopterans were isolated from a<br />

mealworm larva, Tenebrio molitor L. (Coleoptera:<br />

Tenebrionidae). This subspecies was subsequently<br />

named B. thuringiensis subsp. tenebrionis (Kriege et al.,<br />

1983). Another coleopteran specific strain was later<br />

isolated, B. thuringiensis subsp. san diego (Herrnstadt et<br />

al., 1986). These subsp. of B. thuringiensis are effective<br />

against one of the USA’s most important potato pests,<br />

the Colorado potato beetle, Leptinotarsa decemlineata<br />

Say (Coleoptera: Chrysomelidae) (Nault and Kennedy,<br />

1999). Colorado potato beetle has since been reported to<br />

have developed resistance to the Cry3A toxin (Whaldon<br />

et al., 1993). The development of resistance to pesticides<br />

is not uncommon in insects repeatedly subjected to a<br />

single pesticide, especially when only one toxin is<br />

involved (Ferré and Van Rie, 1992; Tabashnik, 1994).<br />

Various other coleopteran specific B. thuringiensis<br />

strains have been isolated and are effective against<br />

*Corresponding author. E-mail: 962082021@ukzn.ac.za.<br />

scarabaeid beetles such as Anomala cuprea Hope<br />

(Coleoptera: Dynastidae), A. rufocuprea Motschulsky<br />

(Coleoptera: Dynastidae) and Popillia japonica Newman<br />

(Coleoptera: Scarabaeidae) (Ohba et al., 1992).<br />

Bioassays on coleopterans are usually conducted on<br />

the Colorado potato beetle because these beetles are of<br />

economic importance and are a major pest in Asia,<br />

Europe and North America (Hare, 1990). These beetles<br />

are also the standard beetles used in bioassays to<br />

determine the International Units (IU) of toxicity (Navon,<br />

2000). However, it is absent from South Africa, and<br />

therefore T. molitor was used as the test organism<br />

because it is readily available in South Africa and easy to<br />

rear (Hinze, 2000).<br />

The aim of this research was to determine and<br />

compare the level of toxicity of five endospore-forming,<br />

entomopathogenic bacterial isolates, namely NDR1,<br />

NDR2, NDR3, NDR5 and NDR11.<br />

MATERIALS AND METHODS<br />

Samples<br />

Bacterial isolates were obtained from diseased insects such as T.<br />

molitor, Coleoptera adults and larvae of Schizonycha spp.


collected in sugarcane areas in KwaZulu-Natal (KZN), and debris<br />

from insect-rich environments such as compost, grain dust from<br />

grain storage bins and T. molitor cultures.<br />

Sample collection<br />

Mushroom compost was sampled from a mushroom farm (Karkloof,<br />

KwaZulu-Natal), compost from an urban garden (Hillcrest, KwaZulu-<br />

Natal) and grain dust was sampled from a chicken grain storage<br />

facility (Pietermaritzburg, KwaZulu-Natal). Diseased T. molitor<br />

larvae were collected from the insect rearing facility (University of<br />

KwaZulu-Natal). Adult beetles were collected from light traps in<br />

sugarcane areas in the KwaZulu-Natal midlands by employees of<br />

the South African Sugar Research Institute (SASRI, Mt. Edgcombe,<br />

KZN). White grubs were collected from sugarcane areas by staff<br />

members of SASRI and were delivered in plastic containers filled<br />

with soil. Diseased grubs were removed from these containers and<br />

placed into sterile Petri dishes. All samples were stored in a<br />

refrigerator kept at 4°C.<br />

Isolation of endospore-forming bacteria<br />

Isolation of Bacillus spp. was conducted using a similar<br />

pasteurization method to that described by Ohba and Aizawa<br />

(1978). Suspensions were shaken vigorously for 30 s at full speed<br />

on a vortex shaker. Mixed suspensions were then left to stand for<br />

1 h at room temperature. Suspensions were then vortexed a<br />

second time at full speed for 30 s and then subjected to a<br />

pasteurization process in a preheated water bath for 10 min at<br />

80°C. After pasteurization 0.1 ml aliquots of the suspensions were<br />

plated out onto nutrient agar (Biolab, Biolab Diagnostics, 259<br />

Davidson Road, Wadeville, Johannesburg) (Thiery and Frachon,<br />

1997).<br />

Plates were incubated for 24 h at 30°C in an incubator and<br />

examined for colonies with typical B. cereus morphology (fast<br />

growing, rhizoid, irregular, raised, smooth, and opaque white) or<br />

‘colonies with an ‘ice crystal’ appearance wider than 2 mm<br />

(Damgaard et al., 1997; Prescott et al., 1999; Selvakumar et al.,<br />

2007). Various other white colored bacterial colonies larger than<br />

2 mm in diameter were selected for microscopic evaluation. The<br />

number of colonies isolated from each isolate was estimated and<br />

counted using a colony counter. Selected colonies were then<br />

purified by sub-culturing onto nutrient agar plates and incubated for<br />

3 days at 30°C in an incubator. Colonies were stored on nutrient<br />

agar slants. Not all colonies from each sample that fitted the<br />

description above were selected due to the large number of<br />

colonies formed.<br />

Selected isolates were subjected to Gram staining as well as<br />

Coomassie Blue staining [Coomassie Blue stain 0.133% (w/v) and<br />

methanol 50% (v/v)] for 1 min. Slides were then rinsed gently for<br />

30 s with distilled water and blotted dry with tissue paper (Ammons<br />

et al., 2002). Isolates were screened for the presence of crystal<br />

proteins using light microscopy with a 100 x oil immersion objective<br />

lens (Bernhard et al., 1997; Young et al., 1998). Crystal proteins<br />

stained as dark blue structures (Ammons et al., 2002). Isolates<br />

were screened for size of cells, spore morphology and the presence<br />

of crystal proteins. Isolates without crystal proteins, but with cells<br />

broader than 0.9 µm, were also selected. This was done in order to<br />

include B. cereus, which does not produce crystal proteins. Rodshaped<br />

bacterial cells containing oval spores were selected<br />

because they fit the description of B. cereus, B. thuringiensis and B.<br />

laterosporus cells. Selected cultures were purified by sub-culturing<br />

and were assigned numbers NDR1-NDR14.<br />

Multiple dose bioassay<br />

A pure culture of T. molitor was obtained from a pet shop in<br />

Rand and Laing 2223<br />

Pietermaritzburg, KZN, SA. The culture was reared on a diet of<br />

commercial chicken meal (Meadow Feeds, P.O. Box 426,<br />

Pietermaritzburg, SA) (Hinze, 2000). Bran was not used because<br />

this contains phytic acid that affects the absorption of calcium which<br />

is and essential mineral for a healthy T. molitor culture (Hinze,<br />

2000). T. molitor larvae were reared in 350 mm x 250 mm square<br />

plastic containers containing chicken meal to a depth of 70 mm. For<br />

moisture, potatoes skins, cabbage leaves and carrot peels were<br />

added once a week (Hinze, 2000). Carrots are an essential additive<br />

because most of the micronutrients required by T. molitor larvae are<br />

present in carrots (Hinze, 2000). Adults and pupae were removed<br />

carefully with forceps from the cultures on a regular basis and<br />

placed into in 350 mm x 250 mm square plastic containers<br />

containing chicken meal to a depth of 70 mm. After 14 mo of<br />

rearing, a population of T. molitor larvae was available that was<br />

large enough for the planned bioassays. It was not possible to<br />

determine the instars’ stages because meal worms have between<br />

10 and 14 instars, and several instars occur with larvae in the same<br />

size range (25 mm in length) (Anonymous, 2008a). Therefore,<br />

larvae sized between 18-20 mm were used in the bioassays.<br />

Test organism preparation<br />

T. molitor larvae were carefully removed from the media by gently<br />

sifting the chicken meal through a sieve. This procedure separated<br />

the T. molitor larvae and the chicken meal effectively. The T. molitor<br />

larvae, sized between 18-20 mm, were placed in a division of a<br />

plastic ice tray, for counting purposes, prior to being inoculated with<br />

the bacterial isolates (NDR1, NDR2, NDR3, NDR5 and NDR11).<br />

Isolates NDR1, NDR3 and NDR11 were isolates of B. thuringiensis.<br />

NDR2 was an isolate of B. laterosporus, formerly Bacillus<br />

laterosporus (De Oliviera et al., 2004). NDR5 was an isolate of<br />

B. cereus.<br />

Inoculum preparation<br />

Bacterial isolates NDR1, NDR2, NDR3, NDR5 and NDR11 were<br />

used to inoculate 150 ml of sterile tryptone soy broth (TSB) (Biolab,<br />

Biolab Diagnostics, 259 Davidson Road, Wadeville, Johannesburg)<br />

and incubated in a shaker water bath at 250 rpm for 5 d at 30°C<br />

(Meadows et al., 1992). A viable spore count was conducted using<br />

a standard viable spore technique (Wheelis and Segel, 1979). The<br />

concentration of the viable spores was measured in colony forming<br />

units (CFUs). The final whole culture (FWC), which consisted of<br />

spores and crystal proteins, was used for inoculation. For NDR5<br />

only spores are present because this isolate does not produce<br />

crystal proteins.<br />

Inoculation<br />

Discs with an 18 mm diameter were cut out of the inner leaves of<br />

freshly purchased cabbage using a pre-sterilized test tube cap. The<br />

FWC of NDR1, NDR2, NDR3, NDR5 and NDR11 were diluted with<br />

sterile distilled water to make up concentrations ranges of 20, 40,<br />

60, 80 and 100%. Cabbage discs were dipped into the whole cell<br />

bacterial suspensions and fed to the T. molitor larvae in Petri<br />

dishes. The trials were conducted in sterile Petri dishes. Control<br />

discs were dipped into sterile distilled water. Twenty larvae per<br />

dose in replicates of five were used for each bioassay for NDR1,<br />

NDR2, NDR3 and NDR11. A total of 28 larvae per dose in<br />

replicates of five were used for NDR5. Dead larvae turned black<br />

and liquefied internally. Koch’s postulate was used to determine if<br />

the isolates were responsible for the insect deaths (Prescott et al.,<br />

1999). The amount of cabbage disc consumption (mm) was a<br />

general observation and was not recorded because larval mortality


2224 Afr. J. Microbiol. Res.<br />

Table 1. (LSTATS) PROBAN analysis of three replicate bioassays of five spore-forming bacteria tested against Tenebrio<br />

molitor, with regression parameters displaying the efficacy of these assays.<br />

Isolate No of larvae affected LC50 a 95% Fl b Slope X 2 (df-13)<br />

NDR1 n<br />

Assay 1 68 7.495 7.414 - 7.548 5.45 1.42<br />

Assay 2 55 7.391 7.256 - 7.497 2.54 3.47<br />

Assay 3 61 7.332 7.176 - 7.436 2.59 3.13<br />

Mean 61 7.406 7.282 - 7.494 3.53 -<br />

NDR2 n<br />

Assay 1 63 6.572 6.488 - 6.630 4.65 2.47<br />

Assay 2 68 6.507 6.358 - 6.603 3.02 0.36<br />

Assay 3 65 6.512 6.388 - 6.600 3.26 2.36<br />

Mean 65 6.530 6.411 - 6.611 3.64 -<br />

NDR3 n<br />

Assay 1 49 7.219 7.092 - 7.344 2.35 2.65<br />

Assay 2 46 7.281 7.170 - 7.403 2.56 1.32<br />

Assay 3 53 7.207 7.084 - 7.312 2.63 5.27<br />

Mean 49 7.236 7.115 - 7.353 2.51 -<br />

NDR5 n<br />

Assay 1 51 7.904 7.786 - 8.070 1.95 4.56<br />

Assay 2 49 7.927 7.809 - 8.101 1.97 2.04<br />

Assay 3 56 7.963 7.832 - 8.142 1.71 3.66<br />

Mean 52 7.931 7.809 - 8.104 1.88 -<br />

NDR11 n<br />

Assay 1 57 7.095 6.953 - 7.199 2.57 2.62<br />

Assay 2 48 7.178 7.087 - 7.266 3.35 2.48<br />

Assay 3 50 7.146 7.037 - 7.248 2.79 0.63<br />

Mean 52 7.140 7.026 - 7.237 2.90 -<br />

a Lethal concentration (LC50); b 95% fiducial limits were log transformed concentrations of bacterial spores per ml; n 20 larvae per<br />

dose in replicates of five (total of 100 larvae per bioassay).<br />

was the objective of this study. Results were recorded after 5 days.<br />

Statistical analyses<br />

The statistical computer programs (LSTATS) P/PROBAN Version<br />

2.1 (1992), as programmed by Van Ark (1983), and SPSS Version<br />

11.5, were used to calculate the regression parameters that<br />

included the determination of lethal concentration (LC50) and their<br />

fiducial limits. Each sample reflected three estimates of LC50 and<br />

fiducial limits. These were compared using a standard one-way<br />

ANOVA. A similar format to that of Hatting (2002) was used to<br />

depict the results obtained (Table 1). The LC50 values were<br />

converted back to concentrations of viable spores to determine the<br />

50% mortality dose.<br />

RESULTS<br />

Mortality of T. molitor larvae in the control Petri dishes<br />

was zero, and the cabbage discs were completely<br />

consumed after 5 days. The complete consumption of the<br />

cabbage discs indicates that no toxic effects were<br />

present in the control. Cabbage discs that were<br />

inoculated with different doses of the various bacterial<br />

isolates were consumed according to dose. An overall<br />

trend observed was that the more concentrated the<br />

inocula of spore-forming bacteria applied to the cabbage<br />

disc, the less the cabbage disc was consumed. Low<br />

bacterial dilutions resulted in levels of cabbage disc<br />

consumption similar to that of the controls but with a low<br />

level of T. molitor mortality. No further larval deaths were<br />

found to occur after 5 days.<br />

The minimum LC50 was 6.530 for NDR2 at a<br />

concentration equal to 3.388 x 10 6 spores ml -1 . The<br />

maximum LC50 was 7.931 (NDR5) at a concentration of<br />

8.531 x 10 7 spores ml -1 (Table 1). NDR5 could thus be<br />

considered the most effective isolate. A comparative<br />

bioassay with B. thuringiensis subsp. tenebrionis was not<br />

conducted because this strain is not a registered biocontrol


Rand and Laing 2225<br />

Table 2. (LSTATS) PROBAN hypothesis test used to determine the homogeneity between the five independent bioassays<br />

using NDR1, NDR2, NDR3, NDR5 and NDR11.<br />

Hypothesis Slopes equal Slopes and intercepts equal<br />

Isolate X 2 Df P X 2 df P<br />

NDR1 0.341 2 0.842 5.404 2 0.065<br />

NDR2 1.445 2 0.49 2.007 2 0.368<br />

NDR3 0.73 2 0.699 0.121 2 0.932<br />

NDR5 0.256 2 0.876 0.317 2 0.851<br />

NDR11 0.846 2 0.661 0.793 2 0.678<br />

Table 3. Results of a parallelism test between the three assays of the five bacterial isolates to determine homogeneity.<br />

Isolate No. of larvae affected Slope ± SE LC50 a 95% Fl X 2 (df-13) G b<br />

NDR1 184 2.82 ± 0.37 7.386 7.314 - 7.443 15.781 0.068<br />

NDR2 196 3.39 ± 0.40 6.530 6.468 - 6.580 7.271 0.055<br />

NDR3 148 2.50 ± 0.34 7.235 7.172 - 7.297 10.132 0.071<br />

NDR5 156 1.85 ± 0.22 7.933 7.861 - 8.021 11.091 0.053<br />

NDR11 155 2.87 ± 0.35 7.140 7.082 - 7.194 8.031 0.058<br />

a The data of the three bioassays were pooled to obtain a more accurate estimation of the LC50 and fiducial limits. b The Fiducial limits<br />

calculated by Fieller’s theorem as a measurement of variation.<br />

agent in SA and is not readily available. B. thuringiensis<br />

subsp. azawai and B. thuringiensis subsp. kurstaki are<br />

the only two subsp. of B. thuringiensis registered in SA<br />

for use as a bio-pesticide (Anonymous, 2008b).<br />

Numbers of larvae used in the assays were either 20<br />

for NDR1, NDR2, NDR3, NDR5 and NDR11 (per single<br />

dose). These were the largest numbers possible at the<br />

time, in this project, due to limited insect rearing facilities,<br />

financial resources and time constraints.<br />

One–way ANOVA was used to determine whether the<br />

slopes and intercepts of the three bioassays of each of<br />

the isolates were comparable. Deviations of the slopes<br />

and intercepts from the Probit lines were homogenous.<br />

These lines were therefore comparable (Tables 2 and 3,<br />

Figure 1).<br />

DISCUSSION<br />

Statistical results<br />

The chi-squared values obtained for each of the<br />

bioassays suggested that the deviations of the observed<br />

mortalities are within the range of accepted parameters<br />

for the calculated Probit line (Table 1) (Van Ark, 1983).<br />

Therefore, it may be concluded that the calculated Probit<br />

line is an acceptable representation of insect response to<br />

B. thuringiensis isolates. Parallelism tests showed that no<br />

significant differences could be detected between the five<br />

bioassays for each of the isolates. This allowed for the<br />

comparison of slopes and intercepts (Table 3). The<br />

slopes of the independent bioassays showed no<br />

significant differences and were found to be homogenous<br />

hence the lines were comparable (Table 2) (Van Ark,<br />

1983). The deviations from the observed mortalities were<br />

within the expected limitations of deviation thus rendering<br />

the Probit line acceptable (Table 2 and Figure 1) (Van<br />

Ark, 1983). Values obtained indicated that the lines for<br />

each of the five bioassays for each isolate were parallel<br />

and hence a similar response was observed for each of<br />

the isolates (Table 2 and Figure 1). The LC50 spore<br />

concentrations for each of the isolates were: NDR1 =<br />

2.432 x 10 7 , NDR2 = 3.388 x 10 6 , NDR3 = 1.718 x 10 7 ,<br />

NDR5 = 8.579 x 10 7 and NDR11 = 1.380 x 10 7 .<br />

G is used as a measure of variation in the calculation of<br />

fiducial limits and is derived from Fieller’s Theorem (Van<br />

Ark, 1983). According to Finney (1971), in a good<br />

bioassay, the value of G will lie between 0.2 and 0.05.<br />

Van Ark (1983) suggested using G values of 0.25 and<br />

0.025. G values above 0.025 indicate that the variation of<br />

mortality is high. Values above 0.25 indicate that the<br />

experimental design is not appropriate and requires<br />

amendment. The fiducial limits cannot be calculated for<br />

assays where the G value is equal to one (Van Ark,<br />

1983). The G values for all five isolates fell within the<br />

parameters set by Van Ark as well as by Finney.<br />

However, the experimental precision was not ideal<br />

because the G values were larger than 0.025, indicating<br />

that the variation in mortalities was large. However, the<br />

values were still acceptable because they did not exceed<br />

0.25 (Table 3).<br />

These results could be improved by conducting six or


2226 Afr. J. Microbiol. Res.<br />

Empirical probit mortality<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Series1<br />

Series2<br />

Series3<br />

Series4<br />

Series5<br />

y = 2.8157x - 15.798<br />

y = 2.5895x - 13.764<br />

y = 1.8497x - 9.6744<br />

y = 3.3919x - 17.15<br />

y = 2.8998x - 15.682<br />

0 1 2 3 4 5 6 7 8 9<br />

Log dose<br />

Figure 1. Separate Probit lines of the pooled data for each of the five isolates. 1. Series 1 = NDR1; Series 2 = NDR2;<br />

Series 3 = NDR3; Series 4 = NDR5 and Series 5 = NDR11.<br />

more bioassays per isolate. This would result in a better it<br />

of line and hence a lower G value (Van Ark, 1983). The<br />

number of insects required in a bioassay for reliable<br />

results depends on the experimental procedures, as well<br />

as the species of insect (Van Ark, 1983).<br />

Toxicity comparison of isolates<br />

The isolate with the best LC50 was NDR2, with a log<br />

value of 6.530 (3.388 x 10 6 spores ml -1 ) (Table 3). This<br />

isolate was identified previously as an isolate of<br />

B. laterosporus. Assays conducted with isolates of<br />

B. laterosporus did not exhibit any toxicity towards<br />

T. molitor. However, toxicity was evident against<br />

mosquitoes (Culex quinquefasciatus and Aedes aegypti)<br />

and a species of snail Biomphalaria glabrata (Favret and<br />

Yousten, 1985). Assays conducted by Rivers et al. (1991)<br />

on T. molitor with strains of B. laterosporus showed these<br />

strains to have toxicity values similar to those obtained<br />

from strains of B. thuringiensis subsp. tenebrionis.<br />

The isolate with the weakest LC50 was NDR5, with a<br />

log value of 7.933 (8.579 x 10 7 spores ml -1 ). This isolate<br />

was identified previously as an isolate of B. cereus. This<br />

species is not often used in entomopathogenic bioassays<br />

because it is considered an organism associated with<br />

gastrointestinal diseases and is often found as a food<br />

contaminant. However, B. cereus strains with insecticidal<br />

properties against coleopteran larvae have been isolated<br />

from species of white grub (Selvakumar et al. 2007;<br />

Sushil et al., 2008). The insecticidal properties of this<br />

organism lie in its ability to produce vegetative<br />

insecticidal proteins (VIP) (Estruch et al., 1996; Moar et<br />

al., 1994; Yu et al., 1997) which have been found to be<br />

effective against Western Corn Rootworm, Diabrotica<br />

virgifera LeConte (Coleoptera, Chrysomelidae) (Warren,<br />

1997).<br />

All five bacterial isolates (NDR1, NDR2, NDR3, NDR5<br />

and NDR11) displayed toxicity against T. molitor. The<br />

three B. thuringiensis isolates (NDR1, NDR3 and NDR11)<br />

displayed high levels of toxicity to T. molitor but were less<br />

toxic than the B. laterosporus isolate (NDR2) (Table 3).<br />

The dose response of the B. cereus isolate (NDR5) was<br />

found to stand alone from the other two species tested<br />

(Figure 1). This is indicative of substantially greater<br />

toxicity than the other isolates.<br />

Standardization assays often involve the use of purified<br />

or extracted crystal proteins (Cry and Cyt toxins) and do<br />

not include any of the other toxins and synergists (e.g.,<br />

chitinases) produced by these species of bacteria


(Thamthiankul et al., 2001; Lee et al., 2007). B.<br />

thuringiensis and B. cereus are able to synthesize<br />

chitinases (Lee et al., 2007). Other toxins may play an<br />

important role in preventing target insects from becoming<br />

resistant because they would have to evolve resistance<br />

against more than one compound concurrently. In the<br />

bioassays used in this research, the FWC were used in<br />

toxicity determination. Hence, other unknown toxins and<br />

synergistic factors were included. Whole cultures<br />

consisting of spore and crystal suspensions are often<br />

used in bioassays to determine toxicity. The use of FWCs<br />

is applied where whole culture products will be<br />

commercialized, as opposed to pure protein crystals (De<br />

Oliviera, 2004; Lambert et al., 1992). Whole culture<br />

products have to be screened for beta-exotoxins because<br />

current registration of B. thuringiensis products requires<br />

the absence of these toxins (Prieto-Samsonova et al.,<br />

1997). Continuous sub-culturing of B. thuringiensis can<br />

be problematic because it may cause a decline in toxicity<br />

(Sachidanandham and Jayaraman, 2003).<br />

ACKNOWLEDGEMENTS<br />

Authors are thankful to M. Ohba and K. Kagoshima for Hserotyping<br />

of some of the isolates and to the team from<br />

The South African Sugar Research Institute (SASRI) for<br />

the collection and delivery of the white grubs and adult<br />

beetles.<br />

REFERENCES<br />

Ammons D, Rampersad J, Khan A (2002). Usefulness of staining<br />

parasporal bodies when screening for Bacillus thuringiensis. J.<br />

Invertebr. Pathol., 79: 203-204.<br />

Anonymous(2008a). www.wikipedia.org (12/12/08)<br />

Anonymous (2008b). www.pesticideinfo.org (21/12/08)<br />

Bernhard K, Jarrett P, Meadows M, Butt J, Ellis DJ, Roberts GM, Pauli<br />

S, Rodgers P, Burges HD (1997). Natural isolation of Bacillus<br />

thuringiensis: Worldwide distribution, characterization, and activity<br />

against insect pests. J. Invertebr. Pathol., 70: 59-68.<br />

Damgaard PH, Hansen BM, Pedersen RC Eilenberg J (1997). Natural<br />

occurrence of Bacillus thuringiensis on cabbage foliage and in insects<br />

associated with cabbage crops. J. Appl. Microbiol., 82: 253-258.<br />

De Oliviera, EJ, Rabinovitch L, Monnerat RG, Passos LKJ, Zahner V<br />

(2004). Molecular characterization of Brevibacillus laterosporus and<br />

its potential use in biological control. Appl. Environ. Microbiol., 70:<br />

6657-6664.<br />

Estruch JJ, Warren GW, Mullins MA, Nye GJ, Craig JA, Koziel MG<br />

(1996). Vip3A, a novel Bacillus thuringiensis vegetative insecticidal<br />

protein with a wide spectrum of activities against lepidopteran<br />

insects. Proc. Nat. Acad. Sci., 93: 5389-5394.<br />

Favret ME, Yousten AA (1985). Insecticidal activity of Bacillus<br />

laterosporus. J. Invertebr. Pathol., 45: 195-203.<br />

Ferré J, Van Rie, J (1992). Biochemistry and genetics of insect<br />

resistance to Bacillus thuringiensis. Ann. Rev. Entomol., 47: 501-533.<br />

Finney DJ (1971). Statistical Method in Biological Assays. Second<br />

edition. Griffin, London, UK.<br />

Hare JD (1990). Ecology and maintenance of the Colorado potato<br />

beetle. Ann. Rev. Entomol., 35: 81-100.<br />

Hatting JL (2002). Fungal cereal parasitism of cereal aphids in South<br />

Africa. PhD thesis. University of Natal, Pietermaritzburg, South<br />

Africa.<br />

Rand and Laing 2227<br />

Herrnstadt C, Soares GG, Wilcox GR, Edwards DL (1986). A new strain<br />

of Bacillus thuringiensis with activity against Coleopteran insects. Bio-<br />

Technology., 4: 305-308.<br />

Hinze I (2000). A guide to keeping and breeding waxbills in a British bird<br />

room. Part III. Cultivating live food. Avizadium., 12: 14-16.<br />

Kriege A, Huger AM, Langernbruch GA, Schnetter W (1983). Bacillus<br />

thuringiensis var. tenebrionis: A new pathotype effective against<br />

larvae of Coleoptera. J. Appl. Entomol., 96: 500-508.<br />

Lambert B, Höfte H, Annys K, Janssens S, Soetaert P, Peferoen M<br />

(1992). Novel Bacillus thuringiensis insecticidal crystal proteins with a<br />

silent activity against Coleopteran larvae. Appl. Environ. Microbiol.,<br />

58: 2536-2542.<br />

Lee HS, Jang JS, Choi SK, Lee DW, Kim EJ, Jung HC, Pan JG (2007).<br />

Identification and expression of GH-8 family chitosanases from<br />

several Bacillus thuringiensis subspecies. FEMS. Micro. Let.,<br />

277: 133-141.<br />

Meadows MP, Ellis DJ, Butt J, Burges HD (1992). Distribution,<br />

frequency, and diversity of Bacillus thuringiensis in an animal feed<br />

mill. Appl. Environ. Microbiol., 58: 1344-1350.<br />

Moar, WJ, Trumble JT, Hice RH, Backman PA (1994). Insecticidal<br />

activity of the CryIIA protein from the NRD-12 isolate of Bacillus<br />

thuringiensis subsp. kurstaki expressed in Escherichia coli Migula<br />

and Bacillus thuringiensis and in a leaf colonizing strain of Bacillus<br />

cereus. Appl. Environ. Microbiol., 60: 896-902.<br />

Nault BA, Kennedy GG (1999). Influence of foliar-applied Bacillus<br />

thuringiensis subsp. tenebrionis and early potato harvest on<br />

abundance and overwinter survival of Colorado potato beetles<br />

(Coleoptera: Chrysomelidae) in North Carolina. Hortic. Entomol., 92:<br />

1165-1171.<br />

Navon A (2000). Bioassays of entomopathogenic microbes and<br />

nematodes: In Navon A, Ascher KRS (eds) Bioassays of Bacillus<br />

thuringiensis, Part 1. CABI, New York, USA, pp. 1-24.<br />

Ohba M, Aizawa K (1978). Serological identification of Bacillus<br />

thuringiensis and related bacteria isolated in Japan. J. Invertebr.<br />

Pathol., 32: 303-309.<br />

Ohba, M, Iwahana H, Asano S, Suzuki N, Sato R, Hori H (1992). A<br />

unique isolate of Bacillus thuringiensis serovar japonensis with a high<br />

larvicidal activity specific for scarabaeid beetles. Lett. Appl.<br />

Microbiol., 14: 54-57.<br />

Prescott LM, Harley JP, Klien DA (1999). Microbiology. Fourth Edition.<br />

WCB McGraw-Hill, New York.<br />

Prieto-Samsonova DL, Vasquez-Parelan RI, Ayra-Pardo C, Gonzalez-<br />

Cabera J, de la Riva GA (1997). “Bacillus thuringiensis: From<br />

biodiversity to biotechnology.” J. Ind. Microbiol. Biotechnol., 19: 202-<br />

219.<br />

Rivers DB, Vann CN, Zimmack HL, Dean DH (1991). Mosquitocidal<br />

activity of Bacillus laterosporus. J. Invertebr. Pathol., 58: 444-447.<br />

Sachidanandham R, Jayaraman K (2003). Formation of spontaneous<br />

asporogenic variants of Bacillus thuringiensis subsp. galleriae in<br />

continuous cultures. Appl. Microbiol. Biotechnol., 40: 504-507.<br />

Selvakumar G, Mohan M, Sushil SN, Kundu S, Bhatta JC, Gupta HS<br />

(2007). Characteristic and phylogenic analysis of an<br />

entomopathogenic Bacillus cereus strain WGPSB-2 (MTCC 7182)<br />

isolated from white grub, Anomala dimidiate (Coleoptera:<br />

Scarabaeidae). Biocontrol Sci., 17: 525-534.<br />

Sushil NS, Mohan M, Selvakumar G, Bhatt JC, Gupta HS (2008).<br />

Isolation and toxicty evaluation of bacterial entomopathogens against<br />

phytophagous white grubs (Coleoptera: Scarabaeidae) in Indian<br />

Himalayan hills. Int. J. Pest Manage., 54: 301-307.<br />

Tabashnik BE (1994). Evolution of resistance to Bacillus thuringiensis.<br />

Ann. Rev. Entomol., 39: 47-79.<br />

Thamthiankul S, Suan-Ngay S, Tantimavanich S, Panbangred, W<br />

(2001). Chitinase from Bacillus thuringiensis subsp. pakistani. Appl.<br />

Microbiol. Biotechnol., 56: 395-401.<br />

Thiery I, Frachon E (1997). Identification, isolation, culturing and<br />

preservation of entomopathogenic bacteria. In Lacey L (ed) Manual<br />

of Techniques in Insect Pathology. <strong>Academic</strong> Press, London, UK, pp.<br />

55-77.<br />

Van AH (1983). Introduction to Probit analysis with (LSTATS)<br />

P/PROBAN. Science Bulletin, Department of Agriculture, Rep. South<br />

Afr., 399: 1-16.<br />

Whaldon ME, Miller DL, Hollingworth RM, Grafius EJ, Miller JR (1993).


2228 Afr. J. Microbiol. Res.<br />

Selection of Colorado potato beetle (Coleoptera: Chrysomelidae) strain<br />

resistant to Bacillus thuringiensis. J. Econ. Entomol., 86: 226-233.<br />

Warren GW (1997). Vegetative insecticidal proteins: novel proteins for<br />

control of corn pests, p. 109-121. In Carozzi N, Koziel M (ed)<br />

Advances in Insect Control: the Role of Transgenic Plants. Taylor<br />

and Francis, London, UK, pp. 109-121.<br />

Wheelis ML, Segel WP (1979). Introduction to the Microbial World. A<br />

Laboratory Manual. Prentice-Hall, New Jersey.<br />

Young JM, Chilcott CN, Broadwell A, Wigley AJ, Lecadet MM (1998).<br />

Identification of serovars of Bacillus thuringiensis Berliner 1915 in<br />

New Zealand. N. Z. J. Crop Hortic. Sci., 26: 63-68.<br />

Yu CG, Mullins MA, Warren GW, Koziel MG, Estruch JJ (1997). The<br />

Bacillus thuringiensis vegetative insecticidal protein VIP3A lyses<br />

midgut epithelium of susceptible insects. Appl. Environ. Microbiol.,<br />

65: 532-536.


African Journal of Microbiology Research Vol. 5(16), pp. 2229-2233, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.108<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

What changed in necrotizing fasciitis in twenty-five<br />

years?<br />

Baris Yildiz 1 *, Derya Karakoc 2 , Erhan Hamaloglu 2 , Arif Ozdemir 2 and Ahmet Ozenc 2<br />

1 Ankara Numune Teaching and Research Hospital, 6th General Surgery, Ankara, Turkey.<br />

2 Department of General Surgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey.<br />

Accepted 30 June, 2011<br />

Necrotizing fasciitis (NF) is a deadly soft tissue infection characterized by necrosis of subcutaneous<br />

tissues. In this study, our aim was to identify variables affecting patient outcome and mortality in<br />

necrotizing fasciitis and their temporal changes. We reviewed records of 45 patients treated at our<br />

institution between 1979 and 2004. Data about gender, age, etiology, site of involvement, bacteriology,<br />

type of surgery, supportive treatment, accompanying diseases, mortality were collected. Factors<br />

contributing to mortality were sepsis, renal failure, liver failure, multi organ failure, disseminated<br />

intravascular coagulopathy and long term intubation. Mortalities accumulated in first 23 patients. There<br />

was not difference in microbiology, demographics, etiology, site of involvement, debridement technics<br />

between first patients and recent patients of the institution. Mortality in necrotizing fasciitis is mostly<br />

because of sepsis and associated disorders. Adequate control of the microbiological agent and<br />

preventing further contamination of the wound is cardinal part of treatment in NF.<br />

Key words: Necrotizing fasciitis, bacteriology, mortality, sepsis, Fournier’s gangrene.<br />

INTRODUCTION<br />

Necrotizing fasciitis (NF) is deeply located infection of<br />

subcutaneous tissue resulting in progressive destruction.<br />

Its histological patterns are large necrosis of the fascia,<br />

subcutaneous fat and skin with thrombosis of the<br />

microvasculature and sparing of the underlying muscles<br />

and the skin (Wilson, 1952; Giuliano et al., 1977; Misago<br />

et al., 1996).<br />

NF can be divided into two groups depending on the<br />

causative organism. In type I, at least one anaerobic<br />

species is isolated in combination with facultative<br />

anaerobic species and members of Enterobacteriaceae.<br />

In type II, group A streptococci or Staphylococcus aureus<br />

are isolated (Levine and Manders, 2005).<br />

There are a number of predisposing factors to NF such<br />

as diabetes mellitus (DM) (Salcido, 2007) immunocompromised<br />

state, corticosteroid use, intravenous drug<br />

abuse, trauma, malnutrition, burns and atherosclerosis<br />

(Thompson et al., 1993). Early diagnosis and prompt<br />

*Corresponding author. E-mail: baris104@yahoo.com. Tel: +90-<br />

532- 445 46 55. Fax: +90-312- 424 15 20.<br />

Abbreviations: NF, Necrotizing fasciitis; DM, diabetes mellitus.<br />

treatment consisting of surgery and antibiotics are key to<br />

improved patient outcome in NF.<br />

Depending on the depth of invasion, necrotizing soft<br />

tissue infections can cause extensive local tissue<br />

destruction, tissue necrosis, systemic toxicity and even<br />

death. Mortality rates for necrotizing soft tissue infections<br />

range from 6% to as high as 76% despite advances in<br />

management (Brandt et al., 2000; Singh et al., 2002;<br />

Bilton et al., 1998).<br />

In this study, our aim was to identify the temporal<br />

changes in incidence, etiology, bacteriology, management<br />

and outcome in NF at our institution to help define<br />

better strategies in treatment for this challenging<br />

condition.<br />

MATERIALS AND METHODS<br />

After ethics commitee approval was obtained, inpatient records of<br />

patients treated for necrotizing fasciitis between 1979 and 2004 in<br />

our university hospital general surgery department were retrospectively<br />

reviewed. Patients were identified from hospital archives<br />

and data was collected from hospital records and electronic<br />

database. There were 29 men and 16 women whose ages ranged<br />

between 16 and 88 years. We reviewed the charts of the patients


2230 Afr. J. Microbiol. Res.<br />

and recorded sex, age, predisposing factors, duration between the<br />

onset of symptoms and the first surgical debridement, bacteria<br />

isolated, duration of hospital stay, reconstructive proce-dures, and<br />

clinical outcome. Plastic and Reconstructive Surgery at our hospital<br />

was consulted for patients who needed reconstructive procedures<br />

after initial debridement was performed in General Surgery<br />

Department. Broad spectrum antibiotics were given initially and<br />

specific antibiotics were advised after confirmation of the organism<br />

isolated from the wound tissue. Patients with minor skin and softtissue<br />

loss or wound healing with secondary intention were referred<br />

for wound evaluation and wound care. The scrotal advancement<br />

flap, pudendal thigh fasciocutaneous flap, gracilis<br />

muscle/myocutaneous flap, or split-thickness skin graft was used as<br />

the reconstructive procedure to correct the defects arising from<br />

failure of secondary intention or primary closure. The statistical<br />

analysis was carried out with SPSS software (SPSS, Chicago,<br />

Illinois) using Chi-Square test. A P-value < 0.05 was considered<br />

significant.<br />

RESULTS<br />

There were totally 45 patients. The median age of the<br />

patients was 50 (minimum 16, maximum 88). Sixteen<br />

patients (35.6%) were female and 29 (64.4%) were male.<br />

The diagnoses of NF were established based on clinical<br />

symptoms and findings including combinations of fever,<br />

pain, swelling, erythematous change of the involved skin<br />

and purulent discharge.<br />

The most common etiology was perianal abcess (n=17,<br />

37.8%) followed by surgical site infection (n=9, %20) and<br />

others (perineal shaving, decubitus ulcer, strangulating<br />

inguinal hernia, vulvar trauma, intramuscular injection,<br />

pilonidal abcess, colocutaneous fistula, diabetic foot, gun<br />

shot wound, enterocutaneous fistula). It was seen that<br />

majority of cases were actually Fournier’s gangrene. This<br />

was supported by the analysis of the site of involvement<br />

as perianal area, scrotum/vulva with highest frequency<br />

(n=16), followed by combinations of localizations<br />

including scrotum, vulva, gluteus and inguinal region.<br />

Twenty-two percent of patients in our study had comorbidities<br />

while thirteen patients (28.9%) did not have<br />

any accompanying disease. The most common associated<br />

disease was DM (17.8%) followed by malignancies<br />

(8.9%), coronary artery disease (6.7%) and combinations<br />

of DM, hypertension, ulcerative colitis, paraplegia,<br />

chronic obstructive airway disease. During management<br />

46.7% of patients had sepsis and 11.1% had multiorgan<br />

failure.<br />

Polymicrobial bacterial strains were isolated from tissue<br />

and pus cultures. In 9 patients there was not any growth<br />

in bacterial cultures. The rest of isolated bacteria are<br />

listed in Table 1.<br />

Antimicrobial agents, surgery and nutritional support<br />

were the mainstay of treatment for patients with NF.<br />

Antibiotic therapy was started empirically and continued<br />

based on the microbiology of the wounds. In 3 patients<br />

antifungal medication was added to antibiotics. Median<br />

duration of antimicrobial therapy was 17 days (minimum<br />

3 – maximum 55).<br />

Surgery was performed as soon as the diagnosis of NF<br />

was suspected. There was delay in 18 patients between<br />

diagnosis and surgical intervention. Median time of delay<br />

was 1.5 days. Debridement under local anesthesia was<br />

performed only to 4.4% of the patients and 95.6% of the<br />

patients had debridement under general anesthesia. Of<br />

those who had debridement under general anesthesia,<br />

27 (60%) had only one debridement while 9 (20%) had<br />

twice and 4 (8.9%) had three times. Overall, colostomy<br />

was performed to 22.2% of patients and to 90% of the<br />

patients with Fournier’s gangrene.<br />

Twenty-six patients (48.8%) required reconstruction.<br />

Primary closure was performed in 15 (57.6%) patients<br />

whereas 4 (15.3%) patients had split-thickness skin<br />

grafting and 3 (11.3%) had flap reconstruction.<br />

Nutritional support was given to 17 patients. Of these,<br />

six patients received total parenteral nutrition whereas 8<br />

had enteral nutrition.<br />

The longest hospital stay was 92 days while the<br />

shortest stay was 9 days (median: 35.5). The overall<br />

mortality was 22.2% (n=10). Twenty-five percent of<br />

women and 20% of men died. Comparison of survivors<br />

and non-survivors are given in Table 2. Statistically<br />

significant factors contributing to mortality were sepsis<br />

(p=0.001), multiorgan failure (p


Table 1. Combinations of bacteriologic strains isolated from tissues in chronological order starting from 1979 to 2004.<br />

Case Year Microorrganisms<br />

Yildiz et al. 2231<br />

1 1979 Enterobacteriaceae spp.<br />

2 1979 Escherichia coli, Clostridium tetani<br />

3 1980 Coagulase positive Staphylococcus aureus (SCP)<br />

4 1983 No growth<br />

5 1983 Escherichia coli, Proteus, SCP, Diphtheroids<br />

6 1985 Escherichia coli, Diphtheroids<br />

7 1986 Escherichia coli, SCP, Pseudomonas aeruginosa<br />

8 1988 SCP, Acinetobacter<br />

9 1991 Klebsiella pneumonia, Pseudomonas aeruginosa<br />

10 1993 Streptococcus pneumonia, SCP<br />

11 1993 No growth<br />

12 1994 No growth<br />

13 1994 Pseudomonas aeruginosa<br />

14 1994 No growth<br />

15 1995 Enterobacteriaceae species<br />

16 1995 No growth<br />

17 1995 Proteus vulgaris<br />

18 1996 Klebsiella oxytoca, Escherichia coli<br />

19 1996 Escherichia coli, Streptococcus viridens<br />

20 1997 Escherichia coli<br />

21 1997 Enterococcus faecalis, Pseudomonas aeruginosa, Klebsiella Oxytoca<br />

22 1997 Escherichia coli, Streptococcus viridens<br />

23 1997 Escherichia coli, Klebsiella pneumonia, Coagulase positive Staphylococcus aureus<br />

24 1998 Escherichia coli, Staphylococcus aureus<br />

25 1998 Morganella morgagni, Pseudomonas aeruginosa<br />

26 1998 Enterobacteriaceae spp.<br />

27 1998 No growth<br />

28 1998 Streptococcus pneumonia, SCP<br />

29 1998 Staphylococcus aureus<br />

30 1998 Escherichia coli, Klebsiella pneumonia<br />

31 1998 No growth<br />

32 1999 Staphylococcus aureus, Serratia marcescens<br />

33 2000 Escherichia coli, Klebsiella pneumonia, Staphylococcus aureus<br />

34 2001 Escherichia coli, Streptococcus agalactiae<br />

35 2001 Escherichia coli, Pseudomonas aeruginosa<br />

36 2002 No growth<br />

37 2002 Acinetobacter, Staphylococcus epidermidis<br />

38 2003 Acinetobacter, Escherichia coli<br />

39 2003 Staphylococcus epidermidis<br />

40 2003 Morganella morgagni<br />

41 2003 Methicillin resistant Staphylococcus aureus, Escherichia coli, Acinetobacter<br />

42 2003 No growth<br />

43 2003 Escherichia coli, Staphylococcus aureus<br />

44 2003 Streptococcus agalactiae<br />

45 2004 Escherichia coli, Enterococcus faecalis, Proteus mirabilis<br />

debrided area (Villanueva-Sáenz et al., 2002). Colostomy<br />

rate of survivors and non-survivors was similar in our<br />

study while there was an increase in tendency to perform<br />

colostomy in the second half of the study group.<br />

The defects after surgical debridement in NF can be<br />

managed with primary closure, grafting or flap<br />

reconstruction (Baharestani, 2008). Our center had a<br />

propensity towards primary closure whenever possible


2232 Afr. J. Microbiol. Res.<br />

Table 2. Comparison of survivors and non-survivors.<br />

Variable Non survivors Survivors<br />

Age (years) 59.7±16.9 45.6±12.6<br />

Gender 6 Males, 4 Females 23 Males, 12 Females<br />

Etiology Predominantly SSI with peritonitis Predominantly perianal abcess<br />

Duration of symptoms (days) 8±4.24 7.71±4.36<br />

Delay in surgery (days) 2.66±2.05 1.66±1.54<br />

Co-morbidity present 60% 60.7%<br />

Site of involvement Predominantly ABW Predominantly perianal area<br />

Number of debridements 1.4±0.51 1.34±0.787<br />

Colostomy 20% 22.9%<br />

Intubation 60% 8.6%<br />

Duration of Intubation (days) 3.66±3.2 0.087±0.288<br />

Red packed cell transfusion (Units) 4.5±1 1.7±2.2<br />

Antibiotic therapy (days) 14.8±10.56 23.18±14.1<br />

TPN administration 20% 20%<br />

Enteral nutrition 10% 20%<br />

Leukocyte count at admission ( /uL ) 21650±12834 16537±6943<br />

Neutropenia 1 patient 1 patient<br />

Renal failure 70% 34.3%<br />

Liver failure 50% 11.4%<br />

Sepsis 100% 31.4%<br />

SSI:Surgical site infection; ABW:Anterior abdominal wall; TPN: Total parenteral nutrition.<br />

Table 3. Characteristics of first 23 and last 22 cases.<br />

Variable First 23 patients (1979-1996) Last 22 patients (1997-2004)<br />

Age (years) 49.56±16.5 47.90±12.95<br />

Gender 65.2% Male 63.6% Male<br />

Etiology Predominantly SSI with peritonitis and perianal abcess Predominantly perianal abcess<br />

Site of involvement Predominantly perianal area Predominantly perianal area<br />

Number of debridements 1.42±0.676 1.28±0.78<br />

Colostomy 15% 33.3%<br />

Mortality 28.6% None<br />

SSI: Surgical site infection.<br />

and all reconstructions were done between 1997 and<br />

2004.<br />

Empirical antibiotic prophylaxis should be started in NF<br />

until the responsible pathogen is identified in cultures. In<br />

our study, when compared with respect to years, recent<br />

regimens started to contain piperacillin-tazobactam,<br />

teikoplanin and imipenem. Of note, 3 patients with sepsis<br />

in the second half of study received fluconazole along<br />

with antibiotics.<br />

Published mortality rates due to NF vary widely. Brandt<br />

et al. (2000) and Lille et al. (1996) from USA reported<br />

24% (Brandt et al., 2000) and 25% (Lille et al., 1996)<br />

respectively while Rietveld et al (1995) from New Zealand<br />

reported 43% (Rietveld et al., 1995). We found<br />

approximately the same result (22.2%) as those reported<br />

from USA.<br />

In our study, factors predicting mortality were mainly<br />

the end results of sepsis and multi-organ failure. In contrary<br />

to what we found, a study from Canada cited that<br />

the contributing factors for mortality were advanced age,<br />

immunocompromised state and presence of<br />

streptococcal toxic shock syndrome (Golger et al., 2007).<br />

In this study, we tried to reflect our 25 years of<br />

experience in NF between 1979 and 2004. Due to<br />

change in institutional policy our department stopped<br />

admitting and following NF patients after 2004.<br />

The first thing in analysis of cases was better mortality<br />

rate in more recent cases when compared to the earlier<br />

days of the institution. In this regard, when we look at the<br />

variables it seems that microbiology, demographics,


etiology, site of involvement, debridement technics are<br />

similar but frequency of colostomy procedure, type of<br />

antibiotics, utilization of antifungals seem to accumulate<br />

in the second chronological half of the series. The main<br />

outcome of our study is the fact that adequate control of<br />

the microbiological agent and preventing further contamination<br />

of the wound is the cardinal part of the treatment in<br />

NF and particularly in Fournier’s gangrene.<br />

REFERENCES<br />

Anaya DA, Dellinger EP (2007). Necrotizing soft-tissue infection:<br />

diagnosis and management. Clin. Infect. Dis., 44(5): 705-710.<br />

Baharestani MM (2008). Negative pressure wound therapy in the<br />

adjunctive management of necrotizing fascitis: Examining clinical<br />

outcomes. Ostomy Wound Manage., 54(4): 44-50.<br />

Bilton BD, Zibari GB, McMillan RW, Aultman DF, Dunn G, McDonald J<br />

(1998). Aggressive surgical management of necrotizing fasciitis<br />

serves to decrease mortality: A retrospective study. Am. Surg., 64(5):<br />

397–401.<br />

Brandt MM, Corpron CA, Wahl WL (2000). Necrotizing soft tissue<br />

infections: A surgical disease. Am. Surg., 66(10): 967–997.<br />

Brandt MM, Corpron CA, Wahl WL (2000). Necrotizing soft tissue<br />

infections: A surgical disease. Am. Surg., 66(10): 967-970.<br />

Elliott DC, Kufera JA, Myers RA (1996). Necrotizing soft tissue<br />

infections. Risk factors for mortality and strategies for management.<br />

Ann. Surg., 224(5): 672-683.<br />

Fulda GJ, Khan SU, Zabel DD (2003). Special issues in plastic and<br />

reconstructive surgery. Crit. Care Clin., 19(1): 91-108.<br />

Yildiz et al. 2233<br />

Giuliano A, Lewis F, Hadley K, Blaisdell FW (1977). Bacteriology of<br />

necrotizing fasciitis. Am. J. Surg., 134(1): 52–57.<br />

Golger A, Ching S, Goldsmith CH, Pennie RA, Bain JR (2007). Mortality<br />

in patients with necrotizing fasciitis. Plast. Reconstr. Surg., 119(6):<br />

1803-1807.<br />

Levine EG, Manders SM (2005). Life-threatening necrotizing fasciitis.<br />

Clin. Dermatol., 23(2): 144-147.<br />

Lille ST, Sato TT, Engrav LH, Foy H, Jurkovich GJ (1996). Necrotizing<br />

soft tissue infections: obstacles in diagnosis. J. Am. Coll. Surg.,<br />

182(1): 7-11.<br />

Misago N, Narisawa Y, Ryu S, Gotoh Y, Tanaka T, Yokoyama M,<br />

Kohda H (1996). Necrotizing fasciitis due to group A streptococci: A<br />

clinicopathological study of six patients. J. Dermatol., 23(12): 876–<br />

882.<br />

Rietveld JA, Pilmore HL, Jones PG, Theis JC, Bowie DA, Dempster AG<br />

(1995). Necrotising fasciitis: A single centre's experience. N Z Med.<br />

J., 108(995): 72-74.<br />

Salcido RS (2007). Necrotizing fasciitis: Reviewing the causes and<br />

treatment strategies. Adv. Skin Wound Care., 20(5): 288-293.<br />

Singh G, Sinha SK, Adhikary S, Babu KS, Ray P, Khanna SK (2002).<br />

Necrotizing Infections of soft tissues: A clinical profile. Eur. J. Surg.,<br />

168: 366–371.<br />

Thompson CD, Brekken AL, Kutteh WH (1993). Necrotizing fasciitis: A<br />

review of management guidelines in a large obstetrics and<br />

gynecology teaching hospital. Infect. Dis. Obstet. Gynecol., 1(1): 16-<br />

22.<br />

Villanueva-Sáenz E, Martínez Hernández-Magro P, Valdés Ovalle M,<br />

Montes Vega J, Alvarez-Tostado FJF (2002). Experience in<br />

management of Fournier's gangrene. Tech. Coloproctol., 6(1): 5-10.<br />

Wilson B (1952). Necrotizing fasciitis. Am. Surg., 18: 416–431.


African Journal of Microbiology Research Vol. 5(16) pp. 2234-2240, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Aspects of bacterial colonization in newborn babies<br />

Fariba Heshmati*, Seyeed Amir Yazdanparast , Seyeed Akbar Moosavi , Hussein Dargahi,<br />

and Farnaz Tabibzadeh<br />

Department of Microbiology, Tehran University of Medical Sciences (Hemmat Campus), Tehran, Iran.<br />

Accepted 23 June, 2011<br />

The study of colonization of microbial flora in neonate is very important and finding the causative agent<br />

may lead to explore these infections in neonate. In this research the colonization in neonate was<br />

measured at the beginning and a few hours after birth. Sampling was performed in operating room,<br />

rooming in and nursery section. Some of the neonates deliver by rapture of membrane in normal way<br />

and the other deliver by caesarean section. All the samples which are collected were from these sites:<br />

1) Forehead 2) Oral cavity 3) External ear 4) Auxiliary region 5) Umbilical region 6) Perennial region.<br />

Also we had some sampling from health care personnel who are worked in operating room or nursery<br />

section. At the time of sampling we used sterile gloves, mask and always sterile sets. Sampling was<br />

done by use of sterile swabs which were weltering by sterile phosphate buffer or sterile normal saline<br />

.Then this sterile swab scrubbed on the special surface of the body of neonate and directly transferred<br />

to BA plate, Mac agar plate, EMB agar plate and Sc agar plate and then streaked on these plates. The<br />

regions which were sampled ,numbered by 1 to 6 then in other hours once more the sampling were took<br />

place from the same site, therefore we began from number 7 to 12 and according to this method for<br />

example sampling from neonate A was showed by A1 to A12. The prevalent microorganisms in oral cavity<br />

were as follow: 63.5% GPC, 18.5% GPB and 11% GNC. Staphylococcus epidermidis was the major<br />

microorganism found in neonate forehead. No bacteria growth was found in 50% of samples through<br />

caesarean section. Staphylococcus and Diphtheroied constitute the highest colonization of neonate<br />

skin's microorganisms and their growth rates were low at the onset of the birth, but increased after 12<br />

hours. Colonization of gram-negative bacteria in infant hours after birth considered to be an important<br />

issue that was studied in this research. Hence hand washing of health-care personnel’s can reduce<br />

such bacteria in infant. There was a significant decrease in (GNB) number due to hand washing, hence<br />

application of disinfectant solution in hand washing play an important role in controlling<br />

microorganisms in care unites.<br />

Key words: Colonization, GPC (Gram- positive cocci), GPB (Gram-positive bacilli), GNC (Gram- negative<br />

cocci), GNB (Gram- negative bacilli), Microorganism.<br />

INTRODUCTION<br />

Microbial colonization of newborn infant begins<br />

immediately after birth. Infants’ skin is colonized by flora<br />

derived from the body of the mother and other human<br />

contacts and from various inanimate objects. Initial<br />

colonization is fortunately, depending on the first suitable<br />

organism to arrive at a particular site as well as factors<br />

such as the type of delivery, the amount of vernix<br />

*Corresponding author. E-mail:<br />

heshmati_99_1999@yahoo.com<br />

caseosa persent at birth, the type of nourishment<br />

received (breast milk or formula) and the degree of<br />

exposure in the hospital environment (Guenthner et al.,<br />

1987). Moreover exclusively formula-fed infants were<br />

more often colonized with E.coli, C.difficle, Bacteroides<br />

and Lactobacillus, when compared with breast-fed<br />

infants. Other factors like hospitalization and prematurity<br />

were also associated with higher prevalence of C. difficil<br />

(Guenthner et al., 1987; Penders et al., 2006). However,<br />

in most cases, after a few days the representation of<br />

microbial species with in the neonatal flora was<br />

remarkably similar to the adult pattern of colonization.


Interest in the bacterial colonization of the newborn was<br />

developed in mid 1940s when epidemic of<br />

Staphylococcal pyoderma began to appear in substantial<br />

number in the developed countries (Falle and Schleifer,<br />

1981; Pezzati et al., 2002; McConnell et al., 2004).<br />

Staphylococcal disease continues to be a serious<br />

problem in some nurseries, but gram-negative bacilli are<br />

currently the major encountered pathogens in hospital<br />

acquired infection (Pass et al., 1980). Infant born through<br />

caesarean section had lower number of Bifidobacteria<br />

and Bacteroides, but more often colonized with C.difficle,<br />

when compared with those borne vaginally (Penders et<br />

al., 2006). Occasionally infections are transmitted by a<br />

member of the nursery staff who harbors pathogenic<br />

bacteria on the hands or by contamination of vaginal,<br />

rectal bacteria. However, the hand transmission is more<br />

common in infant to infant (Goldmann et al., 1978). The<br />

number of nosocomial gram-negative infections has<br />

increased dramatically in the past decade but pathogenic<br />

E.coli, Klebsiella, Enterobacter and Pseudomonas<br />

organisms have been responsible for majority of the<br />

gram-negative infections 8 . Gram-negative bacilli (GNB)<br />

cause more than 50% of hospital acquired infections.<br />

These organisms are not thought to be part of the<br />

resident flora of the skin perhaps because of factors<br />

such as desiccation inhibition by skin lipids or<br />

interference by normal cutaneous bacterial flora<br />

(Guenthner et al., 1987). Most studies have assumed that<br />

GNB are part of the transient flora only and easily<br />

removed by hygienic hand washing with soap (McGarrity<br />

and Coriell, 1973). GNB remaining after removal of<br />

transient hand flora with soap and water have not been<br />

studied extensively (McGarrity and Coriell, 1973). The<br />

purpose of this study was to determine the prevalence of<br />

GNB as a part of the non transient flora on the hands of<br />

hospital personnel during the period of nosocomial<br />

infections after hand washes and also analyzing the<br />

influence of working a shift in an intensive care unit on<br />

hand carriage of GNB by nurses, the contribution of a<br />

broad range of external influences and mode of delivery<br />

on neonate's skin micro biotic composition in early<br />

infancy.<br />

MATERIALS AND METHODS<br />

Subjects (infants)<br />

Twenty- two newborn infants born at Amin hospital part of Esfahan<br />

University were included in this study. This research was conducted<br />

in 2 stages. The colonization was measured in the beginning and a<br />

few hours after birth. In the pilot study, 12 infants born by vaginal or<br />

cesarean delivery were studied. In final work, 10 infants (2 infants<br />

were premature) at operating room, rooming in and nursery section<br />

were studied.<br />

Sampling and culture of the skin micro flora<br />

In pilot study and final work, 96 and 80 samples were collected<br />

Figure 1. Sampling on blood agar plate.<br />

Heshmati et al. 2235<br />

respectively from forehead region, oral cavity, perineum, and<br />

external ear, auxiliary and umbilical regions. Samples from the skin<br />

flora were taken 3, 4, 5, 8, 14 and 20 h after vaginal or caesarean<br />

delivery by using cotton-tipped swab weltering in sodium phosphate<br />

buffer that were scrubbed on the surface of the skin and<br />

immediately streaked on to the blood agar, Mac conkey, EMB, and<br />

Sc (sabburo cholramphenicol) plates. Samples were brought to the<br />

laboratory and incubated at 37°C within 24 h after collection (Figure<br />

1). Media were incubated in candle jar at 37°C for 48 h. The<br />

bacterial colonies were analyzed in terms of number and<br />

morphological characteristics and then preserved in BHI and 15%<br />

glycerol for determining their species. Upon discharge from the<br />

newborn nursery at the Amin Hospital for maintaining the<br />

microorganisms, brain heart infusion broth (BHI) and 15% glycerol<br />

was used.<br />

Identification of isolates<br />

Bacteria were stained by Gram stain and also 3% H2O2 solution we<br />

used to detect the catalase enzyme then identified by standard<br />

bacteriological and biochemical methods such as acid production<br />

from carbohydrates, using of MSA (Manitol salt agar medium),<br />

DNAse medium, sensivity to Furazolidon disk(100 µg), Bacitracin<br />

disk(0.04U)for differentiation Micrococcus from Staphylococcus, of<br />

medium, SIM, 10% Nacl nutrient agar, MR-VP, TSI, Simmon citrate<br />

agar, urea agar, Nitrate reduction test. Any bacterial colonies which<br />

resembled -hemolytic streptococci were tested for sensivity to<br />

bacitracin with disks containing 0.04U of bacitracin, if the zone of<br />

inhibition was greater than 15 mm, the streptococci would<br />

presumed to be group A and then precise serological grouping was<br />

carried out. For Fungi (Candida sp) we used Sc medium and germ<br />

tube and by using of Corn meal agar medium for production of<br />

clamidospore (Figure 2).<br />

RESULTS<br />

In the first phase 12 neonates were used to obtain<br />

bacteria flora samples at various time of day. A variety of<br />

microorganisms were isolated from the samples.<br />

However, some samples demonstrated no bacterial<br />

growth. We observed that neonates during the birth<br />

would have colonization with a single bacterial species.<br />

Nevertheless.after a period of time several species of<br />

bacteria would be intermixed together. Several gram<br />

negative bacilli strains isolated at various hours after birth<br />

were characterized (Figures 3, 4, 5, 6 and 7).<br />

Oral cavity was found to be sterile in most of times<br />

especially during birth and consequently colonization of


2236 Afr. J. Microbiol. Res.<br />

Figure 2. Isolate sub cultured.<br />

Figure 3. Frequency of isolated organisms from forehead at the birth time.<br />

Figure 4. Frequency of isolated organisms from forehead after birth time.<br />

Staphylococcus epidermidis<br />

Staphylococcus aureus<br />

Corynebacterium xerosis<br />

Streptococcus spp.<br />

E. coli<br />

Enterobacter agglomerans<br />

Staphylococcus epidermidis<br />

Staphylococcus aureus<br />

Staphylococcus saprophyticus<br />

Micrococcus Luteus<br />

E. coli<br />

Corynebacterium xerosis<br />

Corynebacterium renale


Figure 5. Frequency of isolated organisms from perinea area at the birth time.<br />

Micrococcus luteus<br />

Staphylococcus epidermidis<br />

Staphylococcus aureus<br />

Bacillus spp.<br />

E. coli<br />

Enterobacter agglomerans<br />

Plesiomonas shigelloides<br />

E. coli<br />

Enterobacter agglomerans<br />

Staphylococcus epidermidis<br />

Staphylococcus aureus<br />

Micrococcus roseus<br />

Micrococcus luteus<br />

Micrococcus varians<br />

Fungi<br />

Figure 6. Frequency of isolated organisms from perinea area after birth.<br />

Figure 7. Frequency of isolated organisms from oral cavity 6 h after birth.<br />

Staphylococcus epidermidis<br />

Rhodococcus equi<br />

Moraxella catarrhalis<br />

Aurococcus viridans<br />

Staphylococcus aureus<br />

Micrococcus luteus<br />

Streptococuus beta hemolytic<br />

Bacillus lentus<br />

Heshmati et al. 2237


2238 Afr. J. Microbiol. Res.<br />

Figure 8. Percentage of infant with organisms in various anatomic sites. * KEC (Klebsiella, Enterobacter, or Citrobacter).<br />

bacteria was initiated by ingesting food and other<br />

environmental factors (Figure 7).<br />

Although 8 to 15% of cultures yield -hemolytic<br />

streptococci group B which was detected by CAMP and<br />

Hippurate hydrolysis test, group A streptococci are also<br />

encountered. During a two-day period 16 of 22 neonates<br />

in one nursery had organisms sensitive to bacitracin and<br />

penicillin cultured from their umbilical areas. None of the<br />

infant showed clinical signs or symptoms of disease.<br />

These organisms were presumptively identified as group<br />

A streptococci and the positive cultures were first<br />

interpreted as evidence of group A streptococcal<br />

infection. Several weeks later, organisms from 14 of<br />

these infants were identified as a single strain of -<br />

hemolytic Sterptococcus (Figure 8).<br />

DISCUSSION<br />

Various factors may influence the role of microorganisms<br />

in relationship to colonization such as antibacterial agent<br />

(bacteriocin), bacterial interference and competitive<br />

inhibition. Production of bacteriocin by a bacterial species<br />

can inhibit colonization of other bacterial species. It is<br />

also known that a floral bacteria growth can be<br />

inhibited by another bacterial species through<br />

competitive inhibition. For instance pathogenic<br />

microorganisms like Staphylococcus aurous can<br />

proliferate on the neonatal skin in the absence of skin<br />

normal flora such as coagulase-negative Staphylococci.<br />

External factors can also alter the skin normal flora<br />

population:<br />

1) The climate: augmentation in humidity and<br />

temperature can increase the bacterial growth and hence<br />

modifying their population ratio.<br />

2) Anatomical condition, body site: the distribution of skin<br />

normal flora on the body depends on different anatomical<br />

places. The transient bacteria grow extensively on<br />

exposed body parts such as face, neck and hands.<br />

3) Hospitalization, in this study has revealed that long<br />

term hospitalization can increases the bacterial<br />

colonization in neonates.<br />

4) The disease effect: The presence of systemic<br />

disease can also make the body susceptible to<br />

bacterial growth. In this study has revealed diabetic<br />

neonates born from diabetic mother have greater<br />

potential for bacterial growth than the normal neonates.<br />

5) Disinfectant effect: Base on this study organisms such<br />

as Pseudomonas was the most common, and the E.coli<br />

and Entrobacter were isolated repeatedly from washing<br />

liquid. Nonetheless, it was shown that none of these<br />

disinfectant washing liquid would change the coagulase-<br />

negative staphylococcus population, but using disinfecting


soap increased the Propionobacterium population<br />

extensively.<br />

6) Irradiation: this study show that bacterial growth was<br />

susceptible to U.V light radiation and staphylococci were<br />

the most susceptible one among all the studied<br />

organisms.<br />

One of the controversial points in this study was the<br />

level of colonization for group B Streptococci among the<br />

neonates.<br />

According to the study in the Cooper Green Hospital, the<br />

rate of colonization for sterp B was established during 28<br />

months. They emphasize that strep B was the most<br />

common cause of infection in neonate and twines in<br />

which being considered as high risk group (William et al.,<br />

1999). Nevertheless we found variation in the amount of<br />

these bacteria in forehead and perinea areas (14.2 and<br />

3.5%).Therefore it can be concluded that the main cause<br />

of strep B colonization in neonate was due to exposure of<br />

birth canal.<br />

Base on reported studied (Falle and Schleifer, 1981). at<br />

this study ,bacterial growth were increased in still-birth<br />

,low-weighted birth neonates and also in some twins<br />

and single-born babies ,this increase however was more<br />

profound in twins due to complex delivery involved. We<br />

also have found that there is an increasing rate of<br />

colonization of bacteria in immature and low weight<br />

neonate.<br />

Some studies showed a significant difference exist in<br />

colonization of bacteria on skin of neonates during the<br />

first, fourth and tenth weeks. We have demonstrated that<br />

although the rate of colonization of bacteria after birth is<br />

undetectable but colonization of gram- negative<br />

organisms was more prevalent.<br />

Nobel and Jackman reported that colonization of<br />

staphyloccocus in axillaries area can decrease rate of the<br />

Coryneforms of bacteria (William et al., 1999).<br />

But we have showed different result and that the rate of<br />

colonization of these bacteria was similar both during and<br />

after birth.<br />

We also concluded that the highest rate of colonization<br />

for various species of staphyloccocus was in axillary's<br />

areas .and the highest rate of colonization for Coryneform<br />

bacteria was reported in forehead and auxiliary’s areas.<br />

The S. aureus and S. epidermidis were the most common<br />

organisms in forehead area.<br />

The most common organisms of external ear were<br />

reported to be S. epidermidis; we found the same result<br />

which confirmed pervious studies.<br />

Some of earlier researches and literatures have<br />

reported that anaerobic Sterptococcus species are the<br />

first cause of infection. It seems that specific species<br />

constitute the normal flora of vaginal and oral cavity.<br />

While being born the skin of neonates with cesarean<br />

surgery was found sterile during delivery, but neonates<br />

born through birth canal was found to be colonized with<br />

the normal flora.<br />

Heshmati et al. 2239<br />

Based on other results.the Satphylococcus and<br />

Coryneforms were reported as the most common<br />

organisms.found in neonate skin and E.coli and Proteus<br />

species (10%) were also reported. In this study the same<br />

result has obtained (Sarkany and Gaylarde, 1967).<br />

The S. epidermidis was colonized in upper parts of<br />

body and consisted more than 50% the of<br />

Staphylococcus species. We found the same result too.<br />

The S. 0hominis and S. hemolyticus were found in<br />

auxiliary’s and perinea areas. Other findings indicated<br />

that coagulase negative staphyloccocus was colonized in<br />

perinea area. According to other researches, intestine of<br />

neonates were colonized with high rates of klebsiella, E.<br />

ntrobacter or Citobacter species (William et al., 1999).<br />

We found that the most common organisms in neonate<br />

s, stool were gram- negative bacteria such as E.coli and<br />

Entrobacter species.<br />

We also observed that gram -negative bacilli were<br />

mostly colonized in neonates care units. Therefore, we<br />

disregarded studying bacteria colonization in the first<br />

hours of birth in care units.<br />

Larson reported an inverse relation between the bacteria<br />

colonization and continuous hand washing of personals<br />

and nurses of hospitals.<br />

Moreover, hands can be a transmission bacteria portal<br />

between the neonates. Therefore, it is highly<br />

recommended, to use disinfectants daily and hand<br />

washing should be done in neonates care units and ICU<br />

as well (Pass et al., 1980).<br />

Ethical aspects<br />

Informed consent was obtained from the parents of the<br />

babies.<br />

REFERENCES<br />

Guenthner S.H, Owen Hendley J, Wenzel RP (1987). Gram-Negative<br />

Bacilli as No transient Flora on the Hands of Hospital Personnel. J.<br />

Clin. Microbiol., 25: 488-490.<br />

Penders J, Thijis C, Vink C, Stelma FF, Snijders B, Kummeling I, Brandt<br />

PAvan den, Stobberingh EE (2006). Factors Influencing the<br />

Composition of the Micro biota in Early Infancy. Pediatrics, 118: 511-<br />

521.<br />

Falle A, Schleifer KH (1981). Modified Oxidize and Benzidine Tests for<br />

Separation of Staphylococci from Micrococcci. J. Clin. Microbiol., 13:<br />

1031-1035.<br />

Pezzati M, Biagioli EC, Martelli E, Gambi B, Biagiotti R, Rubaltelli FF<br />

(2002). Umbilical Cord Care The Effect of Eight Different cord-Care<br />

regimens on: Cord Separation Time and Other Outcomes. Biol.<br />

Neonate, 81: 38-44.<br />

McConnell TP, Lee CW, Couillard M, Westbrook Sherrill W (2004).<br />

Trends in Umbilical Cord Care: Scientific Evidence for Practice.<br />

Newborn Infant Nurs. Rev., 4: 211-222.<br />

Pass MA, Khare S, Dillon HC (1980).Twin pregnancies: Incidence of<br />

group B streptococcal colonization and disease. Brief clinical and<br />

laboratory observations. J. Pediatr., 4: 635-637.<br />

Goldmann DA, Leclair J, Macone A (1978). Bacterial colonization of<br />

neonates admitted to an intensive care environment. J. Pediatr.,<br />

93(2): 288-293.


2240 Afr. J. Microbiol. Res.<br />

McGarrity GJ, Coriell LL (1973). Bacterial Contamination of Children’s<br />

Soap Bubbles. Am. J. Dis. Child., 125: 224-227.<br />

William EB, Jeffrey BG, Maurice LD (1999). Risk Factors for Early-onset<br />

Group B Streptococcal Sepsis: Estimation of Odds Ratios by Critical<br />

Literature Review. Pediatrics, 103: e77.<br />

Sarkany I, Gaylarde CC (1967). Skin flora of the newborn. Lancet, 1:<br />

589-590.


African Journal of Microbiology Research Vol. 5(16), pp. 2241-2244, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.234<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Evaluation of anticarcinogenicity effect of Artemia<br />

urmiana by Salmonella typhimurium TA 100 strain<br />

Masumeh Abbasi 1 , Saman Mahdavi 2* and Sedigheh Mehrabian 3<br />

1 Islamic Azad university- Malekan Branch, Iran.<br />

2 Islamic Azad university- Maragheh Branch, Iran.<br />

3 Department of Biology, Faculty of science, Teacher Training University, Tehran, Iran.<br />

Accepted 20 May, 2011<br />

Free radicals are produced during oxidative metabolism in body and they can attack all components of<br />

cells and cause cellular changes and early aging. Antioxidant materials role for balance maintenance in<br />

cells has currently attracted more attention. The aim of this study is investing of antimutation effect of<br />

Artemia, cyst and decapsulated egg extract by Ames test and Salmonella typhimurium TA 100 strain.<br />

These extract prevent reverse mutation caused by carcinogenic materials. In comparison with positive<br />

controls (sodium azid, KMno4, cresol) and negative control (distilled water) antimutagenicity properties<br />

of Artemia was evaluated and confirmed. In addition, the value of astaxanthin in those extracts was<br />

assayed. For quality analysis and separation of astaxanthin, we used thin-layer chromatography (TLC)<br />

chromatography and column chromatography, respectively. Then, the presense of astaxanthin in these<br />

extracts was confirmed by spectrometer IR test. The results showed that antimutagenicity properties of<br />

Artemia urmiana are the same with other salty water Artemia. Antimutagenicity and anticarcinogenicity<br />

properties in decapsulated egg, dried Artemia and cyst are 85, 70 and 100%, respectively.<br />

Key words: Ames test, anticarcinogenicity, antioxidant, Artemia urmiana, Salmonella typhimurium TA 100.<br />

INTRODUCTION<br />

Investigation of anticancer features of Artemia urmiana<br />

by using the Salmonella typhimurium TA100 strain has<br />

been done. However, human has discovered the<br />

presence of Artemia in salty rivers long a time ago. The<br />

first report about Artemia was registered by Schlosser in<br />

1755 A.D. He observed Artemia in the samples of the<br />

water that he has provided from salty rivers in a place<br />

near Lymington. He called it Artemia linneaus by the<br />

scientific name of the crab of salt water and in 1819 a<br />

person whose name was Litch recalled it Artemia salina.<br />

At last Artemia in Urmia sea was called Artemia urmiana<br />

by Clark and Bawn (1976), and by the time being, many<br />

*Corresponding author. E-mail: S.mahdavi@iau-maragheh.ac.ir.<br />

Tel: 009809144150454.<br />

Abbreviations: ISA, International study of Artemia; TLC, thinlayer<br />

chromatography; ROS, reactive oxygen species; FT-IR,<br />

fourier transform infrared.<br />

researches and investigations have being done about<br />

different subjects of Artemia by ISA (International Study<br />

of Artemia) in different countries (Lorenz and Cysewski,<br />

2000). Now a day Artemia (that is the famous shrimp of<br />

salty water) is a good source of protein for human, a food<br />

for marines and is used in drugs (Magbool and Suresh,<br />

2003). Because of it contains Caretonoids such as<br />

Astaxantin, Kantanxantin and Autooxicuein, Artemia is<br />

able to do different biological performances such as<br />

protection against light, removing the injuries caused by<br />

oxygen, immune adjusting in body, and antioxidant activities.<br />

Also reactive oxygen species (ROS) may play a<br />

major role as endogenous initiators of degenerative<br />

processes such as deoxyribonucceic acid (DNA) damage<br />

and mutation that may be related to cancer (Nair and<br />

Risch, 2000) cancer (Nair and Risch, 2000). Test of<br />

carcinogens on rats, is expensive and time consuming.<br />

Recently, bacteria have been using for assessment of the<br />

carcinogenicity of suspected materials which are not only<br />

cost- effective but also give prompt and excellent results.


2242 Afr. J. Microbiol. Res.<br />

Table 1. Survey of genotype results of Salmonella typhimurium TA 100 strain.<br />

Strain rfa mutation UVRB mutation R-Factor<br />

Salmonella typhimurium TA100 + + +<br />

One of the methods of assaying carcinogenic effects via<br />

bacteria is Ames test by use of Salmonella strains which<br />

because of mutation have lost the histidine synthesis<br />

potential and in the medium without histidine will face a<br />

reverse mutation which leads to histidine synthesis. By<br />

measuring the developed colonies in the medium, the<br />

carcinogenicity effect will be identified (Motorelmansk<br />

zeger, 2000). This article discusses different steps of<br />

growth of Artemia that has antioxidant and more<br />

anticancer activities so that in proper step we can use it<br />

as foodstuff and drug for human, poultries and marines.<br />

MATERIALS AND METHODS<br />

Artemia was caught from Urmia Lake and transferred to<br />

the laboratory on summer 2005. The dried Artemia, cyst, and<br />

decapsulated egg gathered and froze at -10°C until utilization.<br />

Salmonella typhimurium TA 100 strain was received directly from<br />

Professor Ames (University of Beverly, CA).<br />

Confirming genotypes of tester strains<br />

The tester strains genotype should be confirmed, so fresh overnight<br />

nutrient broth cultures were used for this purpose. Salmonella<br />

typhimurium strain has deficient in dark repair of mutations (UVrB),<br />

and unable to synthesize a portion of the cell wall (rfa).<br />

rfa mutation<br />

TA 100 strain was tested for crystal violet sensitivity. For this test,<br />

nutrient agar plates were seeded with cultures of the motional strain<br />

and sterile filter paper disc soaked in crystal violet was placed on<br />

the surface of each cultured plate. After 12 h incubation the clear<br />

zone of inhibition (approximately 14 mm) was appeared around the<br />

disc indicating the presence of the rfa mutation which permits large<br />

molecules such as crystal violet to enter and kill the bacteria.<br />

UvrB (UVrB) mutation<br />

The UVrB mutation was confirmed by demonstrating UV sensitivity<br />

in TA100 strain which showed this mutation.<br />

R- factor<br />

The R- factor of TA 100 strain was tested for the presence of the<br />

ampicillin resistance factor. It is a convenient marker that makes it<br />

possible to test for the presence of the R- factor plasmid. The<br />

specific regions of the R-factor plasmid PKM 101 DNA that are<br />

essential for enhancement of UV, chemical mutagens, replication<br />

and ampicillin resistance have been identified.<br />

Astaxanthin exploitation<br />

10 g of each dried Artemia, cyst, and decapsulated egg separately<br />

were placed into glass flasks. Petroleum ether, acetone and water<br />

were added at a ratio of 1.5:7.5:1 to the samples and left under<br />

cover for 2 h. Then the solvents were evaporated in rotary<br />

evaporator until sample dryness. The resulting pigments were<br />

redissolved in an adequate volume of diethyl ether. These solutions<br />

were analyzed by thin-layer chromatography (TLC) analysis and<br />

Fourier transform infrared (FT-IR) spectroscopy. In TLC, mixture<br />

25% acetone in n- hexane was used as developers (Kobayashi<br />

and Katsuraji, 2001). In addition, the values of Astaxantin in<br />

those extracts were assayed. To quality analyze and<br />

separate Astaxantin, we used TLC chromatography and<br />

column chromatography respectively. Then we proved this test IR in<br />

these extracts.<br />

Ames test<br />

In antimutagenicity test, the inhibitions of mutagenic activity of<br />

sodium azide and permanganate potassium by the test samples<br />

were determined. 1 ml of solution of the tested compounds and 0.1<br />

ml of an overnight bacterial culture suspension cultivation for 16 h<br />

at 37°C, approximate cell density (2-5)×10 8 cell/ml and 0.1 ml of<br />

solution of the positive mutagens were carefully mixed with 3 ml<br />

of melted top agar containing 50 µmol/l of histidine- biotin, and<br />

poured onto minimal glucose agar plates. Positive and negative<br />

controls were also included in each assay. Sodium azide and<br />

potassium permanganate were used as diagnostic mutagens<br />

(0.1 ml) in the positive control and plates without mutagens and<br />

test samples were considered as negative control. Their revertants<br />

were counted after incubation of the plates at 37°C for 48 h.<br />

Tester strains were checked routinely to confirm genetic features by<br />

using the procedure described by Maron and Ames (Negi et al.,<br />

2003).<br />

RESULTS<br />

Salmonella typhimurium TA 100 strain has special<br />

mutation in its operon histidine that makes it dependent<br />

to outer histidine source. In relation to mutagenic material<br />

this mutation is reversible, so independent bacteria to<br />

histidine will come out. According to Tables 2, 3 and 4,<br />

the antimutation properties of dry Artemia extract,<br />

decapsulated egg and cyst extract in the Presence of<br />

Positive mutations were confirmed. By using of<br />

Salmonella typhimurium TA 100 strain was shown that<br />

the positive control containing the mutating materials like<br />

azid sodium, Permanganat Potasium and cresol causes<br />

reverted mutation in operon histidine (His+). According to<br />

Tables 2, 3 and 4, in presence of anti-mutagenic material<br />

comparing with control sample, the number of reverted<br />

colonies reduced. Indeed, rate of mutation suppression<br />

was evaluated higher than 40% that was indicating<br />

strong antimutagenicity effect of comparison with three


Table 2. The results of investigation of preventing and moderating of mutagenesis effects of Azide sodium by dried<br />

Artemia, cyst and decapsulated egg by using Salmonella typhimurium TA 100.<br />

Examined material<br />

Number and percent of reverted colonies of bacterium<br />

Salmonella typhimurium TA 100<br />

1 2 3<br />

Positive control (Azid sodium) 1600 - 1500 - 1440 -<br />

Negative control 440 - 512 - 524 -<br />

Dried Artemia extract 600 86% 720 78% 800 69%<br />

Decapsulated egg extract 500 94% 724 78% 632 88%<br />

Cyst extract 480 96% 360 100% 400 100%<br />

Abbasi et al. 2243<br />

Table 3. The results of investigation of preventing and moderating of mutagenesis effects of permangeant potasium by dried<br />

Artemia, cyst and decapsulated egg by using Salmonella typhimurium TA 100.<br />

Examined material<br />

Number and percent of reverted colonies of bacterium<br />

Salmonella typhimurium TA 100<br />

1 2 3<br />

Positive control (Permangeant potasium) 1680 - 1760 - 2000 -<br />

Negative control 440 - 512 - 524 -<br />

Dried Artemia extract 724 77% 884 70% 1016 66%<br />

Decapsulated egg extract 600 87% 724 83% 800 81%<br />

Cyst extract 500 87% 480 100% 360 100%<br />

Table 4. The results of investigation of preventing and moderating of mutagenesis effects of cresol by dried<br />

Artemia, cyst and decapsulated egg by using Salmonella typhimurium TA 100.<br />

Examined material<br />

Number and percent of reverted colonies of bacterium<br />

Salmonella typhimurium TA100<br />

1 2 3<br />

Positive control (cresol) 1560 - 2400 - 2800 -<br />

Negative control 664 - 520 - 480 -<br />

Dried Artemia extract 1000 62% 920 78% 840 84%<br />

Decapsulated egg extract 724 93% 648 92% 734 89%<br />

Cyst extract 480 100% 800 85% 360 100%<br />

promotagen materials (Azid sodium, Permanganat<br />

Potasium and cresol). The result coming out of<br />

chromatography of thin layer TLC has showed that the<br />

progressing of the samples produced from the extract of<br />

dried Artemia, cyst and decapsulated egg on the silica<br />

gel paper leads to the formation of a dark orange band<br />

with Rf=40%. This band from color and RF<br />

measurement point of view looks like astaxantin<br />

reference, and the main created stigma was in result<br />

of the separation of the primary samples. Five weak<br />

spots from dried Artemia and two weak spots from cyst<br />

and one weak stigma from decapsulated egg have been<br />

observed (Figure 1).<br />

At first it was assumed that the light colored bands are<br />

related to the ester from astaxantin or even to the rest<br />

of the combinations in it, but after doing spectrographic<br />

with spectrophotometer and comparing them, it was<br />

observed that these spots are like to the spectrums in<br />

result of the primary bands and the light colored bands<br />

are related to astaxantin extract, because astaxantin<br />

can combine with oxygen in the air and change to other<br />

isomers.<br />

Comparison of spectrums related to the main band<br />

of dried Artemia extract, cyst and decapsulated egg<br />

showed that the depth and intensity of created pigments<br />

are almost the same. It showed that the thickness<br />

of pigment in Artemia egg was the most and in dried<br />

Artemia and cyst were fewer than the expected sample.


2244 Afr. J. Microbiol. Res.<br />

Figure 1. Chromatography of thin layer of dried Artemia<br />

extract (A), Decapsulated egg (B) and cyst (C) on<br />

Chromatographic paper of silica gel.<br />

DISCUSSION<br />

One of the main reasons of death in industrialized<br />

societies is cancer. In the last two decades, different<br />

kinds of mutating, cancer making and chemical materials<br />

in foods have been known (Sarmineto and Ochoa, 2002).<br />

Today scientists believe these genetic damages and<br />

changes caused by succession of mutating DNA or<br />

mutation in genes and other genetic changes in chromosomes<br />

structure have role in cancer. According to Ames<br />

theory 80% of mutating material cause cancer (Gerber et<br />

al., 2002) and most of these mutating and cancer<br />

makings show their damaging effects by producing the<br />

free radicals and active oxygen. Free radicals are related<br />

to various physiological and pathological events such as<br />

inflammation, immunization, aging and etc. It has been<br />

suggested that compounds which possess antioxidant<br />

activity, can inhibit mutation and cancer because they<br />

can scavenge free radicals or induce antioxidant<br />

enzymes (Mikula and Ikova, 2003). Thus, daily use of<br />

antioxidants has an important role in protecting body<br />

against the free radicals, even most of the antioxidants<br />

are known as anticancers (Rosenkraz, 2003). Antioxidant<br />

materials such as carotenoids, astaxantin, fixed vitamin<br />

C, autooxicuien, lipids, fish oil and etc has shown<br />

anticancer features of them. TLC<br />

analysis showed that astaxantin and its isomers seemed<br />

to be the dominant carotenoied in Artemia (Duarte and<br />

Lunec, 2005). Epidemiological studies indicate that there<br />

is a close relationship between diet, life style and human<br />

cancer (Nair and Risch, 2000; Mehrabian and<br />

Shirkhodaei, 2006). The result of this research is in<br />

accordant with other researches. According to Ong<br />

formula, antimutagenicity effect is higher than 40%, so<br />

recent examined materials are high antimutagenicity<br />

ability (Ong et al., 1980). Presence of carotenoid pigments<br />

such as β-caroten, kantaxanthin and astaxanthin<br />

have been found in Artemia structure that most of them<br />

are antioxidant properties (Duarte and Lunec, 2005).<br />

Compounds such as glutation in marine food materials<br />

play major role in elimination and limitation of free<br />

radicals (Trusheva, 2006).<br />

REFERENCES<br />

Clark LS, Bowen ST (1976). The genetics of Artemia saliva V<br />

reproductive isolation. J. Helved., 47(6): 385-390.<br />

Duarte T, Lunec J (2005). When an antioxidant is not an antioxidant? A<br />

review of novel actions and reactions of vitamin C and free radicals.<br />

Free Radic. Res., 39(7): 671-686.<br />

Gerber GB, Leonard A, Hantson PH (2002). Carcinogenicity,<br />

mutagenicity and teratogenicity of manganese compounds. Crit. Rev.<br />

Oncol. Hematol., 42: 25-34.<br />

Kobayashi M, Katsuraji T (2001). In larged and astaxathin<br />

accumulating cyst cells of the green algae hematoccocus<br />

pluvialis. J. Biosci. Bioeng., 92: 565-568.<br />

Lorenz RT, Cysewski GR (2000). Commercial potential for<br />

hematococcus microalgae as a natural source of astaxanthin. Trends<br />

Biotechnol., 18.<br />

Magbool TK, Suresh Kumar S (2003). Microbial quality of shrimp<br />

products of export trade produced from aquaculture shrimp. Int. J.<br />

Food Microbiol., 82: 213-221.<br />

Mehrabian S, Shirkhodaei E (2006). Modulation of mutagenicity of<br />

Various mutagens by Shrimp Flesh and Skin Extracts in Salmonella<br />

Test. Pak. J. Biol. Sci., 9(4): 598-600.<br />

Mikula M, Ikova B (2003). Modulation of mutagenicity of various<br />

mutagens by lignin derivatives. Mut. Res. Genet. Tox., 535: 171-180.<br />

Motorelmansk zeger E (2000). The Ames salmonella<br />

microsomemutagenicity assay. Mutares, 455(1-2): 29-60.<br />

Nair B, Risch H (2000). Cancer epidemical biomarker. J. Genet.,<br />

1: 3-28.<br />

Negi PS, Jayaprakasha GK, Jena BS (2003). Antioxidant and<br />

antimutagenic activities of pomegranate peel extracts. Food Chem.,<br />

80: 393-397.<br />

Ong TM, Wolf CR, Zeiger E (1980). Diferential effects of cytochrome<br />

p45 inducers on promutagen activation capabilities and enzymatic<br />

activities of S9 from rat liver. J. Environ. Pathol. Toxicol., 4: 55-65.<br />

Rosenkraz HS (2003). Synergy between toxicity and genotoxicity<br />

relevance to human cancer risk. Mut. Res., 529: 117-127.<br />

Sarmineto M, Ochoa M (2002). Mutagenicity and antimutagenicity<br />

studies of lipidic extracts from yellow tail fish, lise fish and Cason fish.<br />

Food Chem. Toxicol., 40: 1469-1474.<br />

Trusheva B (2006). Bioactive constituents of Brazilian red proplis.<br />

Evidence-based complementary and Alternative Medicine. Ecam.<br />

Oxford University press, 3(2): 249-254.


African Journal of Microbiology Research Vol. 5(16), pp. 2245-2248, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.549<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

mRNA expression of iron metabolism relation genes in<br />

macrophages by infection with Salmonella typhimurium<br />

Pan Xin<br />

Department of Microbiology, Second Military Medical University, Shanghai 200433, China. E-mail: xinpanpx@163.com.<br />

Tel: 86-21-81870998 or 13524211027. Fax: 86-21-81870998.<br />

Accepted 30 July, 2011<br />

A total number of 13 iron metabolism relation genes expression profiles of RAW264.7 murine<br />

macrophages infected or uninfected with Salmonella typhimurium were tested by real-time polymerase<br />

chain reaction (RT-PCR) to evaluate the metabolism of iron in host-pathogen interplay. The living wildtype<br />

S. typhimurium induces expression of the transferrin receptor (Tfr1) in host cell macrophages,<br />

which results in a sustained increase of the labile iron pool inside the host cell after 1 or 24 h infection.<br />

Gene expression analysis showed that wild-type S. typhimurium drives an active iron acquisition<br />

program with induction of ferrireductase (Steap3), iron membrane transporter Dmt1, and iron regulatory<br />

proteins (Irp1 and Irp2), while not too much iron efflux changing through ferriportin (Fpn1). The spiA -<br />

Salmonella mutant strain used in our studies also caused an increase in Tfr1 at 1 or 24 h, but leading to<br />

decrease in Fpn1 at 24 h as compared with 1 h. The assessment of the labile iron pool after infection<br />

with spiA - Salmonella after 24 h shows an increase. The same of these two phenotypes allowed iron<br />

overload in macrophage and became one of the reasons for Salmonella survival inside the macrophage.<br />

Key words: Macrophage, Salmonella, iron metabolism.<br />

INTRODUCTION<br />

Iron is a critical determinant in host-pathogen interaction.<br />

It is required for full virulence expression by a wide<br />

variety of intracellular bacteria. Deprivation of iron in-vivo<br />

and in-vitro severely reduces the pathogenicity of<br />

Mycobacterium tuberculosis, Coxiella burnettii, Legionella<br />

pneumophila, and S. typhimurium (Braun, 2001). In fact,<br />

attempts to withhold iron by sequestering free iron during<br />

infection is a major defense strategy used by many<br />

species (Bullen et al., 2006). On the other hand, hostcells<br />

require iron for first-line defense mechanisms. Iron<br />

is needed for Haber-Weiss redox chemistry to produce<br />

hydrogen peroxide or hydroxy radicals (Eaton and Qian,<br />

2002). Iron and proteins involved in iron metabolism have<br />

interesting links to the immune system (Hentze et al.,<br />

2004).<br />

How the bacterium tricks the host cell into providing<br />

iron and how the host, at the same time, keeps a delicate<br />

balance between having enough iron for redox reactions<br />

and withholding iron from intracellular bacteria, however,<br />

is not well understood. Here we investigated the<br />

expression profile of macrophage iron homeostasis<br />

following the infection of RAW264.7 murine macrophages<br />

with S. typhimurium.<br />

MATERIALS AND METHODS<br />

Bacterial strains and cell<br />

Wild-type S. typhimurium strain ATCC 14028 (ST) and S.<br />

typhimurium mutant strains spiA::kan (EG5793) (STA) were used.<br />

RAW264.7 murine macrophage-like cell line was obtained from<br />

ATCC.<br />

Primers (synthesized by Bioasia Company)<br />

The primers in Table 1 were used in this study.<br />

Infection assay<br />

Several colonies of Salmonella were collected, washed twice with<br />

cell culture complete medium (DMEM was supplemented with 10%<br />

fetal bovine serum), and resuspended with the same medium.<br />

Multiplicity of infection was adjusted to 50 using a standardized<br />

calibration curve of OD600 / colony-forming units (cfu). Bacteria<br />

were added to RAW264.7 cells in 100×15 mm cell culture dishes.<br />

Infections were initiated by centrifuging the bacteria onto the cell<br />

monolayers at 170×g for 5 min and then incubated for 1h at 37°C<br />

5% CO2. In the second hour, gentamicin was added to a final<br />

concentration o f 100 g/mL, after which, it was lowered to 10


2246 Afr. J. Microbiol. Res.<br />

Table 1. Primers used for real-time quantitative PCR assay.<br />

Gene name Accession number Forward primer (5’ 3’) Reverse Primer (5’ 3’)<br />

Irp1 NM_007386 ACTTTGAAAGCTGCCTTGG CTCCACTTCCAGGAGACAGG<br />

Irp2 NM_022655 TGAAGAAACGGACCTGCTCT GCTCACATCCAACCACCTCT<br />

Tfr1 BC054522 TGCAGAAAAGGTTGCAAATG TGAGCATGTCCAAAGAGTGC<br />

Dmt1 NM_008732 GCCAGCCAGTAAGTTCAAGG GCTGTCCAGGAAGACCTGAG<br />

LcnR BC062878 GCAAGGCTACCCCATACAAA TCTTTGGGCATAGGATGGAG<br />

Lcn2 NM_008491 CTGAATGGGTGGTGAGTGTG TATTCAGCAGAAAGGGGACG<br />

Steap3 BC037435 CTCTCTGTGCAGTCTCGCTG TGCAGAGATGACGTTGAAGG<br />

Hmox1 NM_010442 CCTCACTGGCAGGAAATCAT CCAGAGTGTTCATTCGAGCA<br />

Fpn1 AF226613 TGCCTTAGTTGTCCTTTGGG GTGGAGAGAGAGTGGCCAAG<br />

Hamp1 NM_032541 GAGAGACACCAACTTCCCCA TCAGGATGTGGCTCTAGGCT<br />

Ftl1 NM_010240 AAGATGGGCAACCATCTGAC GCCTCCTAGTCGTGCTTGAG<br />

Ftl2 NM_008049 TGACTTCCTGGAAAGCCACT GCCTCCTAGTCGTGCTTGAG<br />

Fth1 NM_010239 CTCATGAGGAGAGGGAGCAT GTGCACACTCCATTGCATTC<br />

GAPDH NM_008084 CCCACTAACATCAAATGGGG CCTTCCACAATGCCAAAGTT<br />

g/mL. At different time intervals, the RAW264.7 cells were washed<br />

three times with DMEM, and harvested for RNA analysis.<br />

Uninfected cells were used as control.<br />

RNA analysis<br />

Total RNA was isolated 1 h later and DNased using the Microto-<br />

Midi Total RNA Purification System (Invitrogen) according to the<br />

product instructions. RNA concentrations were determined by a<br />

RiboGreen assay (Molecular Probes). The reverse transcription<br />

reactions were carried out with 20 units of MMuLV reverse<br />

transcriptase (Fisher Scientific); 20 units RNase inhibitor (Fisher<br />

Scientific); RT-PCR buffer containing 10 mM Tris-HCl and 50 mM<br />

KCl, 2.5 mM MgCl2, 10 mM dithiothreitol, and 1 mM of each dNTP.<br />

The concentration of each reverse primer was 5 M. 100 ng of total<br />

RNA from each sample was reverse transcribed using reverse<br />

primers. The reverse transcription reactions were incubated for 1<br />

min at 48°C, 5 min at 37°C, 60 min at 42°C, and then 5 min at<br />

95°C.<br />

Real-time RT-PCR was based on the high affinity, doublestranded<br />

DNA binding dye SYBR Green using a Bio-Rad IQ SYBR<br />

Green Supermix according to manufacturer's instructions. A total of<br />

2 L of cDNA was used in the qPCR reactions (1×SYBR green<br />

PCR master mix, 500 nM gene specific forward and reverse<br />

primers). All qPCR reactions started with 2 min at 95°C followed by<br />

40 cycles of 15 s at 94°C and 20 s at 55°C and 30 s at 72°C in an<br />

Applied Biosystems 7900HT Fast Real-Time PCR System, utilizing<br />

SDS 2.1 software. All reactions were run three times. Differences in<br />

mRNA concentrations were quantified by the cycles to fluorescence<br />

–[ Ct experimental gene- Ct housekeeping<br />

midpoint cycle threshold calculation (2<br />

gene]<br />

), using GAPDH as the housekeeping gene.<br />

Statistical analysis<br />

Data were expressed as mean ± SD. An analysis of variance was<br />

performed with Statview 4.0 statistical analysis software, and P <<br />

0.05 was considered statistically significant.<br />

RESULTS<br />

The realtime PCR results are listed in Table 2.<br />

DISCUSSION<br />

We sought to characterize the metabolic changes of<br />

RAW 264.7 macrophage iron homeostasis following the<br />

infection with S. typhimurium. The SPI-2 encoded SpiA<br />

protein is a component of the outer membrane ring of the<br />

type three secretion systems (TTSS) needle complex<br />

(Tobar et al., 2006). A spiA - mutant strain rendered<br />

bacteria unable to translocate effector molecules into<br />

macrophage cytoplasm (Ochman et al., 1996) and<br />

displayed levels of phagosome-lysosome fusion that<br />

were much higher than those displayed by wild-type<br />

Salmonella and similar to those exhibited by heat-killed<br />

Salmonella. So we used STA as non-virulent S.<br />

typhimurium strain to compare with wild-type strain in<br />

studying iron metabolism. Iron is delivered to<br />

macrophages primarily through the transferrin receptor 1<br />

(Tfr1). Tfr1 mRNA levels were significantly increased<br />

following 1 h both of ST and STA infection by 2.20-fold<br />

(F=2036.27, P=0.0001) and 1.72-fold (F=8.64, P=0.0424)<br />

as compared with non-infected RAW264.7 cells.<br />

However, after 24 h of infection, the mRNA level for Tfr1<br />

in ST group decreased 0.7-fold (F=11.94, P=0.0259) as<br />

compared with infected 1 h, but increased 1.88-fold<br />

(F=14.11, P=0.0198) during infection with STA.<br />

We also investigated the expression of other genes that<br />

regulate host cell iron levels by Real time PCR. There are<br />

two main eukaryotic iron-regulatory proteins, Irp1 and<br />

Irp2, which sense changes in the labile iron pool (Galy et<br />

al., 2008). They both act post transcriptionally by<br />

stabilizing their respective target mRNA and by affecting<br />

initiation of translation. When investigating Irp1 mRNA<br />

levels in RAW264.7 phagocytes, we found significant<br />

difference between controls and 1 h of two Salmonella<br />

strains infected cells (2.37-fold for ST, F=27.44,<br />

P=0.0063; 2.51-fold for STA, F=45.85, P=0.0025); when<br />

we compared 1 h with 24 h infected samples, there was


Table 2. mRNA expression of iron metabolism-related genes in RAW264.7 cells after infection with viable Salmonella.<br />

Gene name Uninfected<br />

ST infection sample<br />

STA infection sample<br />

Infection 1 h Infection 24 h Infection 1 h Infection 24 h<br />

Irp1 1 2.37±0.45 2.15±0.68 2.51±0.39 0.68±0.06<br />

Irp2 1 2.77±0.32 3.73±1.01 2.52±0.78 2.76±0.32<br />

Tfr1 1 2.20±0.04 1.83±0.10 1.72±0.43 4.15±0.35<br />

Dmt1 1 12.36±2.24 3.22±0.17 5.36±0.35 12.26±3.41<br />

LcnR 1 3.06±0.41 0.66±0.12 2.23±0.36 0.24±0.05<br />

Lcn2 1 2.07±0.38 17.44±1.41 1.18±0.15 11.63±0.54<br />

Steap3 1 2.86±0.18 1.87±0.43 5.20±0.68 3.78±1.19<br />

Hmox1 1 1.33±0.12 6.81±0.39 2.87±0.31 6.34±0.58<br />

Fpn1 1 1.04±0.12 0.62±0.09 1.42±0.13 0.57±0.12<br />

Hamp1 1 3.88±0.45 11.85±1.73 2.64±0.33 1.42±0.25<br />

Ftl1 1 0.81±0.05 1.17±0.09 1.16±0.04 1.46±0.07<br />

Ftl2 1 0.88±0.03 1.23±0.07 1.18±0.01 1.49±0.04<br />

Fth1 1 0.77±0.07 1.58±0.17 1.34±0.10 1.66±0.13<br />

Xin 2247<br />

Mean values shown were normalized to GAPDH mRNA levels and compared to correlated values found in uninfected macrophages (=1).<br />

no significant difference in ST-infected cells (1.11-fold,<br />

F=0.60, P=0.4831), but Irp1 expression decreased (0.27fold,<br />

F=3996.75, P=0.0001) in 24 h STA -infected cells.<br />

Irp2 mRNA levels was also induced (2.77-fold, F=92.96,<br />

P=0.0006) in 1 h ST-infected cells as compared with noninfected<br />

RAW264.7 cells, and no substantially alter in 24<br />

h ST-infected cells (1.11-fold, F=0.60, P=0.4831) as<br />

compared with 1 h infection. In STA-infected samples,<br />

Irp2 mRNA levels at 1 h increased 2.52-fold (F=11.37,<br />

P=0.0280) as compared with controls, while the mRNA<br />

levels also no substantially alter (0.90-fold, F=0.40,<br />

P=0.5599) after 24 h of infection as compared with 1 h<br />

infection.<br />

After uptake of iron and vesicle acidification, iron is<br />

reduced to its ferrous form by the ferrireductase sixtransmembrane<br />

epithelial antigen of the prostate 3<br />

(Steap3) prior to being transferred across the endosomal<br />

membrane (Ohgami et al., 2005). After 1 h of infection<br />

with wild-type Salmonella, mRNA levels for Steap3 were<br />

increased 2.86-fold (F=313.07, P=0.0001) as compared<br />

with controls. After 24 h infection with ST, Steap3<br />

moderately decreased 0.66-fold (F=10.41, P=0.0321) as<br />

compared with 1 h infection. In STA infection sample,<br />

mRNA levels for Steap3 at 1 h were increased 5.20-fold<br />

(F=115.98, P=0.0004) as compared with controls, but<br />

declined at 24 h infection period (0.67-fold, F=10.85,<br />

P=0.0301) as compared with 1 h infection.<br />

After reduction, ferrous iron is transported into the<br />

cytosol by Divalent metal transporter 1(Dmt1) (Andrews<br />

and Schmidt, 2007). After 1 h of infection with wild-type<br />

Salmonella, mRNA levels for Dmt1 is increased 12.36fold<br />

(F=64.49, P=0.0013) as compared with controls.<br />

After 24 h infection with ST, Dmt1 decreased 0.27-fold<br />

(F=793.44, P=0.0001) as compared with 1 h infection. In<br />

STA infection sample, mRNA levels for Dmt1 at 1 h<br />

(5.36-fold, F=465.08, P=0.0001) were significantly<br />

different from control. This increase continued over a 24<br />

h infection period (2.11-fold, F=14.66, P=0.0186) as<br />

compared with 1 h infection.<br />

The lipocalin system provides the host with another<br />

way of scavenging iron. At the same time, lipocalin2<br />

(Lcn2) can interact with bacterial siderophores to inhibit<br />

bacterial growth and has now been recognized as an<br />

important arm of the innate immune response (Flo et al.,<br />

2004). The secreted gene product Lcn2 is induced 2.07fold<br />

(F=22.88, P=0.0088) when cells are infected with ST<br />

at 1 h as compared with controls. This increase continued<br />

over a 24 h infection period (8.6-fold, F=70.14, P=0.0011)<br />

as compared with 1 h infection. In STA infection sample,<br />

mRNA levels for Lcn2 at 1 h did not alter (1.18-fold,<br />

F=4.00, P=0.1161) as compared with control, but<br />

increased (11.63-fold, F=1183.82, P=0.0001) at 24 h<br />

infection as compared with 1 h infection. Wild type<br />

Salmonella also raised LcnR mRNA levels 3.06-fold<br />

(F=78.44, P=0.0009) at 1 h as compared with control, but<br />

decreased (0.21-fold, F=7956.57, P=0.0001) at 24 h as<br />

compared with 1 h infection. In STA infection sample,<br />

mRNA levels for LcnR at 1 h (2.23-fold, F=19.26,<br />

P=0.0118) were increased as compared with control, but<br />

also decreased (0.07-fold, F=76729.00, P=0.0001) at 24<br />

h infection period as compared with 1 h infection.<br />

Infected host cells can restrict the intracellular iron pool<br />

available for intracellular parasites by transporting iron<br />

out of the cells via ferroportin 1 (Fpn1), a transmembrane<br />

iron efflux protein (Knutson and Wessling-Resnick, 2003).<br />

Fpn1 is negatively regulated by hepcidin antimicrobial<br />

peptide (Hamp1), thus effectively reducing iron efflux<br />

(Nguyen et al., 2006). Hepcidin is significantly<br />

upregulated 3.88-fold (F=118.54, P=0.0004) in infections<br />

with ST at 1 h with accompanying unchanged in Fpn1


2248 Afr. J. Microbiol. Res.<br />

(1.04-fold, F=0.32, P=0.6030) as compared with control,<br />

and Hamp1 increase continued (3.1-fold, F=32.90,<br />

P=0.0046) at 24 h infection with decreased in Fpn1 (0.60fold,<br />

F=192.64, P=0.0002) as compared with 1 h<br />

infection. Hamp1 is increased (2.64-fold, F=73.42,<br />

P=0.0010) in infections with STA at 1 h with<br />

accompanying increased in Fpn1 (1.42-fold, F=30.77,<br />

P=0.0052) as compared with control, and Hamp1<br />

moderately decreased (0.51-fold, F=41.16, P=0.0030) at<br />

24 h infection with decreased in Fpn1 (0.33-fold,<br />

F=498.78, P=0.0001) as compared with 1h infection.<br />

This suggests a stronger effect for iron retention in the<br />

host cell during infection with ST. Heme oxygenase<br />

(Hmox1) catalyzes the conversion of heme to biliverdin,<br />

iron, and carbon monoxide. In macrophages it has an<br />

important antioxidative protective function, presumably by<br />

reducing pro-oxidant or proapoptotic hemoproteins<br />

(Orozco et al., 2007). The mRNA level for Hmox1 is<br />

moderately increased 1.3-fold (F=23.42, P=0.0084) in<br />

macrophages infected by ST at 1 h. This increase<br />

continued over a 24 h infection period (5.13-fold,<br />

F=886.43, P=0.0001) as compared with 1 h ST infection.<br />

In STA infection sample, mRNA levels for Hmox1 at 1h<br />

were increased (2.87-fold, F=109.54, P=0.0005) as<br />

compared with control, and the increase continued (2.21fold,<br />

F=1324.96, P=0.0001) at 24 h infection as<br />

compared with 1 h infection.<br />

The components of the ferritin iron storage system,<br />

ferritin heavy chain 1(Fth1), ferritin light chain 1 (Ftl1) and<br />

ferritin light chain 2 (Ftl2), are moderately decrease by<br />

infection with ST at 1 h (0.77-fold, F=32.39, P=0.0047;<br />

0.81-fold, F=51.57, P=0.0020; 0.88-fold, F=61.71,<br />

P=0.0014, respectively) as compared with control, and<br />

increased over a 24 h infection period (2.05-fold,<br />

F=2037.88, P=0.0001; 1.45-fold, F=867.86, P=0.0001;<br />

1.40-fold, F=369.23, P=0.0001, respectively) as<br />

compared with 1h ST infection. In STA infection sample,<br />

these three mRNA levels at 1 h were increased (1.34fold,<br />

F=31.65, P=0.0049; 1.16-fold, F=45.63, P=0.0025;<br />

1.18-fold, F=972.00, P=0.0001) as compared with<br />

control, and still increased (1.24-fold, F=576.00,<br />

P=0.0001; 1.24-fold, F=782.29, P=0.0001; 1.27-fold,<br />

F=182.25, P=0.0002) at 24 h infection as compared with<br />

1 h infection.<br />

Thus, wild-type S. typhimurium appears to drive an<br />

active transferrin-mediated iron uptake program after<br />

infection 1 or 24 h. This was supported by increased<br />

mRNA levels for Irp1 and Irp2, induction of genes<br />

required for transfer of iron to the cytosol via Dmt1 and<br />

Steap3, and by not too much iron efflux changing through<br />

Fpn1 and Hamp1. The spiA - Salmonella mutant strain<br />

used in our studies caused an increase in Tfr1 at 1 or 24<br />

h, and led to decrease in Fpn1 and Hamp1 at 24 h as<br />

compared with 1 h. The assessment was that the labile<br />

iron pool increased after infection with spiA - Salmonella<br />

for 24 h. Of interest in this context is that the spiA -<br />

Salmonella mutant strain used in our studies behaves<br />

quite different to the spiC - strain by lacking an increase in<br />

Tfr1, but leading to an increase in Fpn1 (Pan et al.,<br />

2010). Macrophage iron overload inhibits the transcription<br />

of iNOS and the generation of NO (Weiss et al., 1994)<br />

and became one of the reasons for Salmonella survival<br />

inside the macrophage.<br />

ACKNOWLEDGMENT<br />

This work was supported by the National Natural Science<br />

Foundation of China (Contract No. 30972633).<br />

REFERENCES<br />

Andrews NC, Schmidt PJ (2007). Iron homeostasis. Annu. Rev.<br />

Physiol., 69: 69-85.<br />

Braun V (2001). Iron uptake mechanisms and their regulation in<br />

pathogenic bacteria. Int. J. Med. Microbiol., 291(2): 67-79.<br />

Bullen JJ, Rogers HJ, Spalding PB, Ward CG (2006). Natural<br />

resistance, iron and infection: a challenge for clinical medicine. J.<br />

Med. Microbiol., 55(Pt 3): 251-258.<br />

Eaton JW, Qian M (2002). Molecular bases of cellular iron toxicity. Free.<br />

Radic. Biol. Med., 32(9): 833-840.<br />

Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes MA, Strong RK, Akira<br />

S, Aderem A (2004). Lipocalin 2 mediates an innate immune<br />

response to bacterial infection by sequestrating iron. Nature,<br />

432(7019): 917-921.<br />

Galy B, Ferring-Appel D, Kaden S, Gröne HJ, Hentze MW (2008). Iron<br />

regulatory proteins are essential for intestinal function and control key<br />

iron absorption molecules in the duodenum. Cell. Metab., 7(1): 79-85.<br />

Hentze MW, Muckenthaler MU, Andrews NC (2004). Balancing acts:<br />

molecular control of mammalian iron metabolism. Cell, 117(3): 285-<br />

297.<br />

Knutson M, Wessling-Resnick M (2003). Iron metabolism in the<br />

reticuloendothelial system. Crit. Rev. Biochem. Mol. Biol., 38(1): 61-<br />

88.<br />

Nguyen NB, Callaghan KD, Ghio AJ, Haile DJ, Yang F (2006). Hepcidin<br />

expression and iron transport in alveolar macrophages. Am. J.<br />

Physiol. Lung. Cell. Mol. Physiol., 291(3): L417- 425.<br />

Ochman H, Soncini FC, Solomon F, Groisman EA (1996). Identification<br />

of a pathogenicity island required for Salmonella survival in host cells.<br />

Proc. Natl. Acad. Sci. USA., 93(15): 7800–7804.<br />

Ohgami RS, Campagna DR, Greer EL, Antiochos B, McDonald A, Chen<br />

J, Sharp JJ, Fujiwara Y, Barker JE, Fleming MD (2005). Identification<br />

of a ferrireductase required for efficient transferrin-dependent iron<br />

uptake in erythroid cells. Nat. Genet., 37(11): 1264-1269.<br />

Orozco LD, Kapturczak MH, Barajas B, Wang X, Weinstein MM, Wong<br />

J, Deshane J, Bolisetty S, Shaposhnik Z, Shih DM, Agarwal A, Lusis<br />

AJ, Araujo JA (2007). Heme oxygenase-1 expression in<br />

macrophages plays a beneficial role in atherosclerosis. Circ. Res.,<br />

100(12): 1703-1711.<br />

Pan X, Tamilselvam B, Hansen EJ, Daefler S (2010). Modulation of iron<br />

homeostasis in macrophages by bacterial intracellular pathogens.<br />

BMC. Microbiol., 10: 64.<br />

Tobar JA, Carreño LJ, Bueno SM, González PA, Mora JE, Quezada<br />

SA, Kalergis AM (2006). Virulent Salmonella enterica serovar<br />

typhimurium evades adaptive immunity by preventing dendritic cells<br />

from activating T cells. Infect. Immun., 74(11): 6438-6448.<br />

Weiss G, Werner-Felmayer G, Werner ER, Grünewald K, Wachter H,<br />

Hentze MW (1994). Iron regulates nitric oxide synthase activity by<br />

controlling nuclear transcription. J. Exp. Med., 180(3): 969-976.


African Journal of Microbiology Research Vol. 5(16), pp. 2249-2253, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.570<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Expanding drug resistance through integron<br />

acquisition in Salmonella spp. isolates obtained in<br />

Iran<br />

Bahareh Rajaei 1 , Seyed Davar Siadat 1,2* , Mohamad Reza Razavi 3 ,<br />

Mohammad Reza Aghasadeghi 2 , Nahid Sepehri Rad 1 , Farzad Badmasti 4 ,<br />

Somieh Khanjani Jafroodi 3 , Taraneh Rajaei 5 , Arfa Moshiri 6 and Saifuddin Javadian 7<br />

1 Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.<br />

2 Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.<br />

3 Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran.<br />

4 Department of Microbiology, Pasteur Institute of Iran, Tehran, Iran.<br />

5 Department of Immunology and Allergy, Mashhad University of Medical Sciences, Mashhad, Iran.<br />

6 Department of Biotechnology, Faculty of Allied Sciences, Tehran University of Medical Sciences, Tehran, Iran.<br />

7 Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran.<br />

Accepted 15 July, 2011<br />

A total eighty four epidemiologically unrelated clinical isolates of Salmonella enterica serovars were<br />

subjected to antimicrobial susceptibility testing and molecular detection of class 1 and 2 integrons.<br />

Eleven isolates (13.1%) which were resistant to at least 4 groups of antimicrobial agents considered<br />

as MDR (multidrug resistant) Salmonella serovars. PCR assays detected intI1 and intI2 genes in 50<br />

(59.5%) and 14 (16.7%) of Salmonella clinical isolates respectively. Emergence of MDR Salmonella<br />

serovars demonstrates that antimicrobial selection pressure is widespread and increased distribution<br />

of integron carrying gene cassettes which confer resistance to different antibiotics confirms that<br />

integron-mediated antibiotic resistance is considerable in our clinical settings.<br />

Key words: Salmonella spp., integron, multidrug resistance (MDR).<br />

INTRODUCTION<br />

Salmonella is involved in a wide variety of infections<br />

ranging from life-threatening typhoid to gastroenteritis<br />

and bacteremia (Boyd and Hartl, 1998). Antibiotic<br />

resistance in Salmonella is an emerging problem during<br />

the last decades. The intensive use of antibiotics in both<br />

human and veterinary medicine, as well as in agriculture,<br />

has caused bacteria to develop resistance mechanisms<br />

against therapeutic drugs (Stokes and Hall, 1989;<br />

Rodriguez et al., 2008). Dissemination of antibiotic<br />

resistance genes by horizontal gene transfer has led to<br />

the rapid emergence of antibiotic resistance among<br />

bacteria, thus complicating the treatment of infections<br />

(Gu et al., 2008). The worldwide emergence of multidrugresistant<br />

(MDR) phenotypes among Salmonella<br />

*Corresponding author. E-mail: d.siadat@gmail.com. Tel:<br />

00982144865100. Fax: 00982144865105.<br />

serotypes is of increasing concern for both the scientific<br />

community and the general public (Havlickova et al.,<br />

2009; Yang et al., 2009).<br />

Acquired resistance evolves via horizontal transfer of<br />

antimicrobial resistance genes located on various types<br />

of mobile DNA elements (Yang et al., 2009). A key genetic<br />

system involved in spreading antibiotic multiresistance<br />

is the integron, a genetic element that, although normally<br />

immobile itself, can be transferred in companion with<br />

mobile genetic elements (Tamang et al., 2007)<br />

Integrons were first identified by virtue of their<br />

important role in the acquisition and expression of genes<br />

conferring antibiotic resistance. Integrons are composed<br />

of three key elements necessary for the capture of<br />

exogenous genes: a gene (intI) encoding an integrase<br />

belonging to the tyrosine-recombinase family; a primary<br />

recombination site (attI) located immediately adjacent to<br />

intI; and a strong promoter (Pc) that directs transcription<br />

of the captured genes (Carattoli, 2001; Mazel, 2006).


2250 Afr. J. Microbiol. Res.<br />

Table 1. Information about number of MDR and resistant isolates on the basis of the MIC determination according to guidelines of CLSI<br />

(2009).<br />

Integron positive isolates<br />

No. of resistant isolates to the following antibiotics a on the basis of MIC<br />

SMX SXT TMP CIP NAL AMP CAZ CHL STR<br />

intI1 positive isolates(50) 20 45 22 1 32 5 2 21 20 11<br />

intI2 positive isolates(14) 10 12 11 0 11 0 1 4 6 1<br />

a Abbreviation of mentioned antibiotics and breakpoint for resistance (µg/ml) : AMP, ampicillin ( 32); CAZ, ceftazidime ( 16); CHL, chloramphenicol<br />

( 32); CIP, ciprofloxacin ( 4); NAL, nalidexic acid ( 32); STR, streptomycin ( 64); TMP, trimethoprim ( 4); SMX, sulfamethoxazole ( 512); SXT,<br />

sulfamethoxazole-trimethoprim ( 4/76). Breakpoints were adopted from CLSI (Clinical and Laboratory Standards Institute), except for streptomycin,<br />

which has no CLSI breakpoint.<br />

b In this study isolates which were resistant to at least 4 groups of antimicrobial agents considered as MDR Salmonella isolates.<br />

On the basis of nucleotide sequence of the integrase<br />

gene (intI), nine classes of integrons have been identified<br />

(Moura et al., 2007; Macedo-Vinas et al., 2009) but, to<br />

date, only those of class 1 and 2 have been reported in<br />

S. enterica (Rodriguez et al., 2008).<br />

The objective of this study was to evaluate the<br />

antimicrobial resistance profile and to investigate the<br />

contribution of class 1 and 2 integrons in multidrug<br />

resistant Salmonella isolates.<br />

MATERIALS AND METHODS<br />

Isolation and identification of bacteria<br />

A total of eighty four S. enterica isolates attributed to Typhi (n=40),<br />

Paratyphi A (n=2), Typhymurium (n=12), and non Typhi (n=30)<br />

serovars were studied. The clinical isolates were recovered from<br />

stool (n= 69), blood (n= 6), bone marrow (n= 3), synovial fluid (n=<br />

3), ascites (n= 1), abces (n= 1), urine (n= 1). All isolates were<br />

identified by standard microbiological techniques as previously<br />

described (Martin et al., 2008; Ahmed et al., 2009). The serogroup<br />

was checked with O antisera by the slide agglutination method<br />

(Difco Laboratories, Detroit, MI).<br />

Antimicrobial susceptibility test<br />

The antimicrobial susceptibility test was performed using the<br />

standard disk diffusion method on Muller-Hinton agar and the<br />

Minimum Inhibitory Concentrations (MICs) were determined by<br />

broth microdilution method according to the guidelines of the<br />

Clinical and Laboratory Standards Institute (CLSI, 2009). Disks<br />

prepared by MAST company (Mast Co, Merseyside, UK) were<br />

used to determine the susceptibility of isolates to ampicillin (10 µg),<br />

tetracycline (30 µg), chloramphenicol (30 µg), trimethoprim (5 µg),<br />

sulfamethoxazole-trimethoprim (30 µg), streptomycine (10 µg),<br />

nalidexic acid (30 µg), ciprofloxacin (5 µg), ofloxacin (5 µg),<br />

levofloxacin (5 µg), norfloxacin (5 µg), gatifloxacin (5 µg),<br />

moxifloxacin (5 µg), cefotaxime (30 µg), cefixime (5 µg), ceftriaxone<br />

(30 µg), cefepime (30 µg), ceftazidime (30 µg), amikacin (30 µg),<br />

azithromycin (15 µg), spectinomycin (100 µg), gentamicin (10 µg),<br />

colistin-sulfate (10 µg), imipenem (10 µg). The MICs of ampicillin,<br />

chloramphenicol, streptomycin, nalidixic acid, ciprofloxacin,<br />

ceftazidime, trimethoprim, sulfamethoxazole and sulfamethoxazoletrimethoprim<br />

were carried out against all clinical isolates. The<br />

breakpoints used were those defined by the CLSI for<br />

Enterobacteriaceae, with the exception of streptomycin (32_g/mL)<br />

(CLSI, 2009) (Table 1). E. coli ATCC 25922 was used as a quality<br />

control organism in antimicrobial susceptibility test.<br />

Detection of class 1 and 2 integrons<br />

MDR b<br />

Chromosome and plasmid DNA templates were prepared by<br />

phenol-chloroform methods as previously described (Sambrook et<br />

al., 1989). All isolates were screened for detection of class 1 and 2<br />

integrons by the primers described by Goldstein et al. (2001)<br />

designed for the intI1 and intI2 genes respectively. The<br />

amplification program was performed by termocycler (Eppendorf<br />

Mastercycler®, MA) and started with initial denaturation of 4 min at<br />

94°C and programmed with 35 cycles of each: 1min at 94°C, 30 s<br />

at 60°C, 1 min at 72°C. The program finished with the final<br />

extension of 10min at 72°C.<br />

RESULTS<br />

Disk diffusion and minimum inhibitory concentration<br />

(MIC)<br />

A total of eighty four Salmonella isolates including S.<br />

Typhi (n = 40), S.Typhymurium (n=12), S. Paratyphi A (n<br />

= 2), non-Typhi (n=30) serovars were studied. According<br />

to the Disk diffusion and minimum inhibitory concentration<br />

(MIC), antimicrobial resistance patterns were as<br />

follow: 25 isolates (29.8%) were resistant to<br />

streptomycin, 25 isolates (29.8%) to sulfamethoxazoletrimethoprim,<br />

30 isolates (35.7%) to trimethoprim, 23<br />

isolates (27.4%) to chloramphenicol, 57 isolates (67.9%)<br />

to tetracycline, 6 isolates (7.1%) to ampicillin, 54 isolates<br />

(64.3%) to nalidexic acid, 1 isolate (1.2%) to ciprofloxacin,<br />

6 isolates (7.2%) to cefotaxime, 8 isolates (9.5%)<br />

to cefexime, 6 isolates (7.2%) to ceftriaxon, 2 isolates<br />

(2.4%) to ceftazidime, 2 isolates (2.4%) to colistinsulfate,<br />

3 isolates (3.6%) to gentamicin, 24 isolates<br />

(28.6%) to spectinomycin, 5 isolates (5.9%) to azithromycin,<br />

2 isolates (2.4%) to amikacin. All the isolates were<br />

sensitive to imipenem, ofloxacin, levofloxacin,<br />

norfloxacin, gatifloxacin, moxifloxacin. Of the 84 isolates<br />

only 4 isolates (4.7%) were sensitive to the all of the<br />

tested antimicrobial agents.<br />

Identification of multidrug resistant (MDR) isolates<br />

Multi-drug resistance was defined as resistance to at<br />

least 4 groups of antimicrobial agents. Of the eighty four


Figure 1. PCR Amplification of intl1 Gene. Lane M: 200bp DNA Ladder. Lane 1: int1 PCR product.<br />

Figure 2. PCR Amplification of intl2 Gene. Lane M: 100bp DNA Ladder. Lane 1: int2 PCR product.<br />

isolates, eleven isolates (13.1%) were considered as<br />

MDR Salmonella serovars (1). These MDR isolates were<br />

attributed to S. Typhi (n=3), S. Typhimurium (n=3), other<br />

non-Typhi serovars (n=5). One of the MDR isolates were<br />

recovered from bone marrow and the rest ten MDR<br />

isolates were originated from stool.<br />

Detection of class 1 and 2 Integrons<br />

Rajaei et al. 2251<br />

Fifty (59.5%) isolates amplified intI1 gene with the amplicon<br />

size of 280bp and fourteen (16.7%) isolates carried<br />

intI2 gene with the amplicon size of 232bp (Figures 1 and<br />

2). Of 50 isolates carrying class 1 integrons, 8 (16%)


2252 Afr. J. Microbiol. Res.<br />

Table 2. Information about integron positive isolates among different sources of Salmonella isolates.<br />

Isolation<br />

source<br />

No. of intI1 positive isolates<br />

(%)<br />

No. of inti2 positive isolates<br />

(%)<br />

No. of intI1 and inti2 positive isolates<br />

(%)<br />

Stool(69) 41 13 13<br />

Blood(6) 3 0 0<br />

Bone marrow(3) 2 0 0<br />

Synovial fluid(3) 1 0 0<br />

Abces(1) 1 0 0<br />

Ascites(1) 1 0 0<br />

Urine(1) 1 1 1<br />

Total(84) 50(59.5) 14(16.7) 14(16.7)<br />

Table 3. Information about integron positive isolates among serovars of Salmonella spp.<br />

Serovars (No.)<br />

No. of intI1 and intI2positive<br />

isolates (%) c<br />

No. of intI2-positive<br />

isolates (%) b<br />

No. of intI1-positive<br />

isolates (%) a<br />

S. Typhi (40) 5 5 21<br />

Non-typhi serovars (30) 8 8 20<br />

S. Typhimurium (12) 1 1 7<br />

S. Paratyphi A (2) 0 0 2<br />

Total (84) 14(16.7) 14(16.7) 50 (59.5)<br />

a indicates the number and percentage of intI1-posetive in Salmonellae serovars<br />

b indicates the number and percentage of intI2-posetive in Salmonellae serovars<br />

c indicates the number and percentage of serovars carrying both intI1 and intI2 genes.<br />

isolates amplified class 1 integrons exclusively on<br />

chromosome, 12 (24%) isolates exclusively on plasmid,<br />

30 (60%) isolates both on plasmid and chromosome.<br />

Among fourteen isolates harbored class 2 integrons, 3<br />

(21.4%) isolates carried class 2 integrons only on<br />

plasmids and 11 (78.6%) isolates both on plasmid and<br />

chromosome (Tables 2 and 3).<br />

DISCUSSION<br />

The worldwide emergence of multidrug-resistant (MDR)<br />

phenotypes among Salmonella serotypes is causing an<br />

increasing concern. Use of antimicrobials in clinical and<br />

veterinary medicine is a recognized driving force for the<br />

selection of resistant bacteria. Selective pressure has<br />

resulted in the development of strains that are resistant<br />

to more than one antimicrobial agent (Miriagou et al.,<br />

2006; Yang et al., 2009). Integrons are genetic elements<br />

able to recognize and capture mobile gene cassettes<br />

carrying antibiotic resistance genes leading to MDR<br />

distribution and subsequently limitation of treatment<br />

options for infections (Stokes and Hall, 1989).<br />

In this study eighty four clinical isolates of Salmonella<br />

spp. were subjected to molecular investigations to detect<br />

integron-associated multidrug resistance.<br />

Of 84 isolates, 11 (13.1%) isolates were resistant to<br />

more than 4 groups of antimicrobial agents and considered<br />

as MDR Salmonella serovars (Table 1). Most of<br />

the MDR isolates were recovered from stool and related<br />

to the non-Typhi serovars of Salmonella enterica. Fifty<br />

(59.5%) isolates harbored class 1 integrons and<br />

amplified intI1 gene more detected in Typhi serovar and<br />

14 (16.7%) isolates contained class 2 integrons carrying<br />

intI2 gene as well as intI1 gene which were more<br />

considered in non–Typhi serovars of Salmonella enterica<br />

(Table 3).<br />

Our study indicates that all the MDR isolates harbored<br />

class 1 integrons and one of the MDR isolates carried<br />

class 2 in addition to class 1 integrons. This result<br />

highlights the integron role in MDR distribution (Table 1).<br />

To confirm the previous studies, according to the<br />

results of MIC determination, most of the intI-positive<br />

isolates showed high resistance to nalidixic acid,<br />

trimethoprim and trimethoprim–sulphamethoxazole<br />

(Chang et al., 2007) and indicated increased MIC to be<br />

higher than 512 µg/ml. Limitless use of these antibiotics<br />

in hospitals and other health care environments may be<br />

involved in the selection and consequently distribution of<br />

integron-carrying Salmonella. These antibiotics are at<br />

risk for future use and would be prescribed with more<br />

consideration.<br />

In agreement with other studies, fluoroquinolones,<br />

third-generation cephalosporins and imipenem are


suggested to be used as frontline therapeutic drugs in<br />

treatment of Salmonella infections. Carbapenems are the<br />

main class of drugs used for treatment of infections<br />

caused by MDR and extended-spectrum -lactamaseproducer<br />

Gram-negative bacteria such as Salmonella<br />

(Ahmed et al., 2009).<br />

In conclusion our results support the hypothesis that<br />

integron acquisition may lead to dissemination of new<br />

antibiotic resistance determinants and high levels of<br />

multidrug resistance. Epidemiological studies on the<br />

distribution of integrons would be of great use in<br />

detecting MDR isolates.<br />

REFERENCES<br />

Ahmed AM, Younis EA, Ishida Y, Shimamoto T (2009). Genetic basis<br />

of multidrug resistance in Salmonella enterica serovars Enteritidis<br />

and Typhimurium isolated from diarrheic calves in Egypt. Acta Trop.,<br />

111: 144-149.<br />

Boyd EF, Hartl DL (1998). Salmonella virulence plasmid: Modular<br />

acquisition of the spv virulence region by an F-plasmid in Salmonella<br />

enterica subspecies I and insertion into the chromosome of<br />

subspecies II, IIIa, IV and VII isolates. Genetics, 149: 1183-1190.<br />

Carattoli A (2001). Importance of integrons in the diffusion of<br />

resistance. Vet. Res., 32: 243-259.<br />

Chang YC, Shih DYC, Wang JY, Yang SS (2007). Molecular<br />

characterization of class 1 integrons and antimicrobial resistance in<br />

Aeromonas strains from foodborne outbreak-suspect samples and<br />

environmental sources in Taiwan. Diagn. Microbiol. Infect. Dis., 59:<br />

191-197.<br />

Clinical and Laboratory Standards Institute (CLSI) (2009). Performance<br />

standards for antimicrobial susceptibility testing; nineteenth<br />

informational supplement. M100-S19, CLSI, Wayne, PA.<br />

Goldstein C, Lee MD, Sanchez S, Hudson C, Phillips B, Register B,<br />

Grady M, Liebert C, Summers AO, White DG, Maurer JJ (2001).<br />

Incidence of Class 1 and 2 Integrases in Clinical and Commensal<br />

Bacteria from Livestock, Companion Animals, and Exotics.<br />

Antimicrob. Agents Chemother., 45: 723–726.<br />

Gu B, Pan S, Wang T, Zhao W, Mei Y, Huang P, Tong M (2008). Novel<br />

cassette arrays of integrons in clinical strains of Enterobacteriaceae<br />

in China. Int. J. Antimicrob. Ag., 32: 529–533.<br />

Rajaei et al. 2253<br />

Havlickova H, Hradecka H, Bernardyova I, Rychlik I (2009). Distribution<br />

of integrons and SGI1 among antibiotic-resistant Salmonella enterica<br />

isolates of animal origin. Vet. Microbiol., 133: 193-198.<br />

Macedo-Vinas M, Cordeiro NF, Bado I, Herrera-Leon S, Vola M,<br />

Robino L, Gonzalez-Sanz R, Mateos S, Schelotto F, Algorta G,<br />

Ayala JA, Echeita A, Vignoli R (2009). Surveillance of antibiotic<br />

resistance evolution and detection of class 1 and 2 integrons in<br />

human isolates of multi-resistant Salmonella Typhimurium obtained<br />

in Uruguay between 1976 and 2000. Int. J. Infect. Dis., 13: 342-348.<br />

Martin BS, Lapierre L, Cornejo J, Bucarey S (2008). Characterization of<br />

antibiotic resistance genes linked to class 1 and 2 integrons in<br />

strains of Salmonella spp. isolated from swine. Can. J. Microbiol., 54:<br />

569-576.<br />

Mazel D (2006). Integrons: agents of bacterial evolution. Nat. Rev.<br />

Microbiol., 4:608-620.<br />

Miriagou V, Carattoli A, Fanning S (2006). Antimicrobial resistance<br />

islands: resistance gene clusters in Salmonella chromosome and<br />

plasmids. Microbes Infect., 8: 1923-1930.<br />

Moura A, Henriques I, Ribeiro R, Correia A (2007). Prevalence and<br />

characterization of integrons from bacteria isolated from a<br />

slaughterhouse wastewater treatment plant. J. Antimicrob.<br />

Chemother., 60: 1243-1250.<br />

Rodriguez I, Rodicio MR, Herrera-Leon S, Echeita A, Mendoza MC<br />

(2008). Class 1 integrons in multidrug-resistant non-typhoidal<br />

Salmonella enterica isolated in Spain between 2002 and 2004. Int. J.<br />

Antimicrob. Ag., 32: 158-164.<br />

Sambrook J, Fritsch EF, Manaiatis T (1989). Molecular Cloning: A<br />

Laboratory Manual, 2nd ed. Cold Spring Harbor Laboratory Press,<br />

Cold Spring Harbor, NY.<br />

Stokes HW, Hall RM (1989). A novel family of potentially mobile DNA<br />

elements encoding site- specific gene-integration functions:<br />

integrons. Mol. Microbiol., 3: 1669-1683.<br />

Tamang MD, Oh JY, Seol SY, Kang HY, Lee JC, Lee YC, Cho DT, Kim<br />

J (2007). Emergence of multidrug-resistant Salmonella enterica<br />

serovar Typhi associated with a class 1 integron carrying the dfrA7<br />

gene cassette in Nepal. Int. J. Antimicrob. Ag., 30: 330-335.<br />

Yang BW, Zheng J, Brown EW, Zhao SH, Meng JH (2009).<br />

Characterisation of antimicrobial resistance-associated integrons and<br />

mismatch repair gene mutations in Salmonella serotypes. Int. J.<br />

Antimicrob. Ag., 33: 120-124.


African Journal of Microbiology Research Vol. 5(16), pp. 2254-2260, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.580<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

The synthesis and role of hydroxyectoine in halophilic<br />

bacterium Halomonas ventosae DL7<br />

Daochen Zhu 1,2 *, Chenxiang Wang 3 , Shoko Hosoi-Tanabe 4 , Weimin Zhang 2 and<br />

Shinichi Nagata 4<br />

1 School of Environmental Engineering, Jiangsu University, 301 Xuefu Road Zhenjiang 212013, China.<br />

2 Guangdong Provincial Open Laboratory of Applied Microbiology, Guangdong Provincial, Key Laboratory of Microbial<br />

Culture Collection and Application, Guangdong Institute of Microbiology, Guangzhou 510070, China.<br />

3 Environmental Science and Engineering College, Dalian Maritime University, 1 Linghai Road, Dalian 116026, China.<br />

4 Environmental Biochemistry Division, Research Center for Inland Seas, Kobe University, 5-1-1 Fukae, Higashinada-ku,<br />

Kobe 658-0022, Japan.<br />

Accepted 30 July, 2011<br />

Hydroxyectoine and ectoine were accumulated in the moderately halophilic bacterium Halomonas<br />

ventosae DL7 as dominant compatible solutes in response to the external thermo- and osmo-stresses<br />

(30-42°C, 1-3 M NaCl). As the byproduct of ectoine, hydroxyectoine was directly converted from ectoine<br />

mediated by the ectoine hydroxylase in DL7, and the hydroxylase gene sequence was obtained by PCR.<br />

Further, the activity of ectoine hydroxylase of strain DL7 cells in vitro was determined at 30 to 42°C, and<br />

the thermo-stress conduce the inhibition of this enzyme activity. In addition, DL7 cells showed similar<br />

growth level when grown in presence of same level ectoine or hydroxyectoine (1 mM) in medium with<br />

high salinity (2-2.7 M NaCl). However, the presence of hydroxyectoine showed higher growth level than<br />

ectoine when DL7 cells were incubated at 42°C. The results showed that the alteration of temperature<br />

instead of salinity stress obviously stimulated the expression of ectoine hydroxylase, but inhibited its<br />

activity, and hydroxyectoine with ectoine together played an important role as thermo- and osmoprotectants<br />

in strain DL7 to cope with high temperature and salinity environment.<br />

Key words: Halomonas ventosae DL7, compatible solutes, hydroxyectoine, ectoine hydroxylase, thermostress,<br />

osmo-stress.<br />

INTRODUCTION<br />

To cope with a variety of high osmotic environments,<br />

halophilic and halotolerant bacteria developed a strategy<br />

to accumulate a particular class of highly soluble, small<br />

molecular organic compounds which named compatible<br />

solutes (Kunte, 2006; Roberts, 2005). Among compatible<br />

solutes, ectoine and its hydroxylated derivative,<br />

hydroxyectoine are one of the most widely produced<br />

solutes. And ectoines are increasingly concerned for their<br />

superior function on the enzyme, nucleic acid and cell<br />

wall protective ability against a variety of stress factors<br />

such as heating, freeze-thawing and freeze-drying<br />

*Corresponding author. E-mail: dczhucn@hotmail.com. Tel/fax:<br />

+86-511-88796122.<br />

(Lippert and Galinski, 1992; Louis et al., 1994). The OH<br />

group of hydroxyectoine can partly replace the loss of<br />

water molecules from hydrate shell to further stabilize the<br />

native structure of the biopolymers compare to ectoine<br />

(Graf et al., 2008). Hydroxyectoine is proposed as<br />

suitable healthy cells protectant during chemotherapy<br />

and molecular chaperon for Alzheimer's disease treatment,<br />

and it can protect skin cells against the aging and<br />

function as the PCR enhancer to improve the yield of<br />

target production (Furusho et al., 2005; Kanapathipillai et<br />

al., 2005). It would have a significantly applicable value in<br />

the field of bio-industry, medical therapy, and cosmetic<br />

ingredients, etc. Therefore, the synthesis pathway of<br />

hydroxyectoine and its physiological role in microorganisms<br />

were interesting to researchers. Studies have<br />

suggested that the biosynthesis of hydroxyectoine as


thermo-protectant has been proved to parallel with high<br />

environmental temperature (García-Estepa et al., 2006;<br />

Prabhu et al., 2004). The biosynthetic pathway of hydroxyectoine<br />

was directly converted via ectoine hydroxylase<br />

from ectoine in halophilic or halotolerant bacteria and<br />

induced by osmotic stress of high salinity, and ectoine<br />

hydroxylase has been found in strains Streptomyces<br />

chrysomallus, Streptomyces coelicolor and<br />

Chromohalobacter salexigens (Bursy et al., 2008; García-<br />

Estepa et al., 2006; Prabhu et al., 2004). Further study<br />

revealed that the ectoine hydroxylase is a member of the<br />

non-heme-containing iron (II) and 2-oxoglutaratedependent<br />

dioxygenase superfamily, and its in vitro<br />

activity depends on iron (II), molecular oxygen, and 2oxoglutarate<br />

(Bursy et al., 2008; Reuter et al., 2010). The<br />

chemical synthesis of hydroxyectoine, however, is still<br />

difficult at present. Therefore, the production of hydroxyectoine<br />

by microorganisms is currently a simple and<br />

meaningful method. To produce hydroxyectoine using<br />

halophilic bacteria, the fed-batch and milking method has<br />

been developed and the effective factors for the<br />

synthesis of hydroxyectoine in the production process<br />

were studied. For example, the carbon source xylose can<br />

significantly improve the conversion of ectoine to<br />

hydroxyectoine in recombinant E. coli BL21, which<br />

carries ectoine hydroxylase gene (thpD) (Nagata et al.,<br />

2008b).<br />

Moderately halophilic bacterium, Halomonas ventosae<br />

DL7, which has been isolated from Dalian saltern, China,<br />

displays the ability to synthesize a large amount of<br />

ectoine as well as minor hydroxyectoine under hyperosmolality<br />

(Zhu et al., 2007). And the synthesized amount<br />

of hydroxyectoine significantly increased in strain DL7<br />

when subjected to heat stimulation (Zhu et al., 2008b).<br />

Strain DL7 as an excellent ectoine or hydroxyectoine<br />

producer has a potential application value for producing<br />

ectoines in factory on a larger scale. But there is still<br />

unclear for the effect factor of hydroxyectoine synthesize<br />

and activity of ectoine hydroxylase in DL7. Therefore, to<br />

elucidate the role and synthesis characteristics of<br />

hydroxyectoine in DL7 is an interesting work.<br />

The goal of this study was to investigate the effects of<br />

hydroxyectoine and ectoine on the growth of strain DL7<br />

when subjected to high salinity and temperature environments.<br />

And the in vitro activity of ectoine hydroxylase<br />

with the fluctuant salinity and temperature were<br />

determined. We also evaluated hydroxyectoine as a vital<br />

protectant for strain DL7 cells to defend off the<br />

environmental thermo-stress.<br />

MATERIALS AND METHODS<br />

Bacteria and growth media<br />

H. ventosae DL7 was aerobically grown in a nutrient medium (g/l):<br />

Bacto peptone (Difco Laboratories, Detroit, MI, USA), 5.0 and Bacto<br />

yeast extract (Difco), 3.0. Glucose mineral medium (MM63) was<br />

also used in this study: 100 mM KH2PO4, 75 mM KOH, 15 mM<br />

Zhu et al. 2255<br />

(NH4)2SO4, 1 mM MgSO4, 3.9 µM FeSO4, and 20 mM glucose as<br />

carbon source. NaCl was added as described in the text and pH<br />

was adjusted to 7.2 with NaOH.<br />

PCR amplification and gene analysis<br />

To isolate the gene encoding ectoine hydroxylase of H. ventosae<br />

DL7, we designed degeneration of oligonucleotides for PCR. Based<br />

on the ectD conservation regions from Virgibacillus salexigens<br />

(AY935522) and Chromohalobacter salexigens (AM231629), the<br />

universal degenerate primers (5’-GAMAGCMAKGASATCCGKT-3’<br />

and 5’-GARCCATGCAKYGTRTT-3’) were employed. Genomic<br />

DNA of H. ventosae DL7 and C. salexigens DSM 3043T were<br />

isolated using Genomic DNA Purification Kit (Promega, Madison,<br />

WI, USA) according to the manufacturer’s protocol. PCR was<br />

performed under the following condition: 5 min at 94°C, 30 cycles of<br />

30 s at 94°C plus 1 min at 55°C plus 30 s at 72°C, and finally 5 min<br />

at 72°C. Electrophoresis was performed on 1% (w/v) agarose gel in<br />

TAE buffer at 100 V for 30 min. The PCR product on the gel was<br />

extracted by using a PCR Purification Kit (QIAGEN, Hilden,<br />

Germany), cloned into the pGEM-T Vector System (Promega), and<br />

sequenced using an ABI PRISMA 377 sequencer. The nucleotide<br />

sequence of the PCR product has been deposited in the GSDB,<br />

DDBJ, EMBL, and NCBI databases (Accession No. EU732521).<br />

Enzyme assay and the conversion of ectoine to<br />

hydroxyectoine<br />

Ectoine hydroxylase activity in the extracts of DL7 cells were<br />

assayed as described before (Bursy et al., 2007), 70 µl protein<br />

fractions were suspended in 100 µl reaction mixture that contained<br />

10 mM TES buffer, pH 7.5, 10 mM 2-oxoglutarate, 5 mM ectoine, 1<br />

mM FeSO4 . 7H2O, 1.3 kilo units beef liver catalase (Roche,<br />

Germany). The reaction mixture was incubated at 30 or 42°C for 20<br />

min and stopped by adding 100 µl acetonitrile. Supernatant was<br />

analyzed by HPLC. One unit of ectoine hydroxylase activity is<br />

defined as the conversion of 1 µM of ectoine to 1 µM of<br />

hydroxyectoine per min.<br />

Release of hydroxyectoine and ectoine by downshock<br />

H. ventosae DL7 was aerobically grown in the nutrient medium<br />

containing 3 M NaCl until stationary phase of growth and harvested.<br />

The pellets were washed once by KPi buffer with the same NaCl<br />

concentration as that of corresponding nutrient medium, and then<br />

resuspended in deionized water or 2% NaCl solution. The cells<br />

were incubated for 10-120 min at 30°C. After centrifugation the<br />

supernatant fractions were analyzed by HPLC. Release rate of<br />

hydroxyectoine and ectoine from the cells were calculated as<br />

described previously (Nagata et al., 2008a).<br />

Extraction and determination of compatible solutes<br />

Cells were aerobically grown in the nutrient medium containing 1-3<br />

M NaCl at 30 or 42°C. Cells were harvested by centrifuging<br />

(10,000×g, 4°C, 10 min), and then the pellets were washed once<br />

with 50 mM potassium phosphate (KPi) buffer (pH 7.2) containing<br />

the same concentration of NaCl corresponding to growth medium.<br />

Extraction of intracellular compatible solutes was carried out as<br />

described previously (Zhu et al., 2008a). Fraction of 80% (v/v)<br />

ethanol extraction as well as supernatant after osmotic downshock<br />

analyzed and quantified by HPLC system (L-5020, Hitachi, Tokyo,<br />

Japan) with a Bio-Sil C8 90-5S reversed phase column (Bio-Rad


2256 Afr. J. Microbiol. Res.<br />

Growth (OD600)<br />

3.5<br />

3<br />

2.5<br />

2<br />

1.5<br />

1<br />

0.5<br />

Laboratories, Hercules, CA, USA) as described previously (Nagata<br />

et al., 2006; Zhu et al., 2010). Five microliters of samples were<br />

eluted with 50 mM KPi buffer at pH 6.0 with a flow rate of 1 ml/min<br />

at 35°C and detected by UV absorbance at 210 nm. Concentrations<br />

of ectoine and hydroxyectoine were expressed as milligram per<br />

gram CDW.<br />

RESULTS AND DISCUSSION<br />

Effect of temperature on the growth of H. ventosae<br />

DL7<br />

The influence of temperature on the cells growth of strain<br />

DL7 was examined using the nutrient medium containing<br />

3 M NaCl at 30-42°C. Results showed that the growth of<br />

cells was severely inhibited with the increasing of<br />

incubation temperature, and reached stationary phase at<br />

36 h of incubation at 30°C, which prolonged to 57 and 66<br />

h with the incubation temperature of 37 and 42°C,<br />

respectively. Optical density (OD600) was significantly<br />

reduced from 3.3 at 30°C to 2.8 at 37°C and 2.1 at 42°C,<br />

respectively (Figure 1). In our previous report, the amount<br />

of hydroxyectoine synthesized was very low comparing<br />

with ectoine when cells grown at 30°C and the<br />

concentration of hydroxyectoine in the cells remarkably<br />

increased with the increase of temperature and reached<br />

the maximum of 36.6 mg/g CDW at 42°C, (Zhu et al.,<br />

0<br />

A<br />

0 10 20 30 40 50 60 70 80<br />

Incubation Time time (h) (h)<br />

Figure 1. The growth of strain DL7 cells at different incubation<br />

temperatures. Strain DL7 was pre-incubated in a nutrient medium<br />

containing 3 M NaCl for 18 h at 30°C and then separately incubated<br />

at 30°C (cycles), 37°C (diamonds), or 42°C (triangles). Values are<br />

the averages ± SD from three independent experiments.<br />

2008a). It suggests that the synthesis of hydroxyectoine<br />

was a strategy of strain DL7 cells to defend off the<br />

environmental thermo-stress.<br />

Ectoine hydroxylase in H. ventosae DL7<br />

Ectoine hydroxylase catalyzing the conversion of ectoine<br />

into hydroxyectoine had been proved in several halophilic<br />

bacteria (Bursy et al., 2007; García-Estepa et al., 2006;<br />

Nagata et al., 2008a). The PCR strategy was carrying out<br />

to clarify whether this hydroxylase was present in strain<br />

DL7, and we assumed that a homologous hydroxylase<br />

gene to those of C. salexigens and V. salexigens was<br />

existed in this strain to design a pair of degenerated<br />

primers for PCR amplification, and a 480-bp gene<br />

fragment was obtained and subsequently sequenced<br />

(EU732521), the amino acid sequence deduced from the<br />

DNA sequence of this fragment showed 54.8% identity<br />

with the ectoine hydroxylases from C. salexigens and V.<br />

salexigens (Figure 2). These data confirmed the presence<br />

of hydroxylase in H. ventosae DL7, by which the<br />

conversion of ectoine should be the important pathway<br />

for the synthesis of hydroxyectoine. To prove this opinion,<br />

the activity of ectoine hydroxylase of strain DL7 was<br />

determined.<br />

To investigate the activity of ectoine hydroxylase under


Zhu et al. 2257<br />

Figure 2. Alignment of the sequences of reading frame ectD (EU732521) of H. ventosae DL7 with the sequences of the ectoine<br />

hydroxylases of V. salexigens (AY935522) and C. salexigens (AM231629). Identical positions and conservative fragments are shown<br />

against a dark and gray background, respectively.<br />

Table 1. Ectoine hydroxylase activity (cell free extract) from DL7*<br />

Reaction temperature (°C) NaCl (M) Activity (Unit) Protein (mg)**<br />

30 0 43.7 916.5<br />

42 0 29.5 920.3<br />

30 1 42.1 918.4<br />

*Cell free extract was prepared from 20 g cell pellets grown in 3 M NaCl at 30°C.<br />

** Protein (mg): the protein content of 70 µl protein fractions extract from DL7 cells.<br />

the heat or salt stress, we assayed the cell-free extracts<br />

from strain DL7 cells in vitro at 30 or 42°C, without or with<br />

1 M NaCl. Results showed that the activity of hydroxylase<br />

was reduced from 43.7 to 29.5 U, when the temperature<br />

was increased from 30 to 42°C (Table 1). However, the<br />

activity was stable with the fluctuation of NaCl concentration<br />

(0-1 M) at 30°C. It revealed that the conversion of<br />

ectoine to hydroxyectoine was inhibited by the higher<br />

incubation temperature. On the other hand, the activity of<br />

hydroxylase had no obviously fluctuation with or without<br />

salt. In addition, similar result has also been found in our<br />

previous research, i.e., the in vivo activity of hydroxylase<br />

was inhibited by high temperature, and hydroxyectoine<br />

showed similar conversion rate in the presence of<br />

different NaCl concentrations (0.5-1 M) (Zhu et al.,<br />

2008a). Therefore, these in vivo and in vitro data<br />

supported our suggestion that the hydroxylase activity<br />

was sensitive to temperature but was not disturbed by<br />

salinity.<br />

Based on these data, we could deduce that the<br />

increased amount of hydroxyectoine synthesized<br />

following the enhancing of thermo-stress might be that<br />

the heat stimulated the cells to produce more amount of<br />

ectoine hydroxylase instead of induction of the increasing<br />

of the enzyme activity. Further, to detect whether there is<br />

a high temperature intermediated operon to trigger the<br />

synthesis of hydroxylase in DL7 is an interesting work in<br />

future.<br />

Release of hydroxyectoine and ectoine by osmotic<br />

downshock<br />

To investigate the role of hydroxyectoine and ectoine for


2258 Afr. J. Microbiol. Res.<br />

Release rate (%)<br />

96<br />

92<br />

88<br />

84<br />

80<br />

10 20 40 60 120<br />

Time (min)<br />

Figure 3. Releases of hydroxyectoine and ectoine by H. ventosae DL7 subjected to<br />

osmotic downshock. Downshock procedures were handled in deionized water to<br />

release ectoine (white bars) and hydroxyectoine (hatched bars), or in 2% NaCl to<br />

release ectoine (black bars) and hydroxyectoine (macula bars) during 10-120 min<br />

incubation. Values are the averages ± SD from three independent experiments.<br />

strain DL7 in response to the salt or heat stress, it is<br />

necessary to use the “clearing cells”, which have no<br />

intracellularly compatible solutes. Therefore, the release<br />

of compatible solutes from cells and at the same time to<br />

keep alive of cells was concerned for preparation of the<br />

compatible solutes free cells. When halophilic bacteria<br />

are confronted with a rapid osmotic downshock, most of<br />

the small components in the cytoplasm were released<br />

into the environments (Frings et al., 1995; Nagata et al.,<br />

1996; 1998; Sauer et al., 1998). In this consideration, we<br />

tried to examine the release rates of hydroxyectoine and<br />

ectoine following a shift in medium osmolarity, that is,<br />

cells were suspended in 2% (0.34 M) NaCl or deionized<br />

water from 3 M NaCl medium. When the strain was<br />

downshocked in deionized water, both intracellular<br />

ectoine and hydroxyectoine were rapidly released to<br />

environment. During less than 10 min of downshock<br />

treatment, 90.8 and 89.9% of release rates were<br />

achieved for ectoine and hydroxyectoine, respectively<br />

(Figure 3). However, since strain DL7 is a moderately<br />

halophilic bacterium and a minimum concentration of Na +<br />

is essential for growth, the cells would burst by osmotic<br />

downshock from high salinity environment to deionized<br />

water and could not be recovered to use for further study.<br />

To remain the growth of cells after downshock, we tried<br />

to examine 2% NaCl solution instead of deionized<br />

water. As a consequence, the cells were kept intact after<br />

re-incubation in 2% NaCl solution for 1 h (data not<br />

shown). Release rates of ectoine and hydroxyectoine<br />

reached maximum at 60 min of treatment, and resulted in<br />

90.1 and 88.4%, respectively. The survival rates of cells<br />

treated by downshock (3 M NaCl to 2% NaCl and 2 M<br />

NaCl to 2% NaCl) were determined as 95.3 and 97.6%,<br />

respectively. It was suggested that the numbers of<br />

damaged cells were negligible for the present condition,<br />

and 2% NaCl solution was chosen to prepare the solutes<br />

free cells for the following study.<br />

Thermo- and osmo-protection of compatible solutes<br />

To gain insight into the role of hydroxyectoine and<br />

ectoine in strain DL7 when cells were subjected to<br />

thermo-stress, we tried to examine the behavior of strain<br />

DL7 under high temperature and salinity in the presence<br />

or absence of compatible solutes. Cells were osmotic<br />

downshock treated by 2% NaCl solution to avoid the<br />

interference of intracellular solutes, and pellets were then<br />

resuspended in MM63 medium containing 1-2.7 M NaCl<br />

without or with 1 mM each of ectoine or hydroxyectoine at<br />

30 or 42°C. We obviously observed that the presences of<br />

hydroxyectoine and ectoine significantly promoted the


Growth (OD600)<br />

3.5<br />

3<br />

2.5<br />

2<br />

1.5<br />

1<br />

A<br />

A<br />

0 10 20 30 40 50 60 70<br />

Time (h)<br />

Growth (OD600)<br />

2.5<br />

2.25<br />

2<br />

1.75<br />

1.5<br />

1.25<br />

1<br />

B<br />

B<br />

0 12 24 36 48 60 72<br />

Incubation Time (h) time (h)<br />

Figure 4. Supplement effects of hydroxyectoine and ectoine on the growth of H. ventosae DL7 under high<br />

temperatures or high salinities. A. Cells were grown in MM63 medium containing 1 M NaCl at 42°C (open symbols)<br />

or 30°C (closed symbols) without (triangles), with 1 mM ectoine (circles) or 1 mM hydroxyectoine (squares). B.<br />

Cells were grown in MM63 medium containing 2 M (open symbols) or 2.7 M NaCl (closed symbols) at 30°C with 1<br />

mM ectoine (circles) and 1 mM hydroxyectoine (squares). Values are the averages ± SD from three independent<br />

experiments.<br />

Table 2. The accumulation of ectoine and hydroxyectoine by downshocked cells of H. ventosae DL7*<br />

Compatible solutes NaCl (M)<br />

Ectoine<br />

Hydroxyectoine<br />

Concentrations (µM /g CDW)<br />

Incubation time (h)<br />

1 3 5 10<br />

1 241±14.8 245±10.6 253±3.5 258±7.8<br />

2 327±10.6 368±12.7 395±5.6 411±4.2<br />

1 234±12.7 244±2.8 241±7.1 244±10.6<br />

2 304±9.1 355±4.2 369±5.7 390±4.9<br />

*Downshock treatment was performed according to the method described in Materials and Methods. The<br />

downshocked cells were incubated in MM63 medium at 42°C with 1 mM each ectoine or hydroxyectoine. The values<br />

are the means ± SD from three independent experiments.<br />

growth of cells at 42°C from the growth curve, and the<br />

optical density of hydroxyectoine showed 1.5 times<br />

higher than ectoine after 24 h of incubation (Figure 4A).<br />

However, there was no obviously difference between the<br />

growth of DL7 in presence of hydroxyectoine and ectoine<br />

at 30°C. In addition, the downshocked compatible solutes<br />

free DL7 cells to accumulate hydroxyectoine and ectoine<br />

reached almost same level with 1 M or 2 M NaCl<br />

concentrations (Table 2). It suggests that the presence of<br />

hydroxyectoine result in the growth of cells higher than<br />

ectoine were not induced by different concentrations of<br />

hydroxyectoine and ectoine accumulated in cells. The<br />

above data elucidated that hydroxyectoine protected DL7<br />

Zhu et al. 2259<br />

cells more effectively than ectoine at 42°C.<br />

To investigate the protection functions of hydroxyectoine<br />

and ectoine on H. ventosae DL7 to defend off<br />

high salinity, the compatible solutes free cells were<br />

incubated in MM63 medium containing 2 or 2.7 M NaCl in<br />

presence of 1 mM each of hydroxyectoine or ectoine and<br />

the growth was detected. Results verified that the<br />

supplementation of ectoine or hydroxyectoine conferred a<br />

similar cell growth both in presence of 2 and 2.7 M NaCl<br />

(Figure 4B), that is, ectoine and hydroxyectoine have<br />

similar protection effect for cells grown under high salinity<br />

environment.<br />

It was understood that the accumulation of compatible


2260 Afr. J. Microbiol. Res.<br />

solutes was osmo-adapted by environmental stresses for<br />

halophilic bacteria. In this study, we described the<br />

synthesis of hydroxyectoine and activity of hydroxylase in<br />

strain DL7, showed that thermo-stress stimulated the<br />

yield of hydroxyectoine but inhibited the activity of<br />

hydroxylase. We also noticed the hydroxylase express<br />

system might be induced by thermo-stress instead of<br />

osmotic-stress. The study for charactering the<br />

hydroxylase in structure and exploring its molecular<br />

mechanism are interesting work in the near future.<br />

ACKNOWLEDGEMENT<br />

This work was supported by a grant (Code:<br />

9451007002003073) from the Natural Science<br />

Foundation of Guangdong Province, China, and by the<br />

Project Startup Fundation for Distinguished Scholars of<br />

Jiangsu University (Code: 10JDG084).<br />

REFERENCES<br />

Bursy J, Pierik AJ, Pica N, Bremer E (2007). Osmotically induced<br />

synthesis of compatible solute hydroxyectoine is mediated by an<br />

evolutionarily conserved ectoine hydroxylase. J. Biol. Chem., 282:<br />

31147-31155.<br />

Bursy J, Kuhlmann AU, Pittelkow M, Hartmann H, Jebbar M, Pierik AJ,<br />

Bremer E (2008). Synthesis and uptake of the compatible solute<br />

ectoine and 5-hydroxyectoine by Streptomyces coelicolor A3(2) in<br />

response to salt and heat stresses. Appl. Environ. Microbiol., 74:<br />

7286-7296.<br />

Frings E, Sauer T, Galinski EA (1995). Production of hydroxyectoine:<br />

high cell-density cultivation and osmotic downshock of Marinococcus<br />

strain M52. J. Biotechnol., 61: 117-158.<br />

Furusho K, Yoshizawa T, Shoji S (2005). Ectoine alters subcellular<br />

localization of inclusions and reduces apoptotic cell death induced by<br />

the truncated Machado-Joseph disease gene product with an<br />

expanded polyglutamine stretch. Neurobiol. Dis., 20: 170-178.<br />

García-Estepa R, Argandoña M, Reina-Bueno M, Capote N, Iglesias-<br />

Guerra F, Nieto JJ, Vargas C (2006). The ectD gene, which is<br />

involved in the synthesis of the compatible solute hydroxyectoine, is<br />

essential for thermoprotection of the halophilic bacterium<br />

Chromohalobacter salexigens. J. Bacteriol., 188: 3774-3784.<br />

Graf R, Anzali S, Buenger J, Pfluecker F, Driller H (2008). The<br />

multifunctional role of ectoine as a natural cell protectant. Clin.<br />

Dermatol., 26: 326-333.<br />

Kanapathipillai M, Lentzen G, Sierks M, Park CB (2005). Ectoine and<br />

hydroxyectoine inhibit aggregation and neurotoxicity of Alzheimer’s -<br />

amyloid. FEBS Lett., 579: 4775-4780.<br />

Kunte HJ (2006). Osmoregulation in bacteria: compatible solute<br />

accumulation and osmosensing. Environ. Chem., 3: 94-99.<br />

Lippert K, Galinski EA (1992). Enzyme stabilization by ectoine-type<br />

compatible solutes: protection against heating, freezing and drying.<br />

Appl. Microbiol. Biotechnol., 37: 61-65.<br />

Louis P, Truper HG, Galinski EA (1994). Survival of Escherichia coli<br />

during drying and storage in the presence of compatible solutes.<br />

Appl. Microbiol. Biotechnol., 41: 684-688.<br />

Nagata S, Adachi K, Sano H (1996). NMR analyses of compatible<br />

solutes in a halotolerant Brevibacterium sp. Microbiology., 142: 3355-<br />

3362.<br />

Nagata S, Adachi K, Sano H (1998). Intracellular changes in ions and<br />

organic solutes in halotolerant Brevibacterium sp. strain JCM 6894<br />

after exposure to hyperosmotic shock. Appl. Environ. Microbiol., 64:<br />

3641-3647.<br />

Nagata S, Wang C (2006). Effect of duration of osmotic downshock and<br />

coexisting glutamate on survival and uptake of ectoine in halotolerant<br />

Brevibacterium sp. JCM 6894. J. Biosci. Bioeng., 91: 288-293.<br />

Nagata S, Wang Y, Oshima A, Zhang L, Miyake H, Sasaki H, Ishida A.<br />

(2008a). Efficient cyclic system to yield ectoine using Brevibacterium<br />

sp. JCM 6894 subjected to osmotic downshock. J. Biotechnol.<br />

Bioeng., 99: 941-948.<br />

Nagata S, Zhu D, Cui S, Sasaki H (2008b). Yield of compatible solute<br />

hydroxyectoine through direct synthesis and conversion from ectoine.<br />

In: Schafer HA, Wohlbier LM (eds) Biamino Amino Acid Research.<br />

New York, Nova Science Publishers, pp. 139-153.<br />

Prabhu J, Schauwecker F, Grammel N, Keller U, Bernhard M (2004).<br />

Functional expression of the ectoine hydroxylase gene (thpD) from<br />

Streptomyces chrysomallus in Halomonas elongata. Appl. Environ.<br />

Microbiol., 70: 3130-3132.<br />

Reuter K, Pittelkow M, Bursy J, Heine A, Craan T, Bremer E (2010).<br />

Synthesis of 5-Hydroxyectoine from Ectoine: Crystal Structure of the<br />

Non-Heme Iron(II) and 2-Oxoglutarate-Dependent Dioxygenase<br />

EctD. PLoS One., 5: e10647.<br />

Roberts MF (2005). Organic compatible solutes of halotolerant and<br />

halophilic microorganisms. Saline Syst., 1: 5.<br />

Sauer T, Galinski EA (1998). Bacterial milking: A novel bioprocess for<br />

production of compatible solutes. Biotechnol. Bioeng., 57: 306-313.<br />

Zhu D, Niu L, Wang C, Nagata S (2007). Isolation and characterisation<br />

of moderately halophilic bacterium Halomonas ventosae DL7<br />

synthesizing ectoine as compatible solute. Ann. Microbiol., 57: 401-<br />

406.<br />

Zhu D, Osman O, Nagata S (2008a). Physiological role of compatible<br />

solute hydroxyectoine, synthesized in the halophilic Halomonas<br />

ventosae DL7. Proceedings in OCEANS'08 MTS/IEEE.4530985, pp.<br />

1-3.<br />

Zhu D, Cui S, Nagata S (2008b). Isolation and characterization of saltsensitive<br />

mutants of the moderately halophilic bacterium Salinivibrio<br />

costicola subsp. yaniae. Biosci. Biotechnol. Biochem., 72: 1977-1982.<br />

Zhu D, Zhang W, Nagata S (2010). Accumulation and role of compatible<br />

solutes in bacterium Salinivibrio costicola subsp. yaniae with fast<br />

growth rate. Can. J. Microbiol., 56: 1020-1027.


African Journal of Microbiology Research Vol. 5(16), pp. 2261-2265, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.585<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Virulence and characteristics of a new<br />

nucleopolyhedrovirus strain of Dendrolimus kikuchii<br />

(Lepidoptera: Lasiocampidae)<br />

Miao Miao Yang 1,2,6, Meng Lou Li 1 , Yu Zhu Wang 2 , Liang Jian Qu 2 , Ke Yue Huai 3 ,<br />

Xue Wen Nie 4 , Lu Qin Qiao 5 , Jin Ying Ding 6 and Yong An Zhang 2 *<br />

1 Forestry Entomology Laboratory of <strong>Academic</strong> Forestry, Northwest Agriculture and Forest University,<br />

Yangling 712100 China.<br />

2 The Key Laboratory of Forest Protection, State Forestry Administration of China, Research Institute of Forest Ecology,<br />

Environment and Protection, Chinese Academy of Forestry, Beijing 100091 China.<br />

3 Forestry Research Institute of Yunnan, Yunnan 650000 China.<br />

4 Forest Protection Station Mile County of Yunnan, Yunnan 652300 China.<br />

5 College of Plant Protection, Shandong Agricultural University, Tai’an 271018 China.<br />

6 Ningxia agricultural school, Yinchuan 750000 China.<br />

Accepted 30 June, 2011<br />

In present study, a nucleopolyhedrovirus was isolated from the infected larvae of Dendrolimus kikuchii,<br />

which is a serious pest for a variety of conifers in China. Ultrastructural observation revealed that this<br />

virus is a multiple-nucleocapsid nucleopolyhedrovirus (MNPV), which was encapsulated within the<br />

envelope of each virion. Based on the morphological characteristics, it was identified as a new virus<br />

and named as DkNPV. It exhibited high potential as a biocontrol agent to protect the forest because of<br />

the high virulence against the third-instar larvae of D. kikuchii. The virus passage in the host after 4<br />

generations, toxicity and ultrastructural morphology were stable.<br />

Key words: Dendrolimus kikuchii, multiple-nucleocapsid nucleopolyhedrovirus, Dendrolimus Kikuchii<br />

Matsumura nuclear polyhedrosis virus (DkNPV), virulence, ultrastructure, biological control.<br />

INTRODUCTION<br />

Dendrolimus kikuchii (Matsumura) (Lepidoptera:<br />

Lasiocampidae), is a serious pest for a variety of<br />

conifers, including Pinus kesiya, Pinus yunnanensis,<br />

Pinus massoniana, Pinus armandi, Pinus fenzeliana,<br />

Cedrus deodara and Keteleeria evelyniana (Figure 1a).<br />

In China, D. kikuchii mainly occurs in the south part and<br />

chemical pesticides were primarily used as the first<br />

control method (Chen, 1990). However, chemical insecticides<br />

may poison the non-target organisms (e.g.,<br />

humans, livestock and natural enemy), pollute<br />

environment, and induce insecticide-resistant pests.<br />

*Corresponding author. zhangyab@caf.ac.cn. Tel: +86-<br />

1062889520. Fax: +86-10 62884972.<br />

These problems proved that the application of chemical<br />

insecticides to be less than an ideal method to controll D.<br />

kikuchii (Yasuhisa, 2007).<br />

The insect virus is one kind of pathogenic<br />

microorganism in nature and may influence the quantity<br />

of insect population by causing the epidemic disease of<br />

insect. It has high host specificity, so that it is safe to<br />

other non-target species and can maintain dynamic<br />

balance of insect population for a long term by horizontally<br />

and vertically spreading epidemic disease in the<br />

pest population (Fuxa, 1989; Laird et al., 1990; Heinz et<br />

al., 1995). Moreover, it causes no environmental<br />

pollution. Therefore, it is realized that the use of virus to<br />

control pest insects is an ideal and sustainable means. In<br />

1986, a nucleopolyhedrovirus of D. kikuchiii, named as<br />

Dendrolimus Kikuchii Matsumura Nuclear Polyhedrosis


2262 Afr. J. Microbiol. Res.<br />

Virus (DKMNPV), was isolated and its morphological<br />

characteristics and toxicity were described, however, D.<br />

kikuchii larvae were not sensitive to DKMNPV (Yang et<br />

al., 1986). Therefore, the screening of high toxicity and<br />

strong stability virus is an imperative work for the<br />

biological control of D. kikuchii. In present study, a<br />

nucleopolyhedrovirus was isolated from diseased D.<br />

kikuchii larvae in Mile county, Yunnan province, China.<br />

Based on its ultrastructural morphological characteristics<br />

and the virulence against the third-instar larvae of D.<br />

kikuchii, it was identified as a new virus and named as<br />

DkNPV (multiple-nucleocapsid nucleopolyhedrovirus of<br />

D. kikuchii). This virus exhibited high virulence against<br />

the third-instar larvae of D. kikuchii, and high potential as<br />

a biocontrol agent to protect the forest from these insects.<br />

MATERIALS AND METHODS<br />

Virus isolation and purification from D. kikuchii<br />

Diseased D. kikuchii larvae were collected from P. yunnanensis<br />

forest in Mile, Yunnan Province, China, and kept frozen at -20°C<br />

until use. Virus occlusion bodies (OBs) were isolated and purified<br />

from the larval cadavers according to the method developed by EI-<br />

Salamouny (1998). Briefly, 200 g larvae cadavers were<br />

homogenized in 500 ml distilled water and filtered through five<br />

layers of muslin. The filtrate was subjected to centrifugation<br />

alternated between low and high speed several times until the clear<br />

off-white OBs were obtained. For further purification, the obtained<br />

OBs were centrifuged through sucrose gradient of 40-60% (w/w) at<br />

10,000 g for 30 min at room temperature. The bands containing<br />

virus were collected, washed with sterile water, and then<br />

centrifuged at 12,000 g for 30 min at 4°C with three replications.<br />

The pellets of purified OBs were stored at 4°C for the next study.<br />

Ultrastructural studies of virus isolated from D. kikuchii<br />

As to scanning electron microscopy (SEM), the following method<br />

was used. Firstly, pellets of purified virus were diluted in distilled<br />

water and the concentration of virus was adjusted to 10 9 PIB·ml -1<br />

(polyhedral inclusion bodies/ ml -1 ). Secondly, 20-30 l of viral<br />

suspensions were mounted on a clear coverslip and dried naturally<br />

in the ventilate place. After that, the coverslip was placed on an<br />

aluminous specimen stub and coated with gold. Finally, the<br />

specimen was observed by using a scanning electron microscope<br />

(Hitachi 3400N, Japan). One hundred OBs were measured to<br />

determine their mean size.<br />

The transmission electron microscopy (TEM) was carried out<br />

according to the method described by Luft (1961). Briefly, pellets of<br />

purified OBs were fixed directly in 2.5% glutaraldehyde for 2.5 h,<br />

dehydrated in a graded ethanol series (30, 50, 70, 80, 90, 100%),<br />

and then embedded in Epon-Araldite resin. Ultrathin sections were<br />

cut with ultractome, slice sizes range from 50 - 60 nm, double<br />

strained with uranyl acetate and lead citrate, and then examined<br />

with a transmission electron microscope (Hitachi JEM-1230,<br />

Japan).<br />

Preparation of D. kikuchii larvae<br />

Larvae of D. kikuchii were reared from surface-sterilized eggs.<br />

Briefly, the pupae of D. kikuchii were collected from the P.<br />

yunnanensis forest and kept in the clear cages (30 × 30 × 40 cm) in<br />

the laboratory. Once emergence, adults were provided with 50 gl -1<br />

sugar solution as food source and some fresh shoots of P. armandi<br />

as an oviposition substrate. Eggs were collected and sterilized with<br />

10% methanal for 0.5 h. After hatch, larvae were group reared on<br />

fresh needles of the P. armandi within the clear plastic jars (15 × 15<br />

× 20 cm) with some holes in the bottom until the third-instars. The<br />

above experiments were carried out at 20°C, 50-65% relative<br />

humidity and a 18:6 h (day : night) photoperiod (Guo et al., 2007).<br />

Infection studies of isolated virus against the D. kikuchii larvae<br />

The infection activity of isolated virus was tested against the thirdinstars<br />

larvae of D. kikuchii by following the method developed for<br />

Cydia pomonella granulovirus (Huber, 1981). The pellets of purified<br />

virus were diluted in distilled water. Five virus suspensions at<br />

different concentrations (2.2×10 4 , 2.2×10 5 , 2.2×10 6 , 2.2×10 7 and<br />

2.2×10 8 PIB·ml -1 ) were prepared respectively and used for this<br />

infection study. For each concentration, fifty larvae were used and<br />

300 µl of viral suspensions were sprayed onto pine needles, which<br />

were used as food of D. kikuchii larvae and filled into a plastic jar<br />

(20 × 20 × 20 cm) with some holes in the bottom. The tested larvae<br />

were kept at 20°C, 50-65% relative humidity and a 18:6 h (light :<br />

night) photoperiod. As soon as the treated needles were eaten, the<br />

tested larvae were provided with fresh, untreated needles, and<br />

continuously reared in the plastic jar. Larvae with the typical<br />

symptoms of NPV infection such as a bloated body, hanging upside<br />

down from the branches and fragile cuticle were recorded as dying<br />

of virus infection. Mortality of larvae due to the infection of virus was<br />

recorded from the third day and up to the 13th day at a one day<br />

interval. Three repetitions were carried out and as control, pine<br />

needles were treated with distilled water exactly according to the<br />

above method.<br />

Stability studies of isolated virus in toxicity and ultrastructural<br />

morphology<br />

The virus has been propagated in the fourth-instars larvae of D.<br />

kikuchii, for 4 generations. Stability of ultrastructural morphological<br />

and the virulence of the virus were tested according to the above<br />

method.<br />

Statistical analysis<br />

The median lethal time (LT50), the median lethal concentration<br />

(LC50) and the toxicity regression equations of D. kikuchii virus were<br />

estimated by probit analysis software SPSS package 17.0 (Yu and<br />

He, 2003). Death rate and corrected mortality of insect larvae were<br />

calculated based on bioassays.<br />

RESULTS<br />

Ultrastructural characteristics of D. kikuchii virus<br />

The SEM results showed that OBs of D. kikuchii are<br />

mostly polyhedral shape, 0.79~2.31 (1.64 ± 0.1) µm<br />

[minimum~maximum (mean ±SE)] in diameter (n = 100),<br />

with a lots of small holes on their surface, 173.00~254.00<br />

nm × 55.10~116.00 nm in size (Figure 2a). Ultrathin<br />

sections revealed that each OB contains many virions,<br />

252.00~359.00 nm × 70.18~200.00 nm in size (Figure


Figure 1. (a) Image showing the conifers damaged by D. kikuchii larvae; (b) OBs in D. kikuchii larvae body fluids,<br />

and for image (b) the bar represents 20 µm.<br />

Figure 2. The ultrastucture characteristics of DkNPV. (a) Scanning electron micrograph of the DkNPV OBs; (b) The<br />

ultrastructural section of OBs showing the virions (V) inside it. Each virion contains multiple nucleocapsids (N); for images a<br />

and b the bar represents 5.00 µm and 500 nm respectively.<br />

2b). The virion is rod-shaped shaped with the truncated or obtuse<br />

ends, consists of multiple nucleocapsids (up to 9) within a<br />

single viral envelope (Figure 2b). b). The size of nucleo-<br />

capsid is approximately 25.80 ± 0.86 nm (mean ±SE) in<br />

width and 265.00 ± 12.66 nm in length (n = 50) (Figure<br />

2b).<br />

Yang et al. 2263<br />

Based on the ultrastructural observation, the<br />

morphological characteristics aracteristics and the virulence of the D.<br />

kikuchii virus was identified as a new virus in<br />

nucleopolyhedrovirus of Baculoviridae, and named as<br />

DkNPV according to the universal system of virus<br />

taxonomy in the 8th ICTV report (Hong, 2006).


2264 Afr. J. Microbiol. Res.<br />

Table 1. The virulence of DkNPV on 3 rd instars larvae of D. kikuchii.<br />

Concentration (PIB·mL -1 ) Regression equation Correlation coefficient LT50(d) LC50 (PIB·mL -1 )<br />

2.2×10 8 Y=10.457x+9.219 0.976 6.89 1.72×10 5<br />

2.2×10 7 Y=10.051x+8.156 0.986 7.18 —<br />

2.2×10 6 Y=8.633x+5.487 0.947 8.16 —<br />

2.2×10 5 Y=6.536x+2.237 0.980 10.31 —<br />

2.2×10 4 Y=6.039x+0.890 0.988 11.13 —<br />

Table 2. The difference between DkNPV and DKMNPV.<br />

Virus name OBs surface Both ends of rod-shape<br />

The size of nucleocapsids<br />

(nm)<br />

Diameter<br />

(µm)<br />

LC50<br />

(PIB·mL -1 )<br />

DkNPV Lots of small holes Truncate or obtuse ends 265.00 ± 12.66 × 25.80 ± 0.86 0.79~2.31 1.72×10 5<br />

DKMNPV The even surface Obtuse both ends only 410 ± 10 × 40 ± 3 1.3~3.0 1×10 5.9<br />

Infection studies of DkNPV against the D. kikuchii<br />

larvae<br />

The results of infection study showed that DkNPV could<br />

infect and kill the third-larvae of D. kikuchii (Table 1). LC50<br />

was 1.72 × 10 5 OBs·ml -1 . At the tested five concentrations<br />

(from 2.2 × 10 8 to 2.2 × 10 4 PIB·ml -1 ), LT50 were 6.89,<br />

7.18, 8.16, 10.31 and 11.13 days, respectively (Table 1).<br />

A large number of D. kikuchii larvae begin to die from the<br />

seven day of infection, and peak mortality from 7 to 10<br />

days. A large number of virus was found in D. kikuchii<br />

larvae with the typical symptoms of NPV infection by<br />

optical microscope (Figure 1b). Therefore, DkNPV is<br />

virulent to the third-instar larvae of D. kikuchii.<br />

Our research demonstrates that the virus has been<br />

propagated 4 generations and the fourth-instars larvae of<br />

D. kikuchii has stable ultrastructural morphological and<br />

virulence.<br />

DISCUSSION<br />

In present study, a nucleopolyhedrovirus was isolated<br />

from D. kikuchii. The main morphological characteristics<br />

of this virus is consistent with the NPVs. When it was<br />

compared with the reported D. kikuchii virus DKMNPV,<br />

several significant differences were observed between<br />

them (Table 2). It is interesting that lots of small holes<br />

were observed on the surface of DkNPV. Firstly, we<br />

thought they were caused by the process of sample<br />

preparation. Other four samples (HcNPV, BusuNPV,<br />

DnNPV and CzNPV) therefore were prepared exactly<br />

according to the same method. However, no holes were<br />

observed on their OBs surface. One difference lied in<br />

small holes on the surface of DkNPV and the even<br />

surface of DKMNPV. The second difference existed on<br />

the both ends of rod-shape virions. DkNPV had truncate<br />

or obtuse in both ends, but the DKMNPV had obtuse both<br />

ends only. The third difference was that the size of<br />

nucleocapsids of DkNPV was smaller than DKMNPV<br />

(265.00 ± 12.66 nm × 25.80 ± 0.86 nm of DkNPV, 410 ±<br />

10 nm × 40 ± 3 nm of DKMNPV). Besides, the diameter<br />

of OBs differed widely, the diameter of DKMNPV was<br />

1.3~3 µm, but the diameter of DkNPV was 0.79~2.31 µm.<br />

The shape and size of OBs, the size of nucleocapsids<br />

and the features of both ends of visions were the<br />

important basis for the identification of a baculoviruses<br />

(Xie and Hu, 2000). The fourth difference shows the<br />

DkNPV is more virulent than DKMNPV against the larvae<br />

of D. kikuchii. Based on the morphological characteristics,<br />

the virulence of this virus and the difference between<br />

DkNPV and DKMNPV, it was proposed as a new strain<br />

nuclepoyhedrovirus of D. kikuchii and named as DkNPV.<br />

In the next step, the structure of its nucleic acid and<br />

biological characteristics will be further studied to confirm<br />

its identification. Dendrolimus cytoplasmic polyhedrosis<br />

virus (CPV) were used to biologically control the pest of<br />

Dendrolimus in China since the first DpCPV (D.<br />

punctatus Cytoplasmic Polyhedrosis virus) was isolated<br />

in China in 1973. However, as is well known the time<br />

required for CPVs to kill the insect larvae was longer than<br />

that for NPVs obviously. The new isolated DkNPV was<br />

virulent for the third-instar larvae of D. kikuchii and<br />

exhibited high infection activity (Table 1). Therefore, this<br />

virus was very precious for the biological control of D.<br />

kikuchii.<br />

ACKNOWLEDGMENTS<br />

We sincerely thank Yan-Li Duan and Jian-Jie Xue for<br />

assistance both in field research and virus propagation;<br />

Chun-Yan Wang for suggestions of revising the<br />

manuscript. This work was supported financially by<br />

National Natural Science Foundation of China (No.<br />

30671688), and Forestry Industry Special Foundation of


China (No. 200904029; No. 200704003).<br />

REFERENCES<br />

Chen CJ (1990). Integrated Management pine Caterpillars. Beijing,<br />

China Forestry Publishing House Press, pp. 201-210.<br />

EI-Salamouny S (1998). Increasing efficacy of nucleopolyhedroviruses<br />

and a comparative study of an Egyptian isolate. PhD dissertation,<br />

Faculty of Agriculture, Cairo University, Egypt, pp. 216-233.<br />

Fuxa JR (1989). Fate of released entomopathogens with reference to<br />

risk assessment of genetically engineered microorganisms, Bull.<br />

Entomol. Soc. Am., 35: 122-124.<br />

Guo HF, Fang JC, Liu BS, Wang JP, Zhong WF (2007). Enhancement<br />

of the biological activity of nucleopolyhedrovirus through disruption of<br />

the peritrophic matrix of insect larvae by chlorfluazuron. Pest<br />

Manage. Sci., 63: 68-74.<br />

Heinz KM, McCutchen BF, Herrmann R, Parrella MP, Hammock BD<br />

(1995). Direct effects of recombinant nuclear polyhedrosis viruses on<br />

selected non-target organisms. J. Econ. Entomol., 88: 259-246.<br />

Yang et al. 2265<br />

Hong J (2006). The Universal System of Virus Taxonomy in the 8th<br />

ICTV Report. Virol. Sin., 21(1): 84-96.<br />

Huber J (1981). Apfelwickler granulosevirus: production and Biotests.<br />

Mitt. Dtsch. Ges. Allg. Angew. Ent., 2: 141-145.<br />

Laird M, Lacey LA, Davidson EW (1990). Safety of Microbial<br />

Insecticides. Boca Raton. FL. USA, CRC Press, pp. 76-80.<br />

Luft JH (1961). Improvements in epoxy resin embedding methods. J.<br />

Biophys. Biochem. Cytol., 9: 409-414.<br />

Xie TE, Hu ZH (2000). General Virology. Beijing, Science Press. pp.<br />

119-120.<br />

Yang DZ, Wang YX, Li YQ (1986). Studies on the nuclear polyhedrosisvirus<br />

disease of simao pine caterpil-lcar infected by Dendrolimus<br />

kikuchii Matsumura (Lepidoptera: Lasiocampidae). J. Yunnan Univ.,<br />

1: 105-110.<br />

Yasuhisa K (2007). Current status and prospects on microbial control in<br />

Japan. J. Invertebr. Pathol., 95: 181–186.<br />

Yu JY, He XH (2003). Data statistical analysis and SPSS applications.<br />

Beijing, People's Post and Telecommunication Publishing House<br />

Press, pp. 178-182.


African Journal of Microbiology Research Vol. 5(16), pp. 2266-2272, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Characterization of Staphylococcus spp strains<br />

isolated from hospital, community and environmental in<br />

Puebla city, Mexico<br />

Ana Marta de los Ángeles Lobo-Sánchez 1 , Patricia Lozano-Zaraín 2 ,<br />

Ygnacio Martínez-Laguna 1,2 , Zita Gutiérrez-Cázarez 3 , Antonio Rivera-Tapia 2 , Carmen Torres 4<br />

and Rosa del Carmen Rocha-Gracia 1,2 *<br />

1 Posgrado en Ciencias Ambientales, Área de la Salud, Centro de Investigaciones en Ciencias Microbiológicas, Instituto<br />

de Ciencias, Benemérita Universidad Autónoma de Puebla. Complejo de Ciencias, edificio No. 103 J. Ciudad<br />

Universitaria, colonia San Manuel, Puebla 72570, México.<br />

2 Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de<br />

Puebla. Complejo de Ciencias, edificio No. 103 J. Ciudad Universitaria, colonia San Manuel, Puebla 72570, México.<br />

3 Hospital para el Niño Poblano, Blvd. del Niño Poblano No. 5207 Col. Concepción la Cruz, 72190 Puebla, Pue., México.<br />

4 Área Bioquímica y Biología Molecular, Universidad de la Rioja, Logroño, Spain.<br />

Accepted 30 July, 2011<br />

We investigated the prevalence of methicillin-resistant staphylococci (MRS) and evaluated the<br />

antimicrobial resistance patterns of 284 Staphylococcus strains isolated from two hospitals, as well as<br />

from community and the environment in Puebla City, Mexico. Isolates were identified by Gram-stain and<br />

biochemical reactions and antimicrobial susceptibility testing was performed by Kirby-Bauer and in<br />

some cases by determination of minimal inhibitory concentrations of antimicrobial agents. Result<br />

showed that from 284 strains studied, 32% (90 strains) were multidrug-resistant, of which 82% (74<br />

strains) were methicillin-resistant. From 154 Staphylococcus aureus isolates tested, 41 strains showed<br />

methicillin-resistance (27%) and 84 of 130 coagulase-negative staphylococci showed methicillinresistance<br />

(65%). The mecA gene was detected in 38 of 43 oxacillin-resistant staphylococci tested<br />

(14/14 S. aureus and 24/29 coagulase negative staphylococci). Nevertheless, this gene was also<br />

identified in 14 S. aureus that exhibited oxacillin and cefoxitin susceptibility. The percentages of<br />

resistance detected among S. aureus and coagulase negative staphylococcal isolates were: penicillin<br />

(79 and 67%, respectively), oxacillin (27 and 65%), erythromycin (18 and 36%), tetracycline (6 and 24%),<br />

gentamicin (71 and 45%), and trimethoprim sulfamethoxazole (19 and 37%). The �-lactamase production<br />

was positive in more than 80% of isolates. These results show the presence of multiresistant strains in<br />

these three sources, which supports the control measures taken by health authorities with respect to<br />

avoiding the misuse and abuse of antibiotics.<br />

Key words: Staphylococcus, methicillin resistance Staphylococcus aureus (MRSA), methicillin resistance<br />

coagulase-negative staphylococci (MRCoNS), emerging infections.<br />

INTRODUCTION<br />

In the 60's, Staphylococcus aureus was identified as the<br />

*Corresponding author. E-mail: rochagra@yahoo.com. Tel: 00<br />

(52) 2222295500 ext 2546.<br />

main cause of nosocomial infections in the world (Klimek<br />

et al., 1976; Crossley et al., 1979; Doebbeling, 1995). At<br />

present, the methicillin-resistant Staphylococcus (MRS)<br />

and multiresistant Staphylococcus are the leading cause<br />

of infections in hospitals and community (Schaberg et al.,<br />

1991; Ponce de León, 1996; Diekema et al., 2000; Wu et


al., 2006; Martins et al., 2007; David and Daum, 2010).<br />

Resistance to methicillin is due to the presence of<br />

Staphylococal Chromosomal Cassette mec (SCCmec)<br />

which in addition to carrying mecA gene that encodes a<br />

penicillin-binding protein (PBP2a) (Hartman and Tomasz,<br />

1984; De Lencastre et al., 1994; Pinho et al., 2001), also<br />

can contains genes of resistance for non-beta-lactam<br />

agents, causing multidrug resistant strains (Katayama et<br />

al., 2000; Hiramatsu et al., 2001).<br />

It is reported that S. aureus is the causal agent of more<br />

than 20% of bacteremias in USA, Canada and Latin<br />

America (Pfaller et al., 1998; 1999; Diekema et al., 2000;<br />

Wisplinghoff et al., 2003; Chen et al., 2010), skin<br />

infections and soft tissue (Gales et al., 2009; Doern et al.,<br />

1999) and pneumonia (Sader et al., 1998; Gales et al.,<br />

2009). Coagulase-negative staphylococci (CoNS) are the<br />

third most common type of isolates in bacteremias<br />

(Schaberg et al., 1991; Edmond et al., 1999; Diekema et<br />

al., 2001; Wisplinghoff et al., 2003; Chen et al., 2010,<br />

Rosa et al., 2009). In the environment, staphylococci are<br />

the most frequently isolated bacteria in both open and<br />

closed areas (Górny et al., 1999; Górny and Dutkiewicz,<br />

2002; Tsai and Macher, 2005; Eames et al., 2009; Tang,<br />

2009).<br />

The emergence of multidrug-resistant staphylococci,<br />

prompted a warning from the health centers and<br />

organizations, because epidemiological changes, clinical<br />

manifestations and their control can become a significant<br />

Public Health problem in both developed and developing<br />

countries. In Latin America, it was reported a prevalence<br />

of 40% of methicillin resistant Staphylococcus aureus<br />

(MRSA) and over 80% methicillin resistant coagulase<br />

negative Staphylococcus (MRCoNS) and most of these<br />

strains were multidrug resistant (Sader et al., 2009). The<br />

World Health Organization (WHO) has seen the<br />

emergence and spread of antimicrobial resistance as a<br />

priority issue and therefore since September 2001<br />

established a global measure for the containment of<br />

antimicrobial resistance, which includes as a fundamental<br />

measure the surveillance of antimicrobial resistance<br />

caused among other factors by self-medication (WHO,<br />

2002). In Mexico, a law was issued in which the<br />

guidelines to which the sale and dispensing of antibiotics<br />

as a preventive measure to misuse of these drugs would<br />

be subjected (Official Gazette, 2010).<br />

The aim of this study was to determine the methicillin<br />

resistance and other associated resistances of<br />

staphylococci strains isolated from two general hospitals,<br />

the community and the environment of the city of Puebla,<br />

Mexico.<br />

MATERIALS AND METHODS<br />

Bacterial isolates<br />

284 staphylococci were included in this study. One-hundred-andseventeen<br />

strains were obtained in two hospitals of Puebla, México<br />

(Pediatric Hospital in 2003; University Hospital of Puebla City in<br />

Lobo-Sánchez et al. 2267<br />

1998-2000) and they were collected from wound and articulation<br />

secretions, sepsis, pleural and cerebrospinal fluids, peritoneal<br />

dialysis fluids, catheter tips, post-mortem and throat swabs. Onehundred-and-twelve<br />

strains were obtained from community during<br />

2002-2003 and they were collected from outpatients with clinically<br />

reported staph infections (vulvar swabs, throat, skin and conjunctiva)<br />

as well as from hospital personnel and community carriers by<br />

means of throat swabs. By other side, 55 strains from the environment<br />

were collected in 2004, through the Gravity Sedimentation<br />

Technique (Frankland and Hart, 1887); specifically, blood agar and<br />

mannitol salt agar plates, were exposed for a period of approximately<br />

15 and 45 minutes respectively, at a height of between 1.5<br />

and 2.0 m (Lighthart and Shaffer, 1995; De la Rosa and Ullán,<br />

2002; Rosas et al., 2004). Staphylococcus strains were identified by<br />

standard methods, such as colonial morphology, Gram-stain,<br />

catalase, coagulase, clumping factor, urease, DNase, hemolysis on<br />

blood agar and bacitracin test (Finegold and Baron, 1996).<br />

Antimicrobial susceptibility test and �-lactamase production<br />

All obtained staphylococci were tested by Kirby-Bauer test (CLSI,<br />

2008) for susceptibility to the following antimicrobials oxacillin (OX,<br />

(1 �g/disk), penicillin (PE, 10 U), erythromycin (E, 15 �g),<br />

tetracycline (TE, 30 �g), gentamicin (GE, 10 �g), vancomycin (VA,<br />

30 �g) and trimethoprim sulfamethoxazole (SXT, 25 �g) (Sanofi<br />

Diagnostics Pasteur, SA, Mexico City, Mexico). Mueller-Hinton agar<br />

(Oxoid, Cambridge, England) supplemented with 4% NaCl was<br />

used in these assays. The susceptibility of cefoxitin (FOX, 30<br />

�g/disk) (Difco Laboratories, Detroit, MI, USA) was checked in a<br />

group of mecA-positive strains (n=14) that appeared as oxacillinsusceptible.<br />

Additionally, a minimum inhibitory concentration (MIC)<br />

test of oxacillin using the agar dilution method with Steers<br />

Replicator was performed for 90 staphylococci isolated from<br />

different origins (30 from each source). The results of disk diffusion<br />

tests and MICs were interpreted according to CLSI criteria (CLSI,<br />

2008).<br />

The �-lactamase production was performed by Cefinase disks<br />

(Becton Dickinson) according to manufacturer's instructions.<br />

mecA gene amplification by polymerase chain reaction (PCR)<br />

Specific PCR of the mecA gene was performed in 90 staphylococci<br />

from different origins that were also analyzed by MIC. Because the<br />

content of A + T in mecA gene is high (70%) and in order to<br />

minimize the amplification of DNA regions not related to the gene<br />

(Ubukata et al., 1990; Ryffel et al., 1990; Unal et al., 1992), we<br />

chose two sets of primers combined in two reactions, the sense<br />

primer P1, 5'-(911) GGTCCCATTAACTCTGAAG (929)-3' and<br />

antisense P3, 5'-(1956) AGTTCTGCAGTACCGGATTTGC (1935)-3'<br />

(Petinaki et al., 2001) and a pair designed in this study, MMecAF<br />

sense primer, 5'-(539) TCCAGAATGCAGAAAGACC (558)-3'and<br />

antisense MMecAR, 5'-(1076) TGTATGTGCGATTGTATTGCTATT<br />

(1053)-3', giving rise to PCR products of 1046 and 538 bp<br />

respectively.<br />

RESULTS<br />

From 284 isolates studied, 154 of them were identified as<br />

S. aureus (54%), isolated mostly from hospital and<br />

community sources and 130 isolates (46%) were CoNS,<br />

mainly obtained from environment and hospital sources.<br />

Methicillin-resistance in this study was firstly checked by<br />

the study of the susceptibility to oxacillin. Table 1 shows


2268 Afr. J. Microbiol. Res.<br />

Table 1. Species distribution and methicillin resistance (MR) in Staphylococcus strains from three sources analyzed.<br />

Origin<br />

(n isolates)<br />

Studied isolates<br />

a Methicillin-resistance<br />

OXA/FOX-susceptible<br />

mecA-positive S. aureus<br />

S. aureus CoSCN S. aureus (%) CoSCN (%)<br />

Hospital (117) 71 46 24 (34) 36 (78) 5<br />

Community (112) 80 32 17 (21) 15 (47) 6<br />

Environmental (55) 3 52 0 33 (63) 3<br />

Total: 284 154 (54%) 130 (46%) 41 (27) 84 (65) 14<br />

CoSCN: coagulase negative staphylococci; OXA: oxacillin; FOX: cefoxitin<br />

a Isolates included were those that presented oxacillin resistance (by Kirby Bauer or MIC).<br />

Figure 1. mecA gene detection by PCR, amplified in agarose gel and subjected to electrophoresis. A. P1F-P3R<br />

primers (Petinaki et al., 2001) to amplify 1046 bp. Line 1: Marker �X174; 2, 3-5, 7: mecA positive strains; 6: mecA<br />

negative strain and 8: ATCC 25923 (negative control). B. MmecAF-MmecAR primers to amplify 538 bp. Line 1: Marker<br />

�X174; 2, 3-8, 10 and 11: mecA positive strains; 9, 12 and 13: mecA negative strains and 14: ATCC 25923 (negative<br />

control).<br />

the percentage of methicillin resistance among our<br />

isolates using this criterium. Forty-one S. aureus isolates<br />

of the 154 tested showed methicillin-resistance (27%),<br />

and isolates of hospital origin showed higher percentage<br />

of resistance than those of the community (34 and 21%,<br />

respectively). On the other hand, 84 of 130 coagulasenegative<br />

staphylococci showed methicillin-resistance<br />

(65%) and percentages were higher among hospital and<br />

environmental isolates (78 and 63%, respectively) (Table<br />

1).<br />

The presence of mecA gene was analyzed in a group<br />

of 90 staphylococci that included isolates of the three<br />

sources and also isolates with different oxacillin susceptibilities<br />

(Figure 1). The mecA gene was detected in 38 of<br />

43 oxacillin-resistant staphylococci tested (14/14 S.<br />

aureus and 24/29 coagulase negative staphylococci, data<br />

not showed). Nevertheless, this gene was also identified<br />

in 14 S. aureus that exhibited oxacillin and cefoxitin<br />

susceptibility, observing a phenotypic-genotypic discrepancy<br />

(Table 1). From 90 selected strains and tested by<br />

�-lactamase production it was found that 100% hospital<br />

isolates, 83% community isolates and 93% environmental<br />

isolates were positive.<br />

Figure 2 shows the percentages of resistance to<br />

different antimicrobial agents of S. aureus and<br />

coagulase-negative staphylococci. All strains tested were<br />

resistant to two or more agents and it was considered as<br />

multidrug-resistant strains those that were resistant to<br />

more than four antibiotics. From 284 strains studied, 32%<br />

(90 strains) were multidrug-resistant, of which 82% (74<br />

strains) were methicillin-resistant. The percentages of<br />

resistance detected among S. aureus and coagulase<br />

negative staphylococcal isolates were: penicillin (79 and<br />

67%, respectively), oxacillin (27 and 65%), erythromycin<br />

(18 and 36%), tetracycline (6 and 24%), gentamicin (71<br />

and 45%), and trimethoprim sulfamethoxazole (19 and<br />

37%).<br />

DISCUSSION<br />

As expected, the percentages of methicillin resistance<br />

detected among S. aureus and CoSCN in this study were<br />

higher in the hospital isolates in relation to those of the


Figure 2. Resistance staphylococci strains isolated from Hospital, Community and<br />

Environmental by Kirby-Bauer test. PE: penicillin; OX: oxacillin; E: erythromycin; TE:<br />

tetracycline; GE: gentamicin; SXT: trimethoprim-sulfamethoxazole and VA: vancomycin.<br />

RSA, Resistant Staphylococcus aureus strains; RCoNS, Resistant Coagulase Negative<br />

Staphylococcus strains. Intermediate Resistance: a GE SA 8% and CoNS 6%; b TE CoNS 2%<br />

and c SXT 1%.<br />

community. Hospital isolates presented high percentages<br />

of resistance (34 and 78% for S. aureus and CoSCN,<br />

respectively), what indicate that this type of resistant<br />

microorganisms can be a problem in the hospitals tested.<br />

These percentages are similar to other data previously<br />

published for Mexican hospitals and in other Latin<br />

American countries (Guzmán-Blanco et al., 2009).<br />

The results of this study show that in our community<br />

Staphylococcus is a major concern for public health as a<br />

cause of emerging infections, due to the presence of<br />

multiresistant strains with ability to survive and spread.<br />

Among 284 strains collected in this study, the highest<br />

percentage was S. aureus isolated from community,<br />

which is consistent with that reported by the Centers for<br />

disease control and prevention (CDC) in USA (Kuehnert<br />

et al., 2006) and the Instituto Mexicano del Seguro Social<br />

(IMSS) in México (García-Contreras et al., 2000). The<br />

SCoN were isolated mostly from environmental sources,<br />

probably because they are found in greater numbers in<br />

skin and mucous membranes of both human animals as<br />

compared with S. aureus (Bischoff et al., 2004; 2007).<br />

The resistance rates observed in this study agree with<br />

those reported in other regions (Diekema et al., 2001;<br />

Cuevas et al., 2004; Gales et al., 2009; Sader et al.,<br />

2009) and may be related to selective pressure to which<br />

Lobo-Sánchez et al. 2269<br />

bacteria are subjected in our community, because in the<br />

hospitals the committee epidemiological surveillance sets<br />

that the treatment of staphylococcal infections begins<br />

with �-lactam antibiotics, which in some cases, antimicrobial<br />

therapy of choice is subject to the standard antibiotic<br />

treatments available or unfinished treatments due to lack<br />

of resources of both health institutions and patients. In<br />

community, easy access to this type of antibiotics until<br />

2010 year (Official Gazette, 2010) allowed the selfmedication,<br />

resulting in inadequate doses or unfinished<br />

treatments (Dreser et al., 2008), favoring the spread of<br />

multiresistant strains in the community.<br />

The GE resistance above 45%, consistent with that<br />

reported in 2005 by Public Health in Mexico (Benavides-<br />

Plascencia et al., 2005), where GE is among the seven<br />

antibiotics account for 80% of the observed resistance. In<br />

recent years, glycopeptides have been the mainstay of<br />

treatment of infections due to MRS. However, the isolation<br />

of CoNS with decreased susceptibility (Schwalbe et<br />

al., 1987), the report heteroresistant S. aureus strains<br />

(Hiramatsu et al., 1997) and the presence of strains<br />

resistant to VA (CDC, 2002; Palazzo et al., 2005; Tiwari<br />

and Sen, 2006), led that the treatment options for<br />

infections by MRS are committed and required of new<br />

antimicrobial agents (Appelbaum, 2006; Menezes et al.,


2270 Afr. J. Microbiol. Res.<br />

2008). In this paper, strains with intermediate susceptibility<br />

to VA were not identified however; the results<br />

emphasize the need for continuous monitoring of the<br />

levels of MIC to this antibiotic in the MRS, since these<br />

results alert of the risks and implications of the indiscriminate<br />

use of this drug. The E and SXT has been widely<br />

used in the treatment of staphylococcal infections<br />

(Huovinen et al., 1995; Schlegelová et al., 2002),<br />

however, clinical experience has shown that resistance to<br />

both drugs has developed worldwide (Huovinen, 2001;<br />

Gales et al., 2009; Sader et al., 2009). The TE is a<br />

relatively cheap antibiotic, has therefore been widely<br />

used in prophylaxis, treatment of infections and as animal<br />

growth promoter, so the selective pressure for its use,<br />

has led to resistant bacterial strains (Committee on Drug<br />

Use in Food Animals, 1999; Chopra and Roberts, 2001).<br />

The worldwide prevalence of resistance to TE in S.<br />

aureus is higher than CoNS (Diekema et al., 2001; Gales<br />

et al., 2009). However, in Spain was reported a<br />

resistance less than 5% in S. aureus and between 5%<br />

and 15% in CoNS (Pérez-Trallero and Iglesias, 2003).<br />

The TE resistance observed in this study is relatively low 6%<br />

for S. aureus and 24% for CoNS, which could be related<br />

to the TE is one of the lowest drug consumption in<br />

Mexico (Benavides-Plascencia et al., 2005) and is not<br />

considered a treatment of choice for serious<br />

staphylococcal infections (WHO, 1999). It is interesting<br />

intermediate susceptibility to GE, TE and SXT, while not<br />

reaching high levels, confirming the need for selection<br />

and rational use of these antibiotics.<br />

The MRSA and MRCoNS mecA positive showed<br />

uniformity in the expression of resistance (Chambers,<br />

1988), so that might be considered strains with homogeneous<br />

resistant (Weller, 1999). In the MSSA-mecA<br />

positive, the results confirmed with FOX could coincide<br />

with that reported by Hososaka et al. (2007), so the<br />

absence of the phenotypic expression of resistance in<br />

these strains, suggests new molecular targets that could<br />

be related to susceptibility to ME (Rohrer et al., 2003), or<br />

the presence of mutations in the fem genes, which are<br />

essential factors for ME resistance expression<br />

(Chambers, 1997), contributing to low levels of resistant<br />

to this antibiotic, without any alteration in the mecA gene<br />

(Giannouli et al., 2010). In MRCoNS-mecA negative, the<br />

expression of resistance to ME, could be related to<br />

different mechanisms for the production of PBP2a<br />

(McDougal and Thornsberry, 1986; Tomasz et al., 1989;<br />

Suzuki et al., 1993; Weller, 1999), resulting in the<br />

presence of extremely heteroresistant strains (Chambers,<br />

1988). The MSCoNS-mecA positive strains reported as<br />

MR by test MIC, could be pre-MRCoNS (Hiramatsu,<br />

1995), because mutations in regulatory genes of mecA<br />

would originate phenotypically methicillin resistant strains<br />

(Suzuki et al., 1993; Kobayashi et al., 1996; 1998).<br />

In most of the strains tested the �-lactamase production<br />

was detected, suggesting that the regulatory genes blaR1<br />

and blaI could also be related to the phenotypic<br />

expression of resistance (Cohen et al., 1972; Boyce et<br />

al., 1990; Hiramatsu et al., 1990; Ryffel et al., 1992;<br />

Hackbarth and Chambers, 1993; Hackbarth et al., 1994;<br />

Chambers, 1997; Rosato et al., 2003).<br />

Finally, the results of this study confirm the presence of<br />

MRS strains, which are also multiresistant, whose<br />

mechanisms of resistance may not only be related to the<br />

interaction of the mec genes, but with much more<br />

complex regulatory mechanisms, so its worth conducting<br />

more detailed studies and further epidemiological<br />

surveillance of Staphylococcus strains isolated from<br />

different sources, to show the behavior over time of<br />

resistance in these strains, based on the measure<br />

introduced from August 2010 on the sale of antibiotics in<br />

pharmacies throughout Mexico through prescription<br />

(Official Gazette, 2010).<br />

ACKNOWLEDGEMENT<br />

Ana Marta de los Ángeles Lobo Sánchez was supported<br />

by a student scholarship from Consejo Nacional de<br />

Ciencia y Tecnología, México (CONACYT 174306).<br />

REFERENCES<br />

Appelbaum PC (2006). The emergence of vancomycin-intermediate and<br />

vancomycin-resistant Staphylococcus aureus. Clin. Microbiol. Infect.<br />

Dis., 12(1): 16-23.<br />

Benavides-Plascencia L, Aldama-Ojeda AL, Vázquez HJ (2005).<br />

Surveillance of antibiotic utilization and bacterial resistance profiles in<br />

tertiary level hospitals in Mexico City. Salud Publica Mex., 47(3): 219-<br />

226.<br />

Bischoff WE, Bassetti F, Bassetti-Wyss BA, Wallis ML, Tucker BK,<br />

Reboussin BA, D’Agostino RB, Pfaller MA, Gwaltney JM, Sherertz RJ<br />

(2004). Airborne dispersal as a novel transmission Route of<br />

coagulase-negative staphylococci: Interaction between coagulasenegative<br />

staphylococci and rhinovirus infection. Infect. Control. Hosp.<br />

Epidemiol., 25(6): 504-511.<br />

Bischoff WE, Tucker BK, Wallis ML, Reboussin BA, Pfaller MA, Hayden<br />

FG, Sherertz RJ (2007). Preventing the Airborne Spread of<br />

Staphylococcus aureus by Persons with the Common Cold: Effect of<br />

Surgical Scrubs, Gowns, and Masks. Infect. Control Hosp.<br />

Epidemiol., 28(10): 1148-1154.<br />

Boyce JM, Medeiros AA, Papa EF, O’Gara CJ (1990). Introduction of<br />

beta-lactamase and methicillin resistance in unusual strains of<br />

methicillin-resistant Staphylococcus aureus. J. Antimicrob.<br />

Chemother., 25(1): 73-81.<br />

Centers for Disease Control and Prevention (CDC) (2002).<br />

Staphylococcus aureus resistant to vancomycin, United States. Morb<br />

Mortal Wkly Rep (MMWR), 51(26): 565-567.<br />

Chambers HF (1988). Methicillin-Resistant staphylococci. Clin.<br />

Microbiol. Rev., 1(2): 173-186<br />

Chambers HF (1997). Methicillin resistance in staphylococci: molecular<br />

and biochemical basis and clinical implications. Clin. Microbiol. Rev.,<br />

10(4): 781-791.<br />

Chen CY, Tsay W, Tang JL, Tien HF, Chen YC, Chang, SC, Hsueh PR<br />

(2010). Epidemiology of bloodstream infections in patients with<br />

haematological malignancies with and without neutropenia.<br />

Epidemiol. Infec., 138(7): 1044-1051<br />

Chopra I, Roberts M (2001). Tetracycline Antibiotics: Mode of Action,<br />

Applications, Molecular Biology, and Epidemiology of Bacterial<br />

Resistance. Microbiol. Molec. Biol. Rev., 65(2): 232-260.<br />

Clinical and Laboratory Standards Institute (CLSI). 2008. Performance<br />

Standards for Antimicrobial Disk Susceptibility Test. Approved<br />

Standard. Document M100-S18. NCCLS, Wayne, PA.<br />

Cohen SC, Gibson J, Sweeney HM (1972). Phenotypic suppression of


methicillin resistance in Staphylococcus aureus by mutant noninducible<br />

penicillinase plasmids. J. Bacteriol., 112(2): 682-689<br />

Committee on Drug Use in Food Animals (1999). Food-Animal<br />

Production Practices and drug use. In: Committee on Drug Use in<br />

Food Animals, Panel on Animal Health, Food Safety, and Public<br />

Health, National Research Council (eds), The use of drugs in food<br />

animals, benefits and risks, National Academy Press, Washington,<br />

D.C., pp. 27-68.<br />

Crossley K, Loesch D, Landesman B, Mead K, Chern M, Strate R<br />

(1979). An outbreak of infections caused by strains of<br />

Staphylococcus aureus resistant to methicillin and aminoglycosides I.<br />

Clinical studies. J. Infect. Dis., 139(3): 273-279.<br />

Cuevas O, Cercenado E, Videl A, Guinea J, Sánchez-Conde M,<br />

Sánchez-Somolinos M, Bouza E (2004). The Spanish Group for the<br />

Study of Staphylococcus. Evolution of the Antimicrobial Resistance<br />

of Staphylococcus spp. in Spain: Five Nationwide Prevalence<br />

Studies, 1986 to 2002. Antimicrob. Agents Chemother., 48(11): 4240-<br />

4245.<br />

David MZ, Daum RS (2010). Community-Associated Methicillin-<br />

Resistant Staphylococcus aureus: Epidemiology and Clinical<br />

Consequences of an Emerging Epidemic. Clin. Microbiol. Rev., 23(3):<br />

616-687.<br />

De la Rosa MAM, Ullán C (2002). Air: Habitat and mode of<br />

microorganism’s transmission. Rev. Observ. Medioamb., 5: 375-402.<br />

De Lencastre H, De Jonge BL, Matthews PR, Tomasz A (1994).<br />

Molecular aspects of methicillin resistance in Staphylococcus aureus.<br />

J. Antimicrob. Chemother., 33(1): 7-24.<br />

Official Gazette (2010). SECRETARÍA DE GOBERNACIÓN (SEGOB).<br />

José Ángel Córdova Villalobos, Secretary of Health. Law that<br />

determination of guidelines to which it subjects the sale and<br />

dispensation of antibiotics. Date Publication law in the Official<br />

Gazette May 27, 2010. Mexico, D.F.<br />

Diekema DJ, Pfaller MA, Jones RN (2000). Trends in antimicrobial<br />

susceptibility testing of bacterial pathogens isolated from patients<br />

with bloodstream infections in the USA, Canada and Latin America:<br />

report from the SENTRY Antimicrobial Surveillance Program, 1998.<br />

Int. J. Antimicrob. Agents, 13(4): 257-271.<br />

Diekema DJ, Pfaller MA, Schmitz FJ, Smayevsky J, Bell J, Jones RN,<br />

Beach M (2001). Survey of Infections Due to Staphylococcus<br />

Species: Frequency of Occurrence and Antimicrobial Susceptibility of<br />

Isolates Collected in the United States, Canada, Latin America,<br />

Europe, and the Western Pacific Region for the SENTRY<br />

Antimicrobial Surveillance Program, 1997–1999. Clin. Infect. Dis.,<br />

32(2): S114-S132.<br />

Doebbeling BN (1995). The epidemiology of methicillin-resistant<br />

Staphylococcus aureus infection and colonization. J. Chemother.,<br />

7(3): 99-103.<br />

Doern GV, Jones RN, Pfaller MA, Kugler KC, Beach ML (1999).<br />

Bacterial pathogens isolated from patients with skin and soft tissue<br />

infections: frequency of occurrence and antimicrobial susceptibility<br />

patterns from the SENTRY Antimicrobial Surveillance Program<br />

(United States and Canada, 1997). SENTRY Study Group (North<br />

America). Diagn. Microbiol. Infect. Dis., 34(1): 65-72.<br />

Dreser A, Wirtz VJ, Corbett KK, Echániz G (2008). Use of antibiotics in<br />

Mexico: review of problems and policies. Salud Publica Mex., 50(4):<br />

S480-S487.<br />

Eames I, Tang JW, Li Y, Wilson P (2009). Airborne transmission of<br />

disease in hospital. J. R Soc. Interface, 6(6): S697-S702.<br />

Edmond MB, Wallace SE, McClish DK, Pfaller MA, Jones RN, Wenzel<br />

RP (1999). Nosocomial bloodstream infections in United States<br />

hospitals: a three-year analysis. Clin. Infect. Dis., 29(2): 239-244.<br />

Finegold SM, Baron EJ (1996). Identification Methods for etiologic<br />

agents of infectious diseases. In Baron EJ, Drew WL, Edelstein MAC,<br />

García LS, Roberts GD (eds) Bailey and Scott's Diagnostic<br />

Microbiology, The C. V. Mosby Company, St. Louis, Toronto,<br />

Princeton, pp. 339-340.<br />

Frankland PF, Hart TG (1887). Further experiments on the distribution<br />

of micro-organisms in air (by Hesse's method). Proc. Roy. Soc.<br />

London., 42: 267-282.<br />

Gales AC, Sader HS, Ribeiro J, Zoccoli C, Barth A, Pignatari, AC(2009).<br />

Antimicrobial Susceptibility of Gram-Positive Bacteria Isolated in<br />

Brazilian Hospitals Participating in the SENTRY Program (2005-<br />

Lobo-Sánchez et al. 2271<br />

2008). Brazilian J. Infect. Dis., 13(2): 90-98.<br />

García-Contreras F, Del Angel-García G, Ramírez-Cuenca A, Malváez-<br />

Valdes M, Vega-Yáñez A, Amato-Martínez JD (2000). Costeffectiveness<br />

study of ceftriaxone and cefotaxime for the treatment of<br />

community acquired pneumonia. Rev. Invest. Clin., 52(4): 418-426.<br />

Giannouli S, Labrou M, Kyritsis A, Ikonomidis A, Pournaras S,<br />

Stathopoulos C, Tsakris A (2010). Detection of mutations in the<br />

FemXAB protein family in oxacillin-susceptible mecA-positive<br />

Staphylococcus aureus clinical isolates. J. Antimicrob. Chemother.,<br />

65(4): 626-633.<br />

Górny RL, Dutkiewicz J (2002). Bacterial and Fungal Aerosols in Indoor<br />

Environment In Central And Eastern European Countries. Ann. Agric.<br />

Environ. Med., 9(1): 17-23.<br />

Górny RL, Dutkiewicz J, Krysińska-Traczyk E (1999). Size distribution<br />

of bacterial and fungal bioaerosols in indoor air. Ann. Agric. Environ.<br />

Med., 6(2): 105-113.<br />

Guzmán-Blanco M, Mejía C, Isturiz R, Álvarez C, Bavestrello L,<br />

Gotuzzo E, Labarca J, Luna CM, Rodríguez-Noriega E, Salles MJ,<br />

Zurita J, Seas C (2009). Epidemiology of methicillin-resistant<br />

Staphylococcus aureus (MRSA) in Latin America. Int. J. Antimicrob.<br />

Agents, 34(4): 304-308.<br />

Hackbarth CJ, Chambers HF (1993). blaI and blaR1 regulate betalactamase<br />

and PBP 2a production in methicillin-resistant<br />

Staphylococcus aureus. Antimicrob. Agents Chemoter., 37(5): 1144-<br />

1149.<br />

Hackbarth CJ, Miick C, Chambers HF (1994). Altered production of<br />

penicillin-binding protein 2a can affect phenotypic expression of<br />

methicillin resistance in Staphylococcus aureus. Antimicrob. Agents<br />

Chemother., 38(11): 2568-2571.<br />

Hartman BJ, Tomasz A (1984). Low-affinity penicillin-binding protein<br />

associated with beta-lactam resistance in Staphylococcus aureus. J.<br />

Bacteriol., 158(2): 513-516.<br />

Hiramatsu K, Suzuki E, Takayama H, Katayama Y, Yokota T (1990).<br />

Role of penicillinase plasmids in stability of the mecA gene in<br />

methicillin-resistant Staphylococcus aureus. Antimicrob. Agents<br />

Chemother., 34(4): 600-604.<br />

Hiramatsu K (1995). Molecular evolution of MRSA. Microbiol. Immunol.,<br />

39(8): 531-543.<br />

Hiramatsu K, Aritaka N, Hanaki H, Kawasaki S, Hosoda Y, Hori S,<br />

Fukuchi Y, Kobayashi I (1997). Dissemination in Japanese hospitals<br />

of strains of heterogeneously resistant to vancomycin. Lancet,<br />

350(9092): 1670-1673.<br />

Hiramatsu K, Cui L, Kuroda M, Ito T (2001). The emergence and<br />

evolution of methicillin-resistant Staphylococcus aureus. Trends.<br />

Microbiol., 9(10): 486-493.<br />

Hososaka Y, Hanaki H, Endo H, Suzuki Y, Nagasawa Z, Otsuka Y,<br />

Nakae T, Sunakawa K (2007). Characterization of oxacillinsusceptible<br />

mecA-positive Staphylococcus aureus: a new type of<br />

MRSA. J. Infect. Chemother., 13(2):79-86.<br />

Huovinen P, Sundström L, Swedberg G, Sköld O (1995). Trimethoprim<br />

and sulfonamide resistance. Antimicrob. Agents Chemother., 39(2):<br />

279-289.<br />

Huovinen P (2001). Resistance to Trimethoprim-Sulfamethoxazole.<br />

Clin. Infect. Dis., 32(11): 1608-1614.<br />

Katayama Y, Ito T, Hiramatsu K (2000). A new class of genetic element,<br />

Staphylococcus casette chromosome mec, encodes methicillinresistance<br />

in Staphylococcus aureus. Antimicrob Agents<br />

Chemotheer., 44(6): 1549-1555.<br />

Klimek JJ, Marsik FJ, Bartlett RC, Weir B, Shea P, Quintiliani R (1976).<br />

Clinical, epidemiologic and bacteriologic observations of an outbreak<br />

of methicillin-resistant Staphylococcus aureus at a large community<br />

hospital. Am. J Med., 61(3): 340-345.<br />

Kobayashi N, Taniguchi K, Kojima K, Urasawa S, Uehara N, Omizu Y,<br />

Kishi Y, Yagihashi A, Kurokawa I, Watanabe N (1996). Genomic<br />

diversity of mec regulator genes in methicillin-resistant<br />

Staphylococcus aureus and Staphylococcus epidermidis. Epidemiol.<br />

Infect., 117(2): 289-295.<br />

Kobayashi N, Taniguchi K, Urasawa S (1998). Analysis of diversity of<br />

mutations in the mecI gene and mecA promoter/operator region of<br />

methicillin-resistant Staphylococcus aureus and Staphylococcus<br />

epidermidis. Antimicrob. Agents Chemother., 42(3): 717-720.<br />

Kuehnert MJ, Kruzon-Moran D, Hill HA, McQuillan G, McAllister SK,


2272 Afr. J. Microbiol. Res.<br />

Fosheim G, McDougal LK, Chaitram J, Jensen B, Fridkin SK, Killgore<br />

G, Tenover FC (2006). Prevalence of Staphylococcus aureus nasal<br />

colonization in the United States, 2001-2002. J. Infect. Dis., 193(2):<br />

172-179.<br />

Lighthart B, Shaffer BT (1995). Airbone bacteria in the atmospheric<br />

surface layer: temporal distribution above a grass seed field. App.<br />

Environ. Microbiol., 61(4): 1492-1496.<br />

Martins A, Cunha M de L (2007). Methicillin resistance in<br />

Staphylococcus aureus and coagulase-negative staphylococci:<br />

epidemiological and molecular aspects. Microbiol. Immunol., 51(9):<br />

787-795.<br />

McDougal LK, Thornsberry C (1986). The role of �-Lactamase in<br />

Staphylococcal Resistance to Penicillinnase-Resistant Penicillins and<br />

Cephalosporins. J. Clin. Microbiol., 23(5): 832-839.<br />

Menezes GA, Harish BN, Sujatha S, Vinothini K, Parija SC (2008).<br />

Emergence of vancomycin-intermediate Staphylococcus species in<br />

southern India. J. Med. Microbiol., 57(7): 911-912.<br />

Palazzo ICV, Araujo MLC, Darini ALC (2005). First report of<br />

vancomycin-resistant staphylococci isolated from healthy carriers in<br />

Brazil. J. Clin. Microbiol., 43(1): 179-185.<br />

Pérez-Trallero E, Iglesias L (2003). Tetracyclines, sulfonamides and<br />

metronidazole. Enferm. Infect. Microbiol. Clin., 21(9): 520-529.<br />

Petinaki E, Arvaniti A, Dimitracopoulos G, Spiliopoulou I (2001).<br />

Detection of mecA, mecRI and mecI genes among clinical isolates of<br />

methicillin-resistant Staphylococci by combined polymerase chain<br />

reactions. J. Antimicrob. Chermother., 47(3): 297-304.<br />

Pfaller MA, Jones RN, Doern GV, Kugler K (1998). Bacterial pathogens<br />

isolated from patients with bloodstream infection: frequencies of<br />

occurrence and antimicrobial susceptibility patterns from the<br />

SENTRY Antimicrobial Surveillance Program (United States and<br />

Canada, 1997). Antimicrob. Agents Chemother., 42(7): 1762-1770.<br />

Pfaller MA, Jones RN, Doern GV, Sader HS, Kugler KC, Beach ML<br />

(1999). Survey of blood stream infections attributable to Grampositive<br />

cocci: frequency of occurrence and antimicrobial<br />

susceptibility of isolates collected in 1997 in the United States,<br />

Canada, and Latin America from the SENTRY Antimicrobial<br />

Surveillance Program. SENTRY Participants Group. Diagn. Microbiol.<br />

Infect. Dis., 33(4): 283-297.<br />

Pinho MG, Filipe SR, De Lencastre H, Tomasz A (2001).<br />

Complementation of essential peptidoglycan transpeptidase function<br />

of penicillin-binding protein 2 (PBP2) by the drug resistance protein<br />

PBP2A in Staphylococcus aureus. J. Bacteriol., 183(2): 6525-6531.<br />

Ponce de León S (1996). New and old pathogens in nosocomial<br />

infections. In: prevention and control of hospital infections Manual.<br />

(HSP Serie / PALTEX Manuals) Pan Am. Health Organ. (PAHO),<br />

4(13): 52-68.<br />

Rohrer S, Maki H, Berger-Bächi B (2003). What makes resistance to<br />

methicillin heterogeneous? J. Med. Microbiol., 52(8): 605-607.<br />

Rosa J de O, De Moura JP, Palos MA, Gir E, Reis C, Kipnis A, Canini<br />

SR, Belissimo-Rodríguez F, Pimenta FC (2009). Detection of mecA<br />

gene in oxacillin-resistant coagulase-negative staphylococci isolated<br />

from the saliva of nursing professionals. Rev. Soc. Bras. Med. Trop.,<br />

42(4): 398-403.<br />

Rosas I, Cravioto A, Ezcurra E (2004). Environmental Microbiology. In:<br />

Ministry Environment and Natural Resources, Ecology National<br />

Institute (eds), Bacteria in the Atmosphere, University Program of<br />

Environment-UNAM: Program SA Mexico, pp. 1-134.<br />

Rosato AE, Kreiswirth BN, Craig WA, Eisner W, Climo MW, Archer GL<br />

(2003). mecA-blaZ corepressors in clinical Staphylococcus aureus<br />

isolates. Antimicrob. Agent Chemother., 47(4): 1460-1463.<br />

Ryffel C, Tesch W, Birch-Machin I, Reynolds P E, Barberis-Maino L,<br />

Kayser FH, Berger-Bächi B (1990). Sequence comparison of mecA<br />

genes isolated from methicillin-resistant Staphylococcus aureus and<br />

Staphylococcus epidermidis. Gene., 94(1): 137-138.<br />

Ryffel C, Kayser FH, Berger-Bächi B (1992). Correlation between<br />

regulation of mecA transcription and expression of methicillin<br />

resistance in staphylococci. Antimicrob. Agents. Chemother., 36(1):<br />

25-31.<br />

Sader HS, Jones RN, Gales AC, Winokur P, Kugler KC, Pfaller MA,<br />

Doern GV (1998). Antimicrobial susceptibility patterns for pathogens<br />

isolated from patients in Latin American medical centers with a<br />

diagnosis of pneumonia: analysis of results from the SENTRY<br />

Antimicrobial Surveillance Program (1997). SENTRY Latin America<br />

Study Group. Diagn. Microbiol. Infect. Dis., 32(4): 289-301.<br />

Sader HS, Moet GJ, Jones RN (2009). Antimicrobial resistance among<br />

Gram-positive bacteria isolated in Latin American hospitals. J.<br />

Chemother., 21(6): 611-620.<br />

Schaberg DR, Culver DH, Gaynes RP (1991). Major trends in the<br />

microbial etiology of nosocomial infection. Am. J. Med., 91(3B): 72S–<br />

75S.<br />

Schlegelová J, Babák V, Klímová E, Lukásová J, Navrátilová P,<br />

Sustáckova A, Sedivá I, Rysánek D (2002). Prevalence of and<br />

resistance to anti-microbial drugs in selected microbial species<br />

isolated from bulk milk samples. J. Vet. Med. B. Infect. Dis. Vet.<br />

Public Health, 49(5): 216-225.<br />

Schwalbe RS, Stapleton JT, Gilligan PH (1987). Emergence of<br />

vancomycin resistance in coagulase-negative staphylococci. N. Engl.<br />

J. Med., 316: 927-931.<br />

Suzuki E, Kuwahara-Arai K, Richardson JF, Hiramatsu K (1993).<br />

Distribution of mec regulator genes in methicillin-resistant<br />

Staphylococcus clinical strains. Antimicrob. Agents Chemother.,<br />

37(6): 1219-1226.<br />

Tang JW (2009). The effect of environmental parameters on the survival<br />

of airborne infectious agents. J. R. Soc. Interface, 6(6): S737-S746.<br />

Tiwari HK, Sen MR (2006). Emergence of vancomycin resistant<br />

Staphylococcus aureus (VRSA) from a tertiary care hospital from<br />

northern part of India. BMC. Infect. Dis., 6: 156.<br />

Tomasz A, Drugeon HB, De Lencastre HM, Jabes D, McDougal L, Bille<br />

J (1989). New mechanism for methicillin resistance in<br />

Staphylococcus aureus: clinical isolates that lack the PBP 2a gene<br />

and contain normal penicillin-binding proteins with modified penicillinbinding<br />

capacity. Antimicrob. Agents Chemother., 33(11): 1899-1874.<br />

Tsai FC, Macher JM (2005). Concentrations of airborne culturable<br />

bacteria in 100 large US office buildings from. The base study. Indoor<br />

Air., 15(9): 71-81.<br />

Ubukata K, Nonoguchi R, Song MD, Matsuhashi M, Konno M (1990).<br />

Homology of mecA gene in methicillin-resistant Staphylococcus<br />

haemolyticus and Staphylococcus simulans to that of Staphylococcus<br />

aureus. Antimicrob. Agents Chemother., 34(1): 170-172.<br />

Unal S, Hoskins J, Flokowitsch JE, Wu CY, Preston DA, Skatrud PL<br />

(1992). Detection of methicillin-resistant staphylococci by using the<br />

polymerase chain reaction. J. Clin. Microbiol., 30(7): 1685-1691.<br />

Weller TMA (1999). The distribution of mecA, mecR1 and mecI and<br />

sequence analysis of mecI and the mec promoter region in<br />

staphylococci expressing resistance to methicillin. J. Antimicrob.<br />

Chemother., 43(1): 15-22.<br />

Wisplinghoff H, Seifert H, Wenzel RP, Edmond MB (2003). Current<br />

Trends in the Epidemiology of Nosocomial Bloodstream Infections in<br />

Patients with Hematological Malignancies and Solid Neoplasms in<br />

Hospitals in the United States. Clin. Infect. Dis., 36(9): 1103-1110.<br />

World Health Organization (WHO) (1999). WHO Model Prescribing<br />

Information: Drugs Used in Skin Diseases. Bacterial infections.<br />

Geneva: World Health Organization, pp. 1-132.<br />

World Health Organization (WHO) (2002). Antimicrobial resistance. Fact<br />

sheet N°194.<br />

Wu CJ, Lee HC, Lee NY, Shih HI, Ko NY, Wang LR, Ko WC (2006).<br />

Predominance of Gram-negative bacilli and increasing antimicrobial<br />

resistance in nosocomial bloodstream infections at a university<br />

hospital in southern Taiwan, 1996-2003. J. Microbiol. Immunol.<br />

Infect., 39(2): 135-143.


African Journal of Microbiology Research Vol. 5(16), pp. 2273-2277, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.590<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Interaction of severe acute respiratory syndrome<br />

(SARS) nucleocapsid protein with macrophage<br />

migration inhibitory factor protein (MIF)<br />

Yang Lin 1,2 *, Baohua Wang 3 *, Yijun Liu 1 , Zhigang Yu 1 , Manhua Cui 2 and Haichun Ma 1<br />

1 The First Hospital of Jilin University, Changchun130021, China.<br />

2 Department of Gynaecology and Obstetrics, the Second Hospital of Jilin University, Changchun 130041 China.<br />

3 Department of Anesthesia, the Affiliated Hospital of Changchun University of Traditional Chinese Medicine,<br />

Changchun 130021, China.<br />

Accepted 30 July, 2011<br />

The nucleocapsid (N) protein of SARS coronavirus (SARS-CoV) is a major structural component of<br />

virions, which appears to be a multifunctional protein involved in viral RNA replication and translation.<br />

However, how N protein interacts with host protein remains largely elusive. To identify cellular proteins<br />

that interact with the N protein and to elucidate the possible involvement of N protein in SARS-Cov<br />

replication and translation, a human lung cDNA library was screened using a yeast two-hybrid system<br />

assay. In this study, we have identified Macrophage migration inhibitory factor protein (MIF) as a novel<br />

interaction partner of N protein by yeast two-hybrid system. The direct interaction and co-localization of<br />

N protein with MIF were confirmed by immunoprecipitation and confocal microscopy analysis,<br />

respectively. The mapping studies localized the critical N sequences for this interaction to amino acid<br />

140-210 including SR motif by yeast two-hybrid system. To the best of our knowledge, this is the first<br />

report that SARS-N protein interacts with the MIF within host cells, which enhance our understanding of<br />

the molecular mechanisms of SARS replication.<br />

Key words: Severe acute respiratory syndrome (SARS) coronavirus, nucleocapsid protein, Macro|phage<br />

migration inhibitory factor protein, yeast two-hybrid.<br />

INTRODUCTION<br />

Severe acute respiratory syndrome (SARS) is a novel<br />

worldwide infectious disease, which is caused by a newly<br />

identified member of the coronaviridae family, SARS<br />

coronavirus (SARS-CoV) (Ksiazek et al., 2003; Marra et<br />

al., 2003; Peiris et al., 2003; Rota et al., 2003). Like other<br />

known coronaviruses, SARS-CoV is an enveloped virus<br />

containing three outer structural proteins, namely the<br />

membrane (M), nucleocapsid (N), and spike (S) proteins<br />

(Lai et al., 1997; Versteeg et al., 2007). The N protein is<br />

the most abundant viral protein in coronaviruses which is<br />

produced throughout infection and is an important<br />

multifunctional protein. Several functions have been postulated<br />

for the coronavirus N protein throughout the<br />

*Corresponding author. E-mail: Haichun514@yahoo.cn. Fax:<br />

+86-0431-845-14166.<br />

virus life cycle, including viral packaging, viral core<br />

formation, and signal transduction (He et al., 2004;<br />

Hiscox et al., 2001).<br />

Mapping virus–host protein interactions can provide<br />

important clues on the initial stages of infection. For coronaviruses,<br />

the N protein plays an important role during<br />

host cell entry and virus particle assembly and release<br />

(Hatakeyama et al., 2008; Narayanan et al., 2001). It has<br />

been reported that the N protein of SARS interacts with<br />

human cellular heterogeneous nuclear ribonucleoprotein<br />

A1 (hnRNPA1) (Wang et al., 1999), which shown that N<br />

protein plays a crucial role in viral RNP assembly and<br />

replication by interaction with host proteins. However,<br />

how N protein interacts with host protein is still unclear.<br />

To obtain a more detailed insight into the N protein and<br />

host relationship, a yeast two-hybrid system was<br />

employed in the present study to identify host proteins<br />

which bind to N protein of SARS.


2274 Afr. J. Microbiol. Res.<br />

MATERIALS AND METHODS<br />

Strains and general techniques<br />

The strain of Saccharomyces cerevisiae used in this study was<br />

AH109 from Clontech (USA). Yeast cells were cultured at 30°C<br />

either in a complete YPD medium (1% yeast extract, 1% peptone,<br />

2% glucose) or in a synthetic defined (SD) medium supplemented<br />

with required essential nutrients. Plates contained 2% agar.<br />

Transformation of yeast cells was performed by the lithium acetate.<br />

Escherichia coli KC8 were used for general cloning. DNA manipulation<br />

was performed according to established protocol. Betagalactosidase<br />

assays were carried out according to the<br />

CLONTECH Matchmaker manual (PT3024-1, Clontech, USA).<br />

Plasmids and construction of recombinant vectors<br />

For bait construction, the full-length N gene of the SARS was PCRamplified<br />

from a genomic construct of clone, and cloned into the<br />

pMD18-T vector (Takara, China). The full-length N gene was<br />

subjected to DNA sequencing, and the inserts were verified against<br />

the corresponding region of the SARS coronavirus. The full-length<br />

N gene was excised from the pMD18-T –N construct using the<br />

restriction enzymes EcoRI and BamHI, and ligated into the pGBKT7<br />

vector to generate an N-terminal in frame fusion with the GAL4<br />

activation domain (BD), and the resultant plasmid was named as<br />

pGBKT7-N. To identify the putative domain of amino acid sequence<br />

required for MIF/N interaction. The truncated mutants N1–139,<br />

N140–280, N281–422, N140–210 and N211–280 were subcloned<br />

into the yeast two-hybrid vector pGBKT7. The MIF gene was<br />

obtained by PCR from the rat liver cDNA library, and subcloned into<br />

the yeast vector pGADT7. For mammalian cell expression, the fulllength<br />

N gene and MIF were subcloned into the pCMV-Myc vector<br />

(Clontech, USA) and pCMV–HA, and fluorescence vector pEGFP-<br />

N1 and pDsRed-N1, respectively. All DNA manipulations were<br />

performed as described by Sambrook et al. (2001). All constructs<br />

were verified by restriction digestion and sequencing.<br />

Yeast two-hybrid screening<br />

Yeast two-hybrid experiments were performed as described in the<br />

Clontech manual for the MATCHMAKER GAL4 two-hybrid system<br />

and in the Clontech yeast protocols handbook (Clontech, USA).<br />

The rat liver Matchmaker cDNA library (Clontech, USA) cloned in<br />

frame with the GAL4 activation domain in the PGAD vector was<br />

used as a prey. The bait pGBKT7-N was transformed into yeast<br />

strain AH109. The bait construct did not show any toxic effect and<br />

autonomous transcriptional activation on the host strain. The prey<br />

PGAD-library was then transformed into the bait-transformed<br />

AH109 cells, and the cells were incubated on minimal synthetic<br />

dropout medium for yeast (SD)/-His/-Leu/-Trp at 30°C. The fresh<br />

growing clones were assayed for -galactosidase activity by replica<br />

plating the yeast transformants onto Whatman filter papers; the<br />

filters were snap-frozen in liquid nitrogen for 10 sec twice and<br />

incubated in a buffer containing 5-bromo-4-chloro 3-indolyl- -Dgalactopyranoside<br />

solution at 30°C for 1–8 h. Positive interactions<br />

were detected by the appearance of blue clones. The liquid -<br />

galactosidase activity was determined using the substrate ONPG as<br />

described standard Protocols Handbook (PT3024-1, Clontech,<br />

USA). Data for quantitative assays were collected for yeast cell<br />

number and are the mean ± S.E.M. of triplicate assays. Appropriate<br />

positive/negative controls and buffer blanks were used. The positive<br />

pGAD-cDNA plasmids were isolated from positive yeast transformants<br />

by culture in leucine-deficient medium, which resulted in<br />

spontaneous loss of the plasmid pGBK-N and transformed into E.<br />

coli KC8 for sequence analysis. Auto sequencing assay was<br />

performed in Takara Company (China) and the resulting sequence<br />

was analyzed in the database of EMBL\Gene Bank by the BLAST<br />

program.<br />

In vivo co-immunoprecipitation and western blotting<br />

To reaffirm the results observed from yeast two-hybrid assays, another<br />

independent assay, co-immunoprecipitation was carried out.<br />

Vero cells were co-transfected with the plasmids expressing pCMV-<br />

Myc-MIF and pCMA-HA-N using the Iipofectamine TM transfaction<br />

reagent (Invitrogen, USA). At 48 h post-transfection, cells were<br />

washed with PBS and then lysed in lysis buffer. Cell lysate was<br />

then mixed with anti-HA magnetic microbeads for 30 min on ice.<br />

100 l of 10% suspension of protein A–Sepharose was then added<br />

to the samples. The mixture was allowed to shake for 1 h at 4°C,<br />

after following which the beads were washed four times in lysis<br />

buffer, and protein was eluted in 2×SDS dye by boiling the sample<br />

for 5 - 10 min. Samples were resolved by 10% SDS-PAGE and<br />

transferred to a nitrocellulose membrane. The membrane was<br />

blocked using 0.5% BSA in PBST for 1 h, and incubated overnight<br />

with anti-Myc antibodies (1:1000; Clontech, USA). The blot was<br />

then washed three times in PBST, incubated with anti-mouse IgG<br />

HRPO for 1 h and washed three times in PBST and the proteins<br />

were visualized with 5-bromo-4-chloro-3-indolyl phosphate and<br />

nitroblue tetrazolium. The Vero cells transected with the pCMV-Myc<br />

and pCMV-HA vectors were used as negative controls.<br />

Confocal microscopy<br />

Hela cells were grown on coverslips in a 6-well chamber and<br />

simultaneously transfected with the recombinant fluorescence<br />

plasmids pEGFP-N and pDsRed-MIF. After 24 h transfection, the<br />

cells were washed with PBS three times and fixed in 4% paraformaldehyde<br />

for 20 min at room temperature. The coverslips were<br />

then washed with PBS and mounted. Intracellular localization of the<br />

N protein and MIF was observed under a Leica confocal<br />

microscope (Germany).<br />

RESULTS<br />

Yeast two-hybrid identified MIF as an N interacting<br />

protein<br />

A yeast two-hybrid approach was used to identify host<br />

proteins that interact with N protein. For this purpose, the<br />

complete N gene was cloned into a two-hybrid bait<br />

expression vector. The resulting Gal4-N fusion protein<br />

was used to screen a human lung cDNA library. Approximate<br />

2.2 × 10 6 transformants were screened for His-Ade-<br />

Trp-Leu independent growth and blue colony formation in<br />

the -galactosidase assay. Thirty-six positive clones were<br />

obtained. As some AD-cDNA fusion products can activate<br />

reporter gene transcription without interacting with<br />

the BD-N fusion protein, this false-positive clone can be<br />

identified using the technique of segregation analysis.<br />

Only 4 of 36 clones survived all genetic tests and were<br />

considered to be genuine positive clone, DNA sequence<br />

analysis of the fragment revealed that the four cDNA<br />

fragments inserted have a high identity with four genes in<br />

the GeneBank database. One of these clones was identified<br />

as MIF, which encodes a 158-amino acid protein.


Figure 1. MIF protein was immunoprecipitated with the MHV-N protein. Indicated<br />

plasmids were simultaneously transfected into Vero cells. Twenty-four hours after<br />

transfection, coimmunoprecipitation was per-formed using anti-HA magnetic<br />

microbeads, the proteins immuno-precipitated (IP) were assayed with an anti-myc<br />

monoclonal antibody. Cell lysates were immunoblotted (IB) with anti-Myc to confirm<br />

the expression of the object proteins. M: MIF protein; N : N protein.<br />

Lin et al. 2275<br />

Figure 2. Co-localization of the N protein and MIF. pEGFP-N (green) and pDsRed-RACK1 (red) were co-transfected into<br />

HeLo cells. After 24 h, cells were fixed, mounted, and the localization of the proteins was observed with a Leica confocal<br />

microscope. As shown, the N protein and MIF were colocalized in the cytoplasm.<br />

As shown in Figure 3B, the protein encoded by the<br />

pGAD-MIF clones interacted specifically with the N<br />

protein and did not interact with the unfused GAL4-BD<br />

protein expressed from the parental pGBKT7 vector.<br />

Co-immunoprecipitation determined the interaction<br />

of the N protein and MIF<br />

To confirm the specific interaction between N protein and<br />

MIF, co-immunoprecipitation was performed. The N protein<br />

was fused at the amino terminus with a Myc-tag, and<br />

MIF was fused at the carboxyl terminus with a HA tag.<br />

The two plasmids were cotransfected into Vero cells and<br />

immunoprecipitated. The immunoprecipitated complexes<br />

were separated on SDS–PAGE, and analyzed by Western<br />

blot with anti-Myc monoclonal antibodies. As shown<br />

in Figure1, the Myc-fused N protein immunoprecipitates<br />

with HA-MIF. However, it does not immunoprecipitate<br />

with HA alone. These findings indicate that N protein<br />

interacts with MIF in mammalian cells<br />

Confocal microscopy assay the N protein and MIF<br />

To further confirm the interaction between N and MIF<br />

proteins, the localization patterns of the N protein and<br />

MIF were investigated in Vero cells using confocal microscopy<br />

technology. pEGFP-N and pDsRed-MIF were<br />

transfected simultaneously into Vero cells. As shown in<br />

Figure 2, MIF and N protein mainly were localized in the<br />

cytoplasm. The merged image revealed that the N protein<br />

and MIF co-localized in the cytoplasm of Vero cells indicating<br />

that N protein interacts with MIF in cells.


2276 Afr. J. Microbiol. Res.<br />

MIF+N281-422<br />

MIF+N211-280<br />

MIF+N140-210<br />

MIF+N140-280<br />

MIF+N1-139<br />

MIF+N<br />

Negative<br />

Postive<br />

0 20 40 60 80 100<br />

Figure 3. Mapping the interaction domain of N protein. The<br />

empty vectors pGBKT7 and pGADT7 co-transformed were used<br />

as the negative control and the pGBKT7-53 and pGADT7- T cotransformed<br />

were used as the postive con-trol. Every<br />

experiment was repeated for at least three times and the data<br />

were obtained by average. The error bars represent standard<br />

error of the mean. -galactosidase activity assay.<br />

Mapping the MIF binding region of the N Protein by<br />

yeast two-hybrid assays<br />

To map the involved regions of N protein in the N/MIF<br />

interaction, the full-length N protein was divided into three<br />

domains to study the binding domain of the N protein: the<br />

N-terminal domain N1–139, the middle domain N140–<br />

280 containing the SR-rich motif and the C-terminal<br />

domain N281–422 (Figure 3). These three truncated<br />

proteins were tested for MIF binding using yeast twohybrid<br />

assay. The results indicated that N140–280, the<br />

middle domain of the N protein, is responsible for the<br />

interaction (Figure 3). To determine further the region of<br />

the N protein involved in MIF binding, the middle domain<br />

was divided into two parts, N140–280 containing the SRrich<br />

motif and N140–210, In the yeast two-hybrid assay,<br />

the -galactosidase activity of N140–210 fragment is<br />

much higher than the activity of N211–280 fragment<br />

(Figure 3). These results implied that the SR-rich motif of<br />

the N protein is responsible for the majority of the binding<br />

to MIF.<br />

DISCUSSION<br />

In the present study, by employing a series of biochemical<br />

and biophysical methods, we have firstly reported<br />

that SARS-N protein has a specific binding to human MIF<br />

protein, and the further yeast two-hybrid assay demonstrated<br />

that the fragment amino acid 140–210 of SARS-N<br />

probably contribute to the N/MIF interaction. MIF was first<br />

discovered as a lymphokine involved in delayed hypersensitivity<br />

and various macrophage functions, including<br />

phagocytosis, spreading, and tumoricidal activity. It has<br />

been reported that MIF is associated with the pathogenesis<br />

of a variety of diseases (Crichlow et al. , 2009;<br />

Oddo et al., 2005). Multiple clinical studies have indicated<br />

the utility of MIF as a biomarker for different diseases that<br />

have an inflammatory component; these include systemic<br />

infections and sepsis, autoimmune diseases, cancer, and<br />

metabolic disorders such as type 2 diabetes and obesity<br />

(Shen et al., 2003; Wistow et al., 2005). Over the past<br />

few years, the significance of the role of MIF in a variety<br />

of both solid and hematologic tumors has been<br />

established. Both circulating and intracellular MIF protein<br />

levels are elevated in cancer patients and MIF expression<br />

reportedly correlates with stage, metastatic spread and<br />

disease-free survival.<br />

To our knowledge, the fragment amino acid 140–210<br />

contains SR-rich motif (containing rich serine and<br />

arginine) that is multifunctional and conserved in N<br />

protein of coronaviruses (Toney et al., 2004). SR-related<br />

proteins are often involved in protein-RNA and protein–<br />

protein interactions (Blencowe et al., 1999), and the SRrich<br />

motif is conserved in the N protein of coronavirus. It<br />

has been reported that SR-rich motif is indispensable for<br />

SARS N oligomerization and for N protein interaction with<br />

SARS-CoV membrane protein. The SR-rich motif is also<br />

responsible for the interaction with hnRNPA1 in SARS<br />

(Haibin et al., 2005). In addition, this motif is also involved<br />

in interaction with Hubc9 in SARS (Fan et al., 2006). All<br />

of these facts indicate that the SR-rich motif of the N<br />

protein might play a crucial role in SARS infection.<br />

In conclusion, our data have shown for the first time<br />

both in vitro and in vivo that the nucleocapsid protein of<br />

SARS has a high binding affinity to human MIF and such<br />

a protein–protein interaction involves the region amino<br />

acid 140–210 of MHV-N. However, the pathophysiological<br />

significance of the interaction between N and MIF<br />

is largely unknown; elucidation of these questions will<br />

depend on further studies. Moreover, the disruption of<br />

interaction between N and MIF proteins using RNA<br />

interference technology may provide further clues to the<br />

specific function of N and MIF protein. This current<br />

research contributes useful data that will shed light on the<br />

molecular mechanism of N phosphorylation and provide<br />

valuable clues for mutagenic studies in disrupting virion<br />

assembly and replication and developing antiviral agents.<br />

ACKNOWLEDGEMENT<br />

The authors gratefully acknowledge the financial support<br />

provided by Jilin administration of traditional Chinese<br />

medicine TCM science project (20090461042).<br />

REFERENCES<br />

Blencowe BJ, Bowman JA, McCracken S, Rosonina E (1999). SR<br />

related proteins and the processing of messenger RNA precursors.<br />

Biochem. Cell. Biol., 77:257-271.<br />

Crichlow GV, Lubetsky JB, Leng L, Bucala R (2009).Structural and<br />

kinetic analyses of macrophage migration inhibitory factor active site<br />

interactions. Biochem., 48:132-139.<br />

Fan Z, Zhuo Y, Tan X, Zhou Z, Yuan J, Qiang B, Yan J, Peng X, Gao GF


(2006). SARS-CoV Nucleocapsid Protein Binds to hUbc9, a Ubiquitin<br />

Conjugating Enzyme of the Sumoylation System. Med. Virol.,<br />

78:1365-1373.<br />

Haibin L, Qing C, Shen X, Hualiang J (2005). The nucleocapsid protein<br />

of SARS coronavirus has a high binding affinity to the human cellular<br />

heterogeneous nuclear ribonucleoprotein. FEBS. Lett., 579:2623-<br />

2628.<br />

Hatakeyama S, Matsuoka Y, Ueshiba H (2008). Dissection and<br />

identification of regions required to form pseudoparticles by the<br />

interaction between the nucleocapsid (N) and membrane (M) proteins<br />

of SARS coronavirus. Virology, 380:99-108.<br />

He R, Dobie F, Ballantine M, Leeson A, Yan L, Batien N, Cutts T,<br />

Andonov A, Jingxin C, Booth TF, Plummer FA, Tyier S, Baker L, Li XG<br />

(2004). Analysis of multimerization of SARS coronavirus<br />

nucleocapsid protein. Biochem. Biophys. Res. Commun., 316:476-<br />

483.<br />

Hiscox JA, Wurm T, Wilson L, Britton P, Cavanagh D, Brooks G<br />

(2001).The coronavirus infectious bronchitis virus nucleoprotein<br />

localizes to the nucleolus. J. Virol., 75:506-512.<br />

Ksiazek TG, Erdman D, Goldsmith CS, Zaki SR (2003). A novel<br />

coronavirus associated with severe acute respiratory syndrome. N.<br />

Engl. J. Med., 348 (20):1953-1966.<br />

Lai M, Cavanagh D (1997). The molecular biology of coronaviruses.<br />

Advances. Virus. Research., 48:1-100.<br />

Marra MA, Jones SJ, Astell CR, Holt RA (2003). The Genome sequence<br />

of the SARS-associated coronavirus. Science, 300(5624):1399-1404.<br />

Narayanan K, Maeda A, Maeda J, Makino S (2000). Characterization of<br />

the coronavirus M protein and nucleocapsid interaction in infected<br />

cells. J. Virol., 74:8125-8134.<br />

Oddo M, Calandra T, Bucala R, Meylan PR (2005).Macrophage<br />

migration inhibitory factor reduces the growth of virulent<br />

Mycobacterium tuberculosis in human macrophages. Infect. Immun.,<br />

73:3783-3786.<br />

Lin et al. 2277<br />

Peiris JS, Lai ST, Poon LL, Guan Y (2003). Coronavirus as a possible<br />

cause of severe acute respiratory syndrome. Lancet,<br />

361(9366):1319-1325.<br />

Rota PA, Oberste MS, Monroe SS, Nix WA (2003). Characterization of a<br />

novel coronavirus associated with severe acute respiratory<br />

syndrome. Science, 300(5624):1394-1399.<br />

Sambrook J, Russell DW (2001). Molecular cloning: A laboratory<br />

manual. 3rd ed. Cold Spring Harbor (NY): Cold Spring Harbor<br />

Laboratory Press.<br />

Shen L, Hu J, Lu H, Wu M, Qin W, Wan D(2003).The apoptosisassociated<br />

protein BNIPL interacts with two cell proliferation-related<br />

proteins, MIF and GFER.FEBS. Lett., 540:86-90.<br />

Toney J, Navas-Martin S, Weiss S, Koeller A (2004). Sabadinine: a<br />

potential non-peptide anti-severe acute-respiratory-syndrome agent<br />

identified using structure-aided design. J. Med. Chem., 47(5):1079-<br />

1080.<br />

Versteeg G, Bredenbeek P, Spaan W (2007). Group 2 coronaviruses<br />

prevent immediate early interferon induction by protection of viral<br />

RNA from host cell recognition. Virology, 361(1):18-26.<br />

Wang Y, Zhang X(1999).The nucleocapsid protein of coronavirus<br />

mouse hepatitis virus interacts with the cellular heterogeneous<br />

nuclear ribonucleoprotein A1 in vitro and in vivo. Virology., 265(1):96-<br />

109.<br />

Wistow GJ, Shaughnessy M, Lee DC, Hodin J(1993).A macrophage<br />

migration inhibitory factor is expressed in the differentiating cells of<br />

the eye lens. Proc. Natl. Acad. Sci., 90:1272-1275.


African Journal of Microbiology Research Vol. 5(16), pp. 2278-2286, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.611<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Isolation and characterization of high caffeine-tolerant<br />

bacterium strains from the soil of tea garden<br />

Fang-Yuan Fan 1 , Yan Xu 1 , Yue-Rong Liang 1 , Xin-Qiang Zheng 2 , Devajit Borthakur 1,3 and<br />

Jian-Liang Lu 1 *<br />

1 Tea Research Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.<br />

2 Key Laboratory of Horticultural Plants Growth, Development and Biotechnology, Agricultural Ministry of China,<br />

Hangzhou 310058, China.<br />

3 Tocklai Experimental Station, Tea Research Association, Jorhat, Assam 785001, India.<br />

Accepted 30 July, 2011<br />

Microbial biodegradation is an important and promising decaffeination approach because of its low<br />

cost and high security. The efficiency of this approach mainly depends on the characteristics of the<br />

strain. Twenty bacterium strains were isolated from soil of tea garden and exhibited high caffeinetolerance,<br />

and these strains could grow on the medium supplemented with 20 g L -1 caffeine as the sole<br />

source for nitrogen and carbon. According to the physiological-biochemical characteristics and 16S<br />

rRNA gene sequence blast, two of them (CT25 and CT75) were identified as Pseudomonas putida. The<br />

results also showed that caffeine was not the preferential nutrition source for growth of strain CT25,<br />

and high level caffeine inhibited the bacterium amplification although caffeine could be metabolized by<br />

this strain. The CT25 grew well in agitated liquid medium when the incubation temperature was around<br />

30°C. High initial concentration of strain inoculums would improve the efficiency of caffeine<br />

degradation.<br />

Key words: Caffeine-tolerant bacterium, identification, 16S rRNA gene, caffeine biodegradation, growth<br />

condition.<br />

INTRODUCTION<br />

Caffeine (1,3,7-trimethylxanthine), a member of purine<br />

alkaloids, was isolated first from tea and coffee in the<br />

early 1820s and then reported in the beans, leaves and<br />

fruits of more than 60 plants, such as coffee, tea, mate,<br />

guarana, cola and cocoa (Heckman et al., 2010).<br />

Medicinally, caffeine is used as a cardiac, cerebral and<br />

respiratory stimulant (Buerge et al., 2003). However, it<br />

can also produce adverse effects on human health<br />

unavoidably. Caffeine consumption during pregnancy is<br />

associated with an increased risk of fetal growth<br />

restriction (CARE Study Group, 2008). Excessive intake<br />

of caffeine can increase the risk of miscarriage,<br />

independent of pregnancy-related symptoms (Weng et<br />

*Corresponding author. E-mail: jllu@zju.edu.cn. Tel: 86-571-<br />

88982704.<br />

al., 2008). Statistically significantly higher risk of ovarian<br />

cancer among women who reported drinking five or more<br />

cups of caffeinated coffee per day compared to nonconsumers<br />

of coffee (Lueth et al., 2008). The ingestion of<br />

either a high or low glycemic index meal with caffeinated<br />

coffee significantly impairs acute insulin sensitivity<br />

compared with the effect of decaffeinated coffee (Moisey<br />

et al., 2008). Large sample and population-based studies<br />

indicated that regular daily dietary caffeine intake is<br />

associated with disturbed sleep and associated daytime<br />

sleepiness (Roehrs and Roth, 2008). For older women,<br />

blood pressure response is augmented, and subjective<br />

feelings of behavioral mood state are attenuated to a<br />

greater degree following acute caffeine ingestion (Arciero<br />

and Ormsbee, 2009). Higher level of caffeine also leads<br />

to higher risk of developing bone problems, such as<br />

osteoporosis (Hallstrom et al., 2006), alveolar bone loss<br />

in ligature-induced periodontitis (Bezerra et al., 2008), an


d disturbing the early stages of bone healing (Duarte et<br />

al., 2009). Tea polyphenols, an excellent antioxidant<br />

extracted from tea (Panza et al., 2008), is always contaminated<br />

with amount of caffeine because of the technical<br />

constraints of decaffeination, which hampers their<br />

application in the field of pharmaceutical and functional<br />

food.<br />

Environmentally, caffeine has been suggested as a<br />

chemical indicator of ecosystem since it is difficultly<br />

metabolized (Ogunseitan, 2002). When the exposure<br />

dosage of caffeine in water was higher than 300 mg L -1 ,<br />

no zebrafish embryos could survive, and caffeine-treated<br />

embryos exhibited significantly reduced tactile sensitivity<br />

frequencies of touch-induced movement even when<br />

exposure dosages were very low (Chen et al., 2008).<br />

Reports also showed that caffeine is toxic to most of the<br />

aquatic organism such as Ceriodaphnia dubia (Moore et<br />

al., 2008) and coral algae (Pollack et al., 2009). Therefore,<br />

release of some caffeine-containing wastes, such as<br />

coffee pulp/husk from coffee producing factory, infused<br />

tea leaves and sewage from the factory of tea<br />

polyphenols preparation, leads serious ecological<br />

pollutions and becomes a big disposal problem (Pandey<br />

et al., 2000).<br />

To date, four major approaches for reducing caffeine<br />

content from caffeine-containing products are conventional<br />

breeding, physicochemical methods, genetic<br />

engineering and microbial degradation, whilst approach<br />

for wastes decaffeination is rarely concerned. Decaffeination<br />

through conventional breeding is difficult to achieve<br />

because of non-availability of the low-caffeine containing<br />

germplasms. Genetically-modified (GM) plants with low<br />

caffeine can be directly obtained either by down-regulating<br />

the caffeine synthesis pathway or by up-regulating<br />

the caffeine degradation pathway (Yadav and Ahuja,<br />

2007). However, this approach can not be applied<br />

recently because of their instability and ecological risk of<br />

GM plants, even though successful study in laboratory<br />

scale was reported (Kato et al., 2000). Decaffeination<br />

with special reagents extraction (Sun et al., 2010) and<br />

chromatograph separation is usually adopted in practices<br />

(Dong et al., 2011; Lu et al., 2010; Ye et al., 2009)<br />

although these techniques need to be optimized furthermore<br />

since their lack of security or/and yield. Comparatively,<br />

decaffeination through microbial degradation is more<br />

beneficial and attractive than other methods since it can<br />

be conducted safely and at a low budgetary requirement<br />

(Gummadi and Santhosh, 2006; Ramarethinam and<br />

Rajalakshmi, 2004).<br />

Microbial decaffeination, mainly including bacteria and<br />

fungi, was reported first in the early 1970’s (Kurtzman<br />

and Schwimmer, 1971). Reports showed that Aspergillus<br />

and Penicillium (Hakil et al., 1998), Rhizopus delemar<br />

(Tagliari et al., 2003) and Aspergillus tamari (Gutierrez-<br />

Sanchez et al., 2004) can effectively degrade caffeine.<br />

Major bacteria with capability of caffeine degradation<br />

belong to Pseudomonas (Gokulakrishnan et al., 2005;<br />

Mazzafera, 2004; Sarath Babu et al., 2005; Dash and<br />

Fan et al. 2279<br />

Gummadi, 2006a), beside some other species within the<br />

genera Alcaligenes, Rhodococcus, Klebsiella (Dash and<br />

Gummadi, 2006b), Acinetobacter, Enterobacter,<br />

Stenotrophomonas, Pantoea spp. (Padmanabhan et al.,<br />

2003) and Serratia marcescens (Mazzafera et al., 1996).<br />

The characteristic and mechanism of caffeine biodegradation<br />

were investigated in many Pseudomonas strains<br />

which were isolated from the wastewater (Ogunseitan,<br />

1996, 2002) and from the soils of coffee plantation area<br />

(Dash and Gummadi, 2007, 2008; Gokulakrishnan and<br />

Gummadi, 2006; Gokulakrishnan et al., 2007; Gummadi<br />

et al., 2009b) and normal agricultural land (Topp et al.,<br />

2006; Yu et al., 2008, 2009). Comparatively, the<br />

Pseudomonas isolated from coffee plantation area<br />

exhibits the highest caffeine tolerance, indicating that<br />

caffeine content in the growth environment might affect<br />

the caffeine tolerance capability of the strains. Recently,<br />

bioreactor for decaffeination was also developed<br />

(Gummadi et al., 2009a, 2009b) and optimized<br />

(Gummadi and Santhosh, 2010) by using Pseudomonas<br />

strains isolated from coffee plantation area. However,<br />

researchers never stop exploring the new strains with<br />

high caffeine tolerance and efficient caffeine degradation<br />

since the strain is the one of most important factors for<br />

developing caffeine biodegradation technique. In this<br />

study, twenty strains with high caffeine-tolerant capability<br />

were isolated from the caffeine-containing soil of tea<br />

garden, and two of them were identified as Pseudomonas<br />

putida based on the physiological-biochemical tests and<br />

16S rRNA gene analysis. Effect of caffeine concentration<br />

and other culture conditions on the strain growth was also<br />

investigated.<br />

MATERIALS AND METHODS<br />

Preparation mediums for screening and amplifying the<br />

caffeine-tolerant bacteria<br />

Solution with mineral substances, including KCl (0.370 g L -1 ),<br />

MgSO4 (0.205 g L -1 ), Fe2(SO4)3 (0.710 g L -1 ), Na2HPO4·12H2O<br />

(0.205 g L -1 ), CaCl2 (80 mg L -1 ), ZnSO4·7H2O (15 g L -1 ),<br />

NaMoO4·2H2O (12 g L -1 ), MnSO4 (11 g L -1 ), CuSO4 (10 g L -1 )<br />

and H3BO4 (10 g L -1 ), was prepared and centrifuged (Beckman J2<br />

HS; Beckman Instruments Inc., Fullerton, CA, USA) at 5000 ×g for<br />

10 min after its pH value was adjusted to 7.0. The supernatant was<br />

collected and used for preparation of different culture mediums.<br />

Solid screening medium (SSM) for isolating the caffeine-tolerant<br />

bacteria was prepared by mixing the mineral solution with caffeine<br />

(2.5 g L -1 ) and agar (1.5%) and autoclaved at 121°C for 10 min.<br />

Solid purifying medium (SPM) was also prepared as SSM except<br />

20.0 g L -1 caffeine was supplemented. Liquid amplifying medium<br />

(LAM) was obtained after addition of caffeine (0.5 g L -1 ) and<br />

sucrose (5.0 g L -1 ) in the mineral solution and disinfection.<br />

Isolation, purification and amplification of caffeine-tolerant<br />

bacteria<br />

Soils were collected randomly from experimental tea garden of<br />

Zhejiang University. Some of them (100 g) were soaked in 1 L<br />

distilled water for 30 min. Solution containing microorganisms were


2280 Afr. J. Microbiol. Res.<br />

obtained by filtration and diluted 10 3 -10 6 times, and spread over the<br />

SSM surface in Petri dishes (0.5 ml solution for each dish). After<br />

incubation at 30°C for 2-3 d, the fast-growing single colony was<br />

picked up with tooth tip and inoculated on SPM surface for further<br />

purification. Twenty colonies, which could grow normally on the<br />

SPM, were separately picked up again and transferred to the tubes<br />

containing 1.0 ml LAM medium (each colony for one tube), then<br />

incubated at 150 r min -1 and 30°C in a shaking incubator (Taicang<br />

Experimental Instruments Factory, Jiangsu, China) for amplification.<br />

Three days later, 0.3 ml of bacterial solution was mixed with 0.1 ml<br />

dimethyl sulfoxide and stored at -70°C, and the remaining solution<br />

was used for further tests.<br />

Physiological-biochemical identification of caffeine-tolerant<br />

bacteria<br />

Two of the above twenty strains with high-caffeine tolerant<br />

capability, recorded as CT25 and CT75, were randomly selected for<br />

physiochemical identification. The capability to metabolize carboncontaining<br />

compounds was assessed by glycolysis test, methyl red<br />

test (MR) and Voges-Proskauer test (VP), citrate utilization test,<br />

oxidase activity and starch hydrolysis test. The activity of<br />

metabolizing nitrogen-containing compounds was assessed by<br />

indole production, hydrogen sulfide production, urease production<br />

tests and gelatin liquefaction test. In order to make further<br />

determination of the objective strains, the arginine dihydrolase<br />

activity test was also performed. The identification was carried out<br />

in detail according to the “Bergey's Manual of Determinative<br />

Bacteriology” (Buchanan and Gibbons, 2000).<br />

16S rRNA gene identification of caffeine-tolerant bacteria<br />

Amplified caffeine-tolerant bacteria solution (0.5 ml) of strain CT25<br />

and CT75 was centrifuged at 2000 ×g for 5 min and washed with<br />

PBS buffer, and suspended in distilled water (0.1 ml). Degenerate<br />

primers (forward primer: 5’-agagtttgatcmtggctcag; reverse primer:<br />

5’-ggytaccttgttacgactt, predicted length 1498 bp) were used to<br />

amplify the partial 16S rRNA gene fragments. PCRs were carried<br />

out in a 25 l mixture containing 17.3 l double-distilled water, 2.5<br />

l 10×PCR buffer, 1.5 l 25 mM MgCl2, 0.5 l 10 mM dNTPs, 0.5 l<br />

each primer, 0.2 l Taq DNA polymerase (2.5 U l -1 ) and 2 l strain<br />

resuspension. Amplification was performed on Veriti 96-well Fast<br />

Thermal Cycler (Applied Biosystems, Inc., Carlsbad, CA, USA)<br />

according to the following time program: predenaturation at 94°C<br />

for 5 min, 1 cycle; denaturation at 94°C for 45 s, annealing at 50°C<br />

for 60 s and extension at 72°C for 90 s, 30 cycles; and final<br />

extension at 72°C for 10 min, 1 cycle. Partial amplified solutions (10<br />

l) were subjected to electrophoresis for checking presence of the<br />

target band, the remaining was sent to Invitrogen (Shanghai) Life<br />

Science Corporation for sequencing. The obtained sequences were<br />

deposited in GenBank (www.ncbi.nlm.nih.gov) after blasting with<br />

known sequences by using the tool of “blastn”.<br />

Effect of culture conditions on the growth of isolated strain<br />

For evaluating the effect of oxygen, the amplified bacteria (50 l) of<br />

strain CT25 was inoculated in a conical flask containing 50 ml<br />

mineral solution supplemented with 2.0 g L -1 caffeine, and<br />

incubated in a shaking incubator at 150 r min -1 and 25ºC for 30-72<br />

h. Static culture (without shaking) was also carried out as control.<br />

For evaluating the effect of nitrogen source, the CT25 strain was<br />

inoculated in a mineral medium supplemented with 2.0 g L -1<br />

peptone instead of 2.0 g L -1 caffeine, and incubated at 150 r min -1<br />

and 25°C for 14 to 42 h.<br />

For evaluating the effect of caffeine concentration, the CT25<br />

strain was incubated in mineral medium with supplement of different<br />

concentration caffeine (0.1, 0.5, 1.0, 2.0, 5.0, 10.0 and 20.0 g L -1 ) at<br />

150 r min -1 and 25°C for 30 h.<br />

For evaluating the effect of temperature, the CT25 strain was<br />

inoculated in a caffeine-supplemented (2.0 g L -1 ) medium and<br />

shook (150 r min -1 ) at different temperature (20, 25, 30 and 35°C)<br />

for 12-72 h.<br />

After incubation, the optical density of bacterium solutions was<br />

measured at 600 nm (OD600) with a HP8453 UV-visible<br />

spectrophotometer (Hewlett Packard, CA, USA), and the residue<br />

level of caffeine in caffeine supplemented medium was also<br />

monitored by HPLC (LC20A, Shimadzu Corporation, Tokyo, Japan)<br />

method as described previously (Liang et al., 2007).<br />

Data analysis<br />

Observations were made in triplicates and the mean ± standard<br />

deviation (SD) was presented. Statistics was carried out on Version<br />

8.1 SAS System for Windows (SAS Institute Inc, Cary, NC, USA)<br />

software.<br />

RESULTS<br />

The isolated high caffeine-tolerant bacteria<br />

After screening, a total of 103 colonies were obtained<br />

based on rapid proliferation on the SSM medium<br />

containing 2.5 g L -1 caffeine. These colonies exhibited a<br />

similar appearance, circular in shape and ivory-white in<br />

colour. Twenty strains of them could grow normally but<br />

somewhat slowly on the SPM medium supplemented with<br />

20.0 g L -1 caffeine after further purification, and were<br />

recorded as CT03, CT12, CT25, CT26, CT33, CT38,<br />

CT39, CT42, CT45, CT47, CT55, CT57, CT66, CT70,<br />

CT75, CT78, CT79, CT80, CT94 and CT97 respectively.<br />

These strains can be barely differentiated from each<br />

other according to their appearances and growths,<br />

suggesting they might belong to a same genus.<br />

The characteristics of high caffeine-tolerant bacteria<br />

A series of determinative tests were carried out for<br />

identifying the randomly selected caffeine-tolerant strains<br />

(CT25 and CT75) because different genera have specific<br />

metabolic characteristics and different capacities to utilize<br />

amino acids, carbohydrates and proteins (Harley, 2008).<br />

Table 1 summarized the physiological-biochemical<br />

characteristics of the strains CT25 and CT75. Both CT25<br />

and CT75 were preliminary identified as Pseudomonas<br />

putida after comparison with Bergey’s Manual of<br />

Determinative Bacteriology (Buchanan and Gibbons,<br />

2000).<br />

The partial gene (1498bp) of 16S rRNA genes from<br />

strains of CT25 and CT75 were amplified and sequenced<br />

(Figures 1 and 2), then deposited in GenBank<br />

(GU828029 for CT25 and GU828030 for CT75).


Table 1. Physiological-biochemical characteristics of caffeine-tolerant bacteria*.<br />

Items CT25 CT75<br />

Glucose fermentation + +<br />

Lactose fermentation + +<br />

VP & MR - -<br />

Citrate utilization test - -<br />

Starch hydrolysis - -<br />

Oxidase + +<br />

Indole production - -<br />

Hydrogen sulfide production - -<br />

Gelatin hydrolysis - -<br />

Urease + +<br />

Nitrate reduction + +<br />

Arginine dihydrolase + +<br />

“+” for positive and “-” for negative.<br />

Figure 1. Partial sequence of 16S rRNA gene from CT25 strains (GU828029).<br />

Homology search with the tool of “blastn” showed that the<br />

sequences of 16S rRNA gene from CT25 and CT75 were<br />

99% identical with corresponding sequences of<br />

Pseudomonas putida (Table 2). The result confirmed the<br />

previous identification on the basis of physiologicalbiochemical<br />

characteristics, therefore, the isolated twenty<br />

strains, including randomly selected CT25 and CT75,<br />

were the Pseudomonas putida strains with high caffeinetolerant<br />

capability.<br />

Fan et al. 2281<br />

The growth of high caffeine-tolerant bacteria<br />

Incubation results showed that the OD600 value of CT25<br />

strain under shaking culture condition was significantly<br />

higher than that undern static culture condition (Table 3),<br />

suggesting the strain CT25 is an aerobe and its growth<br />

depends obviously on oxygen concentration in medium.<br />

Different source of N and C test showed that the OD600<br />

value of CT25 cultured in mineral medium with addition of


2282 Afr. J. Microbiol. Res.<br />

Figure 2. Partial sequence of 16S rRNA gene from CT75 strains (GU828030).<br />

Table 2. Blast result of 16S rRNA gene from CT25 and CT75.<br />

Accession Description E-Value Consistency (%)<br />

GU828029 Pseudomonas putida strain 25 16S ribosomal RNA gene, partial sequence Sequence from CT25<br />

GU828030 Pseudomonas putida strain 75 16S ribosomal RNA gene, partial sequence Sequence from CT75<br />

CP002290 Pseudomonas putida BIRD-1, complete genome 0.0 99<br />

CP000926 Pseudomonas putida GB-1, complete genome 0.0 99<br />

AE015451 Pseudomonas putida KT2440 complete genome 0.0 99<br />

CP000712 Pseudomonas putida F1, complete genome 0.0 99<br />

AM411058 Pseudomonas putida partial 16S rRNA gene, strain 5zhy 0.0 99<br />

DQ657850 Pseudomonas sp. ND9 16S ribosomal RNA gene, partial sequence 0.0 99<br />

Table 3. Effect of oxygen on growth of CT25 in the medium with<br />

2.0gL -1 caffeine * .<br />

Items 30 h 72 h<br />

Static culture 0.0633±0.0034 a 0.0634±0.0067 a<br />

Shaking culture 0.1461±0.0032 b 0.2827±0.0029 b<br />

* The OD600 was used as growth indicator of bacterium, and the<br />

different letters in column indicated significant difference at p


600<br />

OD<br />

Table 4. Comparison of CT25 growth cultured with different sources for nitrogen and carbon * .<br />

Nitrogen source 14 h 30 h 42 h<br />

Caffeine (2.0g L -1 ) 0.0305±0.0012a 0.1461±0.0032a 0.2514±0.0033a<br />

Peptone (2.0g L -1 ) 0.8911±0.0045b 1.0084±0.0049b 1.3896±0.0174b<br />

* The OD600 was used as growth indicator of bacterium, and the different letters in column indicated significant<br />

difference at p


2284 Afr. J. Microbiol. Res.<br />

Percentage of residual caffeine in %)<br />

110<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

%)<br />

30 C<br />

35°C<br />

0 20 40 60 80<br />

Time (h)<br />

Figure 5. Caffeine-degrading efficiency of CT25 cultured in medium<br />

supplemented with 2.0 g L-1 initial caffeine at different temperature.<br />

The bar shows the standard deviation.<br />

DISCUSSION<br />

The isolated twenty strains could grow normally on the<br />

medium supplemented with 20 g L -1 caffeine, indicating<br />

these strains can tolerate high concentration of caffeine<br />

and metabolize it as sole nitrogen (N) and carbon (C) for<br />

their growth demand since no other source of N and C<br />

was present in that medium. Previous reports showed<br />

that some Pseudomonas sp. strains isolated from coffee<br />

plantation area could survive in the mediums<br />

supplemented with caffeine at initial concentration of 10 g<br />

L -1 (Gokulakrishnan and Gummadi, 2006; Gummadi and<br />

Santhosh, 2006; Dash and Gummadi, 2008), 15 g L -1<br />

(Dash and Gummadi, 2007; Dash et al., 2008) and 20 g<br />

L -1 (Gummadi et al., 2009b; Gummadi and Santhosh,<br />

2010) and degrade the caffeine sufficiently in short<br />

period. From this view, the isolated strains in this study<br />

might possess similar high caffeine-tolerant capability to<br />

the reported ones although some investigation should be<br />

further conducted with these strains. It was also<br />

suggested that soil microorganisms have the ability for<br />

natural enrichment under different ecological condition<br />

according to their chemotaxis (Mazzafera, 2002). High<br />

caffeine-tolerant bacteria can be isolated from caffeinecontaining<br />

soils such as tea and coffee garden where<br />

caffeine is continuously released from the litters of these<br />

plants.<br />

Series physiological-biochemical reactions (Buchanan<br />

and Gibbons, 2000) and the sequence of 16S rRNA gene<br />

(Garrity et al., 2004) are widely used for bacterial<br />

identification. The randomly selected strains (CT25 and<br />

CT75) with high caffeine-tolerant capability were<br />

identified as Pseudomonas putida according to the<br />

similarity of physiochemical characteristics and 16S rRNA<br />

gene sequences. It seemed that all of the isolated twenty<br />

strains might belong to Pseudomonas putida since the<br />

appearance of the colonies was quite similar, and this<br />

species might be a dominant bacterium in soil of tea<br />

garden because special metabolic pathway(s) related to<br />

caffeine degradation were reported to be present<br />

inherently in many Pseudomonas strains (Dash and<br />

Gummadi, 2006b; Mazzafera, 2004; Yamaoka-Yano and<br />

Mazzafera, 1999; Yu et al., 2009).<br />

The growth of strain CT25 was highly depended on<br />

oxygen in liquid medium, which is consistent with other<br />

reports on the growth of Pseudomonas strains (Beltrán et<br />

al., 2006; Gummadi and Santhosh, 2006; Gummadi et<br />

al., 2009b; Middelhoven and Lommen, 1984; Woolfolk,<br />

1975). Although caffeine could be used as sole source for<br />

N and C, the strain CT25 amplified more slowly in<br />

medium supplemented with caffeine than that with<br />

peptone, and its growth was significantly inhibited especially<br />

when high level caffeine was supplemented. Similar<br />

results were reported previously in the studies about<br />

other caffeine-degrading strains (Sarath Babu et al.,<br />

2005; Gokulakrishnan and Gummadi, 2006; Dash and<br />

Gummadi, 2007, 2008; Gummadi et al., 2009b). It was<br />

suggested that the isolated bacteria can utilize the<br />

caffeine as their nonpreferential N and C sources and<br />

may take time to activate their metabolic pathway(s) to<br />

transform the caffeine into some components directly<br />

used for growth. This result agrees with some findings<br />

that the caffeine-degrading enzymes of bacteria were<br />

induced (Dash and Gummadi, 2007; Gummadi and<br />

Santhosh, 2006; Sarath Babu et al., 2005; Ogunseitan,<br />

2002; Woolfolk, 1975). The fastest amplification was<br />

observed when the strain CT25 was incubated at around<br />

30°C, while amplification would dramatically decrease<br />

when the strain was incubated at 35°C, which coincides<br />

with the previous observations on the growth of other<br />

Pseudomonas strains (Dash and Gummadi, 2007; Sarath<br />

Babu et al., 2005) and the optimum catalytic temperature<br />

of caffeine-degrading enzyme (Beltrán et al., 2006;<br />

Yamaoka-Yano and Mazzafera, 1999). However, this<br />

result is not in accordance with another report in which<br />

the optimum temperature for the growth of Pseudomonas<br />

sp. strain was 35°C (Gummadi and Santhosh, 2006). The<br />

difference might be explained as different strain or other<br />

culture parameters such as caffeine concentration.<br />

Caffeine was metabolized rapidly when the CT25 was<br />

incubated at 30°C, while slowly at 35°C (Figures 4 and<br />

5), indicating that the caffeine degradation was positively<br />

correlated with the bacterial growth as more bacterial<br />

cells would metabolize much more caffeine into nutrition<br />

for further bacterial growth. Thus, in order to improve the<br />

decaffeination efficiency, the initial inoculums concentration<br />

should be optimized. Compared with the caffeine<br />

residue, metabolites of caffeine were barely detected in<br />

the incubated medium (Figure 4), suggesting that initial<br />

degradation of caffeine might be a rate-limiting step and


the metabolites might be instantly broken down<br />

furthermore.<br />

The bacteria isolated from soil of tea garden (such as<br />

CT25) have high capability for caffeine-tolerance and<br />

caffeine-degradation, and can be used as an alternative<br />

for decaffeination of tea products or caffeine-containing<br />

wastes. Further studies like usage method<br />

(immobilization or not), optimization of decaffeination<br />

efficiency and assessment of products quality and<br />

security, should be conducted prior to exploitation of the<br />

bacterial strains for caffeine degradation purpose.<br />

ACKNOWLEDGMENT<br />

This study was financed by the National Natural Science<br />

Foundation of China and “R&D Project” of Education<br />

Office of Zhejiang Province.<br />

REFERENCES<br />

Arciero PJ, Ormsbee M J (2009). Relationship of blood pressure,<br />

behavioral mood state, and physical activity following caffeine<br />

ingestion in younger and older women. Appl. Physiol. Nutr. Metab.,<br />

34: 754-762.<br />

Beltrán JG, Leask RL, Brown WA (2006). Activity and stability of<br />

caffeine demethylases found in Pseudomonas putida IF-3. Biochem.<br />

Eng. J., 31: 8-13.<br />

Bezerra JP, da Silva LR, de Alvarenga Lemos VA, Duarte PM, Bastos<br />

MF (2008). Administration of high doses of caffeine increase alveolar<br />

bone loss in ligature-induced periodontitis in rats. J. Periodontol., 79:<br />

2356-2360.<br />

CARE Study Group (2008). Maternal caffeine intake during pregnancy<br />

and risk of fetal growth restriction: a large prospective observational<br />

study. BMJ, 337: a2332.<br />

Buchanan RE, Gibbons NE (2000). Bergey's manual of determinative<br />

bacteriology. Williams & Wilkins Co., Baltimore.<br />

Buerge IJ, Poiger T, Muller MD, Buser HR (2003). Caffeine, an<br />

anthropogenic marker for wastewater contamination of surface<br />

waters. Environ. Sci. Technol., 37: 691-700.<br />

Chen YH, Huang YH, Wen CC, Wang Y H, Chen WL, Chen LC, Tsay<br />

HJ (2008). Movement disorder and neuromuscular change in<br />

zebrafish embryos after exposure to caffeine. Neurotoxicol. Teratol.,<br />

30: 440-447.<br />

Dash SS, Gummadi SN (2006a). Biodegradation of caffeine by<br />

Pseudomonas sp. NCIM 5235. Res. J. Microbiol., 1: 115-123.<br />

Dash SS, Gummadi SN (2006b). Catabolic pathways and<br />

biotechnological applications of microbial caffeine degradation.<br />

Biotechnol. Lett., 28: 1993-2002.<br />

Dash SS, Gummadi SN (2007). Degradation kinetics of caffeine and<br />

related methylxanthines by induced cells of Pseudomonas sp. Curr.<br />

Microbiol., 55: 56-60.<br />

Dash SS, Gummadi SN (2008). Inducible nature of the enzymes<br />

involved in catabolism of caffeine and related methylxanthines. J.<br />

Basic Microbiol., 48: 227-233.<br />

Dong ZB, Liang YR, Fan FY, Ye JH, Zheng XQ, Lu JL (2011).<br />

Adsorption behavior of the catechins and caffeine onto<br />

polyvinylpolypyrrolidone. J. Agric. Food Chem., 59: 4238–4247.<br />

Duarte PM, Marpues MR, Bezerra JP, Bastos MF (2009). The effects of<br />

caffeine administration on the early stage of bone healing and bone<br />

density: A histometric study in rats. Arch. Oral. Biol., 54: 717-722.<br />

Garrity GM, Bell JA, Lilburn TG (2004). Bergey’s manual of systematic<br />

bacteriology. Springer, New York. Gokulakrishnan S, Chandraraj K,<br />

Gummadi SN (2005). Microbial and enzymatic methods for the<br />

removal of caffeine. Enzyme Microb. Technol., 37: 225-232.<br />

Gokulakrishnan S, Gummadi SN (2006). Kinetics of cell growth and<br />

Fan et al. 2285<br />

caffeine utilization by Pseudomonas sp. GSC 1182. Process Biochem.,<br />

41: 1417-1421.<br />

Gokulakrishnan S, Chandraraj K, Gummadi SN (2007). A preliminary<br />

study of caffeine degradation by Pseudomonas sp. GSC 1182. Int. J.<br />

Food Microbiol., 113: 346-350.<br />

Gummadi SN, Santhosh D (2006). How induced cells of Pseudomonas<br />

sp. increase the degradation of caffeine. Cent. Eur. J. Biol., 1: 561-<br />

571.<br />

Gummadi SN, Santhosh D (2010). Kinetics of growth and caffeine<br />

demethylase production of Pseudomonas sp. in bioreactor. J. Ind.<br />

Microbiol. Biotechnol., 9: 901-908.<br />

Gummadi SN, Dash SS, Devarai S (2009a). Optimization of production<br />

of caffeine demethylase by Pseudomonas sp. in a bioreactor. J. Ind.<br />

Microbiol. Biotechnol., 36: 713-720.<br />

Gummadi SN, Ganesh KB, Santhosh D (2009b). Enhanced degradation<br />

of caffeine by immobilized cells of Pseudomonas sp. in agar-agar<br />

matrix using statistical approach. Biochem. Eng. J., 44: 136-141.<br />

Gutierrez-Sanchez G, Roussos S, Augur C (2004). Effect of the<br />

nitrogen source on caffeine degradation by Aspergillus tamarii. Lett.<br />

Appl. Microbiol., 38: 50-55.<br />

Hakil M, Denis S, Viniegra-Gonzalez G, Augur C (1998). Degradation<br />

and product analysis of caffeine and related dimethylxanthines by<br />

filamentous fungi. Enzyme Microb. Tech., 22: 355-359.<br />

Hallstrom T, Wolk A, Glynn A, Michaelsson K (2006). Coffee, tea and<br />

caffeine consumption in relation to osteoporotic fracture risk in a<br />

cohort of Swedish women. Osteoporo.s Int., 17: 1055–1064.<br />

Harley JP (2008). Laboratory exercises in microbiology (7 th ed.).<br />

McGraw-Hill Co., New York.<br />

Heckman MA, Weil J, Mejia G (2010). Caffeine (1, 3, 7trimethylxanthine)<br />

in foods: a comprehensive review on consumption,<br />

functionality, safety, and regulatory matters. J. Food Sci., 75: 77-87.<br />

Kato M, Mizuno K, Crozier A, Fujimura T, Ashihara H (2000). Plant<br />

biotechnology: caffeine synthase gene from tea leaves. Nature, 406:<br />

956-957.<br />

Kurtzman RH, Schwimmer S (1971). Caffeine removal from growth<br />

media by microorganism. Experientia, 27: 481-482.<br />

Liang HL, Liang YR, Dong JJ, Lu JL, Xu HR, Wang H (2007).<br />

Decaffeination of fresh green tea leaf (Camellia sinensis) by hot<br />

water treatment. Food Chem., 101: 1451-1456.<br />

Lu JL, Wu MY, Yang XL, Dong ZB, Ye JH, Borthakur D, Sun QL, Liang<br />

YR (2010). Decaffeination of tea extracts by using poly (acrylamideco-ethylene<br />

glycol dimethylacrylate) as adsorbent. J. Food Eng., 97:<br />

555-562.<br />

Lueth NA, Anderson KE., Harnack LJ, Fulkerson JA, Robien K (2008).<br />

Coffee and caffeine intake and the risk of ovarian cancer: the Iowa<br />

women’s health study. CCC, 19: 1365-1372.<br />

Mazzafera P, Olsson O, Sandberg G (1996). Degradation of caffeine<br />

and related methylxanthines by Serratia marcescens isolated from<br />

soil under coffee cultivation. Microb. Ecol., 31: 199-207.<br />

Mazzafera P (2002). Degradation of caffeine by microorganisms and<br />

potential use of decaffeinated coffee husk and pulp in animal feeding.<br />

Sci. Agric., 59: 815-821.<br />

Mazzafera P (2004). Catabolism of caffeine in plants and<br />

microorganisms. Front. Biosci., 9: 1348-1359.<br />

Middelhoven WJ, Lommen A (1984). Degradation of caffeine by<br />

Pseudomonas putida C3024: the effect of oxygen concentration. J.<br />

Microbiol., 50: 298-300.<br />

Moisey LL, Kacker S, Bickertom AC, Robinson LE, Graham TE (2008).<br />

Caffeinated coffee consumption impairs blood glucose homeostasis<br />

in response to high and low glycemic index meals in healthy men.<br />

Am. J. Clin. Nutr., 87: 1254-1261.<br />

Moore MT, Greenway SL, Farris JL (2008). Assessing caffeine as an<br />

emerging environmental concern using conventional approaches.<br />

Arch. Environ. Contam. Toxicol., 54: 31-35.<br />

Ogunseitan OA (1996). Removal of caffeine in sewage by<br />

Pseudomonas putida: implications for water pollution index. World J.<br />

Microbiol. Biotechnol., 12: 251-256.<br />

Ogunseitan OA (2002). Caffeine-inducible enzyme activity in<br />

Pseudomonas putida ATCC 700097. World J. Microbiol. Biotechnol.,<br />

18: 423-428.<br />

Padmanabhan P, Padmanabhan S, Derito C, Gray A, Gannon D, Snape<br />

JR, Tsai CS, Park W, Jeon C, Madsen EL (2003). Respiration of 13 C-


2286 Afr. J. Microbiol. Res.<br />

labeled substrates added to soil in the field and subsequent 16S rRNA<br />

gene analysis of 13 C-labeled soil DNA. Appl. Environ. Microbiol., 69:<br />

1614-1622.<br />

Pandey A, Soccol CR, Nigam P, Brand D, Mohan R, Roussos S (2000).<br />

Biotechnological potential of coffee pulp and coffee husk for<br />

bioprocesses. Biochem. Eng. J., 6: 153-162.<br />

Panza VSP, Wazlawik E, Schütz GR, Comin L, Hecht KC, Silva EL<br />

(2008). Consumption of green tea favorably affects oxidative stress<br />

markers in weight-trained men. Nutrition, 24: 433-442.<br />

Pollack K, Balazs K, Ogunseitan O (2009). Proteomic assessment of<br />

caffeine effects on coral symbionts. Environ. Sci. Technol., 43: 2085-<br />

2091.<br />

Ramarethinam S, Rajalakshmi N (2004). Caffeine in tea plants<br />

[Camellia sinensis (L.) O. Kuntze]: in situ lowering by Bacillus<br />

licheniformis (Weigmann) Chester. Indian J. Exp. Biol., 42: 575-580.<br />

Roehrs T, Roth T (2008). Caffeine: sleep and daytime sleepiness. Sleep<br />

Med. Rev., 12: 153-162.<br />

Sarath Babu VR, Patra S, Thakur MS, Karanth NG, Varadaraj MC<br />

(2005). Degradation of caffeine by Pseudomonas alcaligenes CFR<br />

1708. Enzyme Microb. Technol., 37: 617-624.<br />

Sun QL, Shu H, Ye JH, Lu JL, Zheng XQ., Liang YR (2010).<br />

Decaffeination of green tea by supercritical carbon dioxide. J. Med.<br />

Plants Res., 4: 1161-1168<br />

Tagliari CV, Sanson RK, Zanette A, Franco TT, Soccol CR (2003).<br />

Caffeine degradation by Rhizopus delemar in packed bed column<br />

bioreactor using coffee husk as substrate. Braz. J. Microbiol., 34:<br />

102-104.<br />

Topp E, Hendel JG, Lu ZX, Chapman R (2006). Biodegradation of<br />

caffeine in agricultural soils. Can. J. Soil Sci., 86: 533-544.<br />

Weng XW, Odouli R, Li DK (2008). Maternal caffeine consumption<br />

during pregnancy and the risk of miscarriage: a prospective cohort<br />

study. Am. J. Obstet. Gynecol., 198: 279.e1 - 279.e8.<br />

Woolfolk CA (1975). Metabolism of N-methylpurines by a Pseudomonas<br />

putida strain isolated by enrichment on caffeine as sole source of<br />

carbon and nitrogen. J. Bacteriol., 123: 1088-1106.<br />

Yadav SK, Ahuja PS (2007). Towards generating caffeine-free tea by<br />

metabolic engineering. Plant Foods Hum. Nutr., 62: 185-191.<br />

Yamaoka-Yano DM, Mazzafera P (1999). Catabolism of caffeine and<br />

purification of a xanthine oxidase responsible for methyluric acids<br />

production in Pseudomonas putida L. Rev. Microbiol., 30: 62-70.<br />

Ye JH, Jin J, Liang HL, Lu JL, Du YY, Zheng XQ, Liang YR (2009).<br />

Using tea stalk lignocellulose as an adsorbent for separating<br />

decaffeinated tea catechins. Bioresour. Technol., 100: 622-628.<br />

Yu CL, Kale Y, Gopishetty S, Louie TM, Subramanian M (2008). A<br />

novel caffeine dehydrogenase in Pseudomonas sp. strain CBB1<br />

oxidizes caffeine to trimethyluric acid. J. Bacteriol., 190: 772-776.<br />

Yu C L, Louie TM, Summers R, Kale Y, Gopishetty S, Subramanian M<br />

(2009). Two distinct pathways for metabolism of theophylline and<br />

caffeine are coexpressed in Pseudomonas putida CBB5. J.<br />

Bacteriol., 191: 4624-4632.


African Journal of Microbiology Research Vol. 5(16), pp. 2287-2296, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.613<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Biopreservative potential of marine lactobacillus spp<br />

K. Indira*, S. Jayalakshmi, A. Gopalakrishnan and M. Srinivasan<br />

Centre of Advanced Study in Marine Biology, Faculty of Marine Sciences,<br />

Annamalai University Parangipettai – 608 502, Tamil Nadu – India.<br />

Accepted 30 July, 2011<br />

Bacteriocins are of special interest due to their potential value as natural preservative. The present<br />

study is a trial for production of such bacteriocin from a marine Lactobacillus sp. Lactic acid bacteria<br />

(LBA) was isolated from fish gut (Mugil cephalus) and prawn muscle (Peneaus monodon) samples and<br />

their density were found to be 5.2 x 10 7 and 6.4 x 10 7 CFU/g respectively. Various pathogens were<br />

isolated from ready to eat pickle samples (bottled). The LAB strains were tested against 10 different<br />

commercial antibiotics. Among them Vancomycin was the only antibiotic that showed a minimum of<br />

40% resistance to the LAB strains tested. LAB strains were optimized at different parameters and<br />

maximum bacteriocin production was at pH 6, temperature of 35°C, 3.5% of salt concentration, 24th h of<br />

incubation period. Bacteriocin produced by these strains were precipitated from the culture filtrate<br />

using methanol and TCA which was further dialyzed, centrifuged and lyophilized. Lactobacillus<br />

fermentum was selected as the most potential strain for both bacteriocin production as well as<br />

antimicrobial activity. sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis<br />

proved that the obtained protein is homologous with a molecular weight of 18 kDa. FT-IR spectrum also<br />

confirmed the obtained protein as a bacteriocin. The study revealed that Lactobacillus strains of marine<br />

origin are having the potential to be used as biopreservatives especially in seafood industries. The<br />

production of bacteriocin from L. fermentum was found to be ideal for industrial scale production and<br />

commercial utilization.<br />

Key words: Bacteriocin, Lactobacillus fermentum, biopreservative, fourier transform infrared (FT-IR).<br />

INTRODUCTION<br />

Food fermentation has a great economic value and the<br />

products obtained through this process put in improving<br />

human health. In this way LAB has contributed in a lot to<br />

fermented foods worldwide. The major genera of LAB is<br />

of importance in the food industry are Bifidobacterium,<br />

Enterococcus, Lactobacillus, Lactococcus, Leuconostoc,<br />

Oenococcus, Pediococcus, Streptococcus and<br />

Tetragenococcus. They are significant in production and<br />

preservation of milk and dairy products, meat and meat<br />

products, vegetable products and as forage crops for<br />

animals (Lee, 2004).<br />

Mankind had exploited lactic acid bacteria (LAB) for the<br />

production of fermented foods because of their ability to<br />

produce desirable changes in taste, flavor and texture as<br />

well as to inhibit pathogenic and spoilage microbes.<br />

Since they are involved in numerous food fermentations<br />

*Corresponding author. E. mail: microind03@yahoo.co.in. Tel:<br />

04144 – 243070 – 243071. Fax: 04144-243555.<br />

for millennia, it is assumed that most representatives of<br />

this group do not pose any health risk to man and are<br />

designated as GRAS (generally recognized as safe)<br />

organisms (Holzapfel et al., 1995). Different antimicrobials,<br />

such as lactic acid, acetic acid, hydrogen peroxide,<br />

carbon dioxide and bacteriocins produced by these<br />

bacteria, can inhibit pathogenic and spoilage microorganisms,<br />

extending the shelf-life and enhancing the<br />

safety of food products (Aymerich et al., 2000).<br />

One important attribute of LAB is their ability to produce<br />

antimicrobial compound called bacteriocin. Bacteriocins<br />

are proteinaceous compound, inhibitory effects towards<br />

sensitive strains produced by both Gram-positive and<br />

Gram-negative bacteria (Tagg et al., 1976). Bacteriocins<br />

producing lactic acid bacteria are used in food fermentations<br />

especially in dairy products. In USA, only nisin<br />

produced by Lactobacillus lactis has been permitted as a<br />

food preservative (FDA, 1988). It has also been used in<br />

health care products and cosmetics for treatment of acne.<br />

They are also being used in toothpaste and mouthwash<br />

for the inhibition of dental caries and periodontal diseases


2288 Afr. J. Microbiol. Res.<br />

(Harlander, 1993). Bacteriocins can be exploited to inhibit<br />

undesirable microorganisms in the fermentation of wine<br />

(Navarro et al., 2000), beer (Ogden et al., 1985), vegetables<br />

(Daeschel and Fleming, 1984) and dairy products<br />

(Castla et al., 1996; Ross et al., 1999). The present study<br />

is on bacteriocin production by LAB of marine environment<br />

and its use as a biopreservatives in sea foods.<br />

MATERIALS AND METHODS<br />

The fin fish and prawn samples were collected from the Annan koil<br />

landing centre (Latitude 11° 29 ’N; Longitude 79° 46’ E) for the<br />

isolation of lactic acid bacteria. The pickle samples were collected<br />

from Tuticorin. Tuna and Prawn pickles were analyzed in the<br />

present study. Homogenized extract of the fish and prawn samples<br />

were serially diluted up to 10 -5 dilutions and were plated on MRS<br />

agar (HI-MEDIA) by spread plate technique and incubated at 28 ±<br />

2°C for 24 h. The isolated potential strains were identified based on<br />

Bergey’s manual of systematic bacteriology (Buchanan et al.,<br />

1974). Approximately 10 g of sample (pickle) was homogenized in a<br />

sterile mortar and pestle using 90 ml of sterile 50% sea water and<br />

then serially diluted. About 0.1ml of the serially diluted samples was<br />

inoculated into the selective media to isolate the specific<br />

pathogens. The isolated pathogens were identified based on<br />

Bergey’s manual of systematic bacteriology (Buchanan et al.,<br />

1974).<br />

Primary screening for antibacterial activity<br />

A preliminary test for inhibitory assay of LAB against food borne<br />

pathogens, isolated from the seafood pickles was done.<br />

Well diffusion method (Reinheimer et al., 1990)<br />

After swabbing the pathogens on the Muller Hinton agar plates, 0.1<br />

ml of cell free culture broth of Lactic acid bacteria (LAB) centrifuged<br />

at 10,000 rpm for 20 min, poured into the wells and plates were<br />

incubated at 37°C for 24 h. The bacterial culture filtrate inhibiting<br />

the growth of pathogen was assessed based on the inhibition zone<br />

around the well and the results were recorded.<br />

Test for antibiotic resistance of LAB<br />

Muller Hinton Agar was used to check the antibiotic resistance of<br />

LAB against antibiotics like Ampicillin (A), Bacitracin (B),<br />

Cephotaxime (Ce), Ciprofloxacin (Cf), Erythromycin(E), Nalidixic<br />

acid (Na), Novobiocin (Nv), Penicillin-G(P), Tetracyclin (T) and<br />

Vancomycin (Va). After swabbing the LAB on MHA plates, the<br />

selected antibiotic discs were placed onto the agar medium and the<br />

incubated plates were observed for antibiotic assay.<br />

Screening of bacteriocin producing bacteria<br />

Screening was done by well diffusion method using the crude<br />

extract. The inhibitory activity was tested against the seafood borne<br />

pathogens.<br />

Optimization of cultural condition for bacteriocin production<br />

The effect of incubation period on growth and bacteriocin<br />

production was studied for 0 - 52h with 12 h intervals. The pH<br />

ranges (3, 4, 5, 6 and 7) were tested. A temperature range 20, 25,<br />

30, 35 and 40°C were optimized to find out the optimum<br />

temperature. The range of 3-5% of salt concentration was<br />

optimized. Based on the results observed in the optimization the<br />

mass scale culture was carried out with the ideal parameters.<br />

Extraction and partial purification of bacteriocins<br />

The precipitation was made by the solvent extraction method. To<br />

the filtrate obtained 70% Methanol and 10% of Tri chloroacetic acid<br />

(TCA) were added in equal proportion and kept for 48 h at room<br />

temperature. This precipitate was dissolved in deionized water and<br />

dialyzed through a 1000 molecular weight cut of dialysis membrane<br />

against deionized water. The dialysis was done for 24 h and then<br />

centrifuged. The precipitate was lyophilized and used for further<br />

analysis.<br />

Protein estimation<br />

The Folin-Ciocalteu phenol method (Lowry et al., 1951) was used<br />

for the estimation of the total protein content in the sample.<br />

FT-IR analysis<br />

In the study of molecular vibrations, Infrared spectroscopy has<br />

contributed more to this field than Raman due to the rapid<br />

developments in Infrared instrumentation (Merritt et al., 1986). The<br />

vibrational spectra can be utilized directly and simply as molecular<br />

“finger prints” to characterize and identify the molecule (Roberts et<br />

al., 1985). The lyophilized bacteriocin sample from LAB6 was<br />

subjected to FT-IR analysis. The IR spectrum of the bacteriocin was<br />

recorded with a perkin-Elmer model 297 IR spectrophotometer. One<br />

part of the extract was mixed with 99 part of dried potassium<br />

bromide and it was scanned between 600-4000 wave number (cm -<br />

1 ) at a speed of 1 micron. and with a programmed slit opening 2x<br />

and air as reference.<br />

RESULTS<br />

The total heterotrophic and lactic acid bacterial count in<br />

fish gut was found to be 2.14 x 10 9 and 5.2 x 10 7 CFU/g<br />

respectively. Similarly, the total heterotrophic and lactic<br />

acid bacterial count in prawn muscle was found to be 2 x<br />

10 9 and 6.4 x 10 7 CFU/g respectively. A total of 116<br />

morphologically distinct strains were isolated from the<br />

serially diluted fish gut and prawn muscle samples. 4<br />

strains from fish gut and 6 strains from prawn muscle<br />

samples were identified as potent strains for bacteriocin<br />

production. After the screening procedures the potential<br />

strains were identified. They are LAB1 Corynebacterium<br />

bovis, LAB2 Corynebacterium xerosis, LAB3<br />

Lactobacillus alimentarius, LAB4 Lactobacillus animalis,<br />

LAB5 Lactobacillus casei, LAB6 Lactobacillus fermentum,<br />

LAB7 Lactobacillus plantarum, LAB8 Micrococcus<br />

varians, LAB9 Staphylococcus epidermidiis, and LAB10<br />

Streptococcus mitis.<br />

Selective media used for the isolation of seafood<br />

pathogens are EMB agar, MRS agar, Listeria isolation<br />

agar, SS agar, TCBS agar, Yersinia identification agar


Figure 1. Antibiotic activity against LAB strains.<br />

Table 1. Antibiotic activity of lactic acid bacteria.<br />

LAB A B Ce Cf E Na Nv P T Va<br />

LAB1 R R R R R R R R R -<br />

LAB2 R R R R R R R R R -<br />

LAB3 R R R R R R R R R -<br />

LAB4 R R R R R R R R R -<br />

LAB5 R R R R R R R R R R<br />

LAB6 R R R I R R R R R R<br />

LAB7 R R R R R R R R R -<br />

LAB8 R R R R R R R R R -<br />

LAB9 R R R R R R R R R R<br />

LAB10 R R R R R R R R R R<br />

*I- intermediate; R- resistant.<br />

Figure 2. Antibacterial activity of bacteriocin<br />

against Vibrio parahaemolyticus<br />

where in the density of E. coli, Lactobacillus spp, Listeria<br />

spp, Salmonella, Vibrio spp and Yersinia spp were<br />

enumerated to be in the fish pickle 1.9x10 4 , 2.7x10 4 ,<br />

2.1x10 4 , 3.2x10 4 , 1.5x10 4 CFU/g. Likewise in the prawn<br />

pickle the pathogens were enumerated to be 1.3x10 4 ,<br />

1.7x10 4 , 2.2 x10 4 , 1.5 x10 4 , 2.8 x10 4 and 1.0 x10 4 CFU/g.<br />

Isolated pathogens were identified as FBP1 E.coli, FBP2<br />

Indira et al. 2289<br />

Lactobacillus vulgaris, FBP3 Listeria monocytogenes,<br />

FBP4 Listeria spp, FBP5 Salmonella spp, FBP6 Shigella<br />

spp, FBP7 Staphylococcus aureus, FBP8 Vibrio cholera,<br />

FBP9 Vibrio parahaemolyticus and FBP10 Yersinia spp.<br />

All the 10 LAB strains isolated were found to be<br />

resistant to Ampicillin (A), Bacitracin (B), Cephotaxime<br />

(Ce), Erythromycin (E), Nalidixic acid (Na), Novobiocin<br />

(Nv), Penicillin-G(P), and Tetracyclin (T) (Figure 1). The<br />

resistance against Ciproflaxacin (Cf) was shown by all<br />

the LAB strains except LAB6, which showed an intermediate<br />

activity. Resistance to Vancomycin (Va) was shown<br />

by LAB 5, LAB 6, LAB 9 and LAB 10, these results are<br />

given in Table 1.<br />

The bacteriocin obtained from LAB6 showed the<br />

maximum zone of inhibition compared to all the other<br />

strains. It showed a zone of inhibition of 7 mm against V.<br />

parahaemolyticus (Figure 2), 6 mm against L.<br />

monocytogenes, Listeria spp. 5 mm against E. coli,<br />

Salmonella spp. S. aureus and Yersinia spp and 4mm<br />

against Shigella spp. Vibrio cholerae and Lactobacillus<br />

vulgaris, whereas the supernatant of the same strain<br />

showed zone of inhibition of 5 mm against Vibrio<br />

paraheamolyticus (Figure 3), 4 mm against L.<br />

monocytogenes, S. aureus and Yersinia spp and 3 mm<br />

against E. coli, Listeria spp, Salmonella spp, Vibrio


2290 Afr. J. Microbiol. Res.<br />

Figure 3. Antibacterial activity of LAB strains against Vibrio parahaemolyticus.<br />

Figure 4. Antibacterial activity of LAB cell free extracts (Zone of clearance in mm).<br />

cholerae and Lactobacillus vulgaris. These results are<br />

given in Figures 4 and 5 respectively.<br />

The bacteriocin production was found maximum at an<br />

incubation period of 24 h. The results were depicted in<br />

(Figure 6). Among the various pH ranges studied, the<br />

growth of the potential strain was found to be highest at<br />

pH 6 (Figure 7). The optimum temperature for bacteriocin<br />

production was found to be 30 - 35°C (Figure 8). The<br />

optimum salt concentration for bacteriocin production was<br />

found to be 3.5% salt concentration (Figure 9).<br />

The amount of protein was estimated to be 0.36 mg/ml.<br />

The FT-IR Spectrum of the protein revealed the presence<br />

of peaks at the wave numbers of 3288, 3090, 2958, 2924<br />

and 2849 cm -1 which indicated the presence of NH, NH3,<br />

CH3, CH2 and CH2 groups respectively. The wave<br />

numbers 1660, 1632, 1589 and 1540 cm -1 indicated the<br />

presence of their bending mode of amide, methyl, and<br />

amide groups respectively. The asymmetric mode of the<br />

NH band occurred at the wave number 827 cm -1 . The<br />

molecular weight of bacteriocin from L. fermentum was<br />

found to be 18 kDa.<br />

DISCUSSION<br />

In the present study the isolation, partial characterization<br />

and activity of bacteriocin produced by L. fermentum was<br />

done. The bacteriocin producing lactic acid bacteria<br />

(LAB) were isolated from the fresh meat of marine fin fish<br />

and shell fish. It is interesting to note that majority of the<br />

Lactobacillus spp. that have been isolated from fresh and<br />

frozen fish/prawns were those species which were commonly<br />

found in animals and human beings (Kandler and<br />

Weiss, 1986). There are only a few reports available on<br />

isolation of LAB from fresh and seawater fish (Cone,<br />

1982; Okafor and Nzeako, 1985). The LAB strains were<br />

evaluated for the production of inhibitory substances<br />

against various food borne pathogens. The pathogens<br />

used in the present were isolated from fish pickle. All the<br />

LAB strains showed a moderate inhibitory activity against<br />

the pathogens isolated from pickle samples. The use of<br />

bacteriocinogenic starter/protective cultures could<br />

improve the quality and increase safety by inhibiting the<br />

food-borne pathogens and spoilage microorganisms.


Figure 5. Activity of bacteriocins against pathogens (Zone of clearance in mm).<br />

Figures 6. Bacteriocin activity at (a)12th h (b) 24th h (c) 36th h (d) 48th h (e) 60th h of incubation period.<br />

Indira et al. 2291


2292 Afr. J. Microbiol. Res.<br />

Figures 7. Bacteriocin activity between pH range of (a) 3, (b) 4, (c) 5, (d) 6 and 7 (e).<br />

Recent outbreaks of emerging pathogens such as L.<br />

monocytogenes that has caused severe illness through<br />

food ingestion have prompted the scientific community to<br />

focus their studies on the anti-Listeria activity of<br />

bacteriocins produced by Lactobacillus and Pediococcus<br />

strains (Todorov et al., 1999; Aymerich et al., 2000;<br />

Messens et al., 2002).<br />

In the present investigation 100% of the isolated LAB<br />

strains were resistance to Ampicillin, Bacitracin,<br />

Cephotaxime, Erythromycin, Nalidixic acid, Novobiocin,<br />

Penicillin-G and T etracyclin. However only 40% of the<br />

strains showed resistance to Vancomycin. Generally<br />

many LABs are resistant to antibiotics. This resistance<br />

attributes are often intrinsic and non transmissible<br />

(Curragh and Collins, 1992). Among antibiotics,<br />

Vancomycin is one of the last antibiotics broadly<br />

efficacious against clinical infections caused by multidrug<br />

resistant pathogens (Johnson et al., 1990).<br />

The effect of incubation period, pH, temperature and<br />

salinity of medium on the production of bacteriocin was<br />

also investigated in all LAB strains. The pH 6, temperature<br />

of 35°C, salinity of 35 ppt, 24 h of incubation was


Indira et al. 2293<br />

Figures 8. Bacteriocin activity between (a) 20°C, (b) 25°C, (c) 30°C, (d) 35°C, and (e) 40°C of temperature ranges with 5°C interval.<br />

found to be the optimal parameters for the most potential<br />

producers of bacteriocin (that is) L. fermentum. According<br />

to Ogunbanwo et al. (2003) the use of constituted<br />

medium at 30°C of incubation temperature, initial pH of<br />

5.5 and 48 to 60 h fostered the best production of<br />

bacteriocin by Lactobacillus brevis OG1 which seemed to<br />

differ from the results of the present study.<br />

L. fermentum produced bacteriocin in high level and the<br />

strain showed the maximum inhibitory activity against<br />

Vibrio parahaemolyticus, L. monocytogenes , Listeria sp.<br />

and S. aureus. Besides, the productions of bacteriocins<br />

having a wide spectrum of antibacterial activity against<br />

seafood borne pathogens like Listeria, Clostridium and<br />

even Gram-negative pathogens like Pseudomonas and<br />

E. coli to employ as biopreservatives. Accordingly L.<br />

acidophilus and L. casei (Stiles and Holzapfel, 1997) may<br />

be of great interest as probiotics strains because of their<br />

ability to adhere to intestinal epithelial cells and being of<br />

human origin.<br />

The protein purification was done by methanol and<br />

trichloroacetic acid precipitation followed by dialysis<br />

against deionised water. Extraction of bacteriocin using<br />

organic solvents indicated that bacteriocin was removed<br />

from the aqueous phase and could be recovered from the<br />

organic phase. This suggested that part of the bacteriocinmolecule<br />

has a hydrophobic character, and shares this<br />

property with most other bacteriocins (Klaenhammer,<br />

1993). In the present study also Trichloroacetic acid


2294 Afr. J. Microbiol. Res.<br />

Figures 9. Bacteriocin activity between (a) 3%, (b) 3.5%, (c) 4%, (d) 4.5% and to (e) 5% of salt concentration with 0.5% of interval.<br />

precipitation at the rate of 70% methanol and 10% tri<br />

chloroacetic acid aggregated bacteriocin from cell free<br />

broth and the antibacterial activity was also observed<br />

which the same in the bacteriocin R from L. fermentum.<br />

In the present study FT-IR Spectrum of the protein<br />

revealed the presence of peaks at the wave numbers of<br />

3288, 3090, 2958, 2924 and 2849 cm -1 which indicated<br />

the presence of NH, NH3, CH3, CH2 and CH2 groups<br />

respectively. The wave numbers 1660, 1632, 1589 and<br />

1540 cm -1 indicated the presence of their bending mode<br />

of amide, methyl, and amide groups respectively.<br />

Comparing the results with the nisin standard, the protein<br />

in the sample was confirmed as a bacteriocin. The peak<br />

at 1546 cm -1 indicated a secondary amide was reported<br />

(Silverstein et al., 1991; Yakimov et al., 1995). The FT-IR<br />

spectrum offered concrete evidence that the substance<br />

contained a peptide in its structure. Acidocin 8912<br />

(Tahara et al., 1992) and lactacin B (Barefoot and<br />

Klaenhammer, 1984) were reported to be 5.4 and 6.5<br />

kDa, respectively. In the present study the molecular<br />

weight determination thorough SDS-PAGE showed that<br />

the molecular weight of bacteriocin as 18 kDa protein.<br />

Compared to many other studies the molecular weight<br />

obtained in the present study seemed to be high.


Conclusion<br />

Figure 10. The FT-IR spectrum of Bacteriocin from L. fermentum.<br />

Lane 1 Lane 2<br />

Figure 11. SDS-PAGE of partially<br />

purified protein Lane 1-Marker,<br />

Lane 2-Partial purified Bacteriocin.<br />

The present study showed that the bacteriocin of L. fer<br />

effects on some clinically important food borne<br />

Indira et al. 2295<br />

pathogens. This revealed the potential application of<br />

bacteriocin produced by L. fermentum as a biopreservatives<br />

for the improvement of the microbial safety of<br />

fermented foods and reduction in food contamination<br />

which causes illness to human beings. The study<br />

revealed that Lactobacillus strains of marine origin are<br />

having the potential to use as biopreservatives especially<br />

in seafoods. The production of bacteriocin from L.<br />

fermentum, seems to be ideal for industrial scale<br />

production and commercial utilization.<br />

ACKNOWLEDGMENT<br />

We thank Dr. T. Balasubramanian, Dean, Faculty of<br />

Marine Sciences, Annamalai University, Parangipettai for<br />

providing facilities.<br />

REFERENCES<br />

Aymerich MT, Garriga M, Monfort JM, Nes I, Hugas M (2000).<br />

Bacteriocin-producing lactobacilli in Spanishstyle fermented<br />

sausages: Characterization of bacteriocins. Food Microbiol., 17: 33–<br />

45.<br />

Barefoot SF, Klaenhammer TR (1984). Purification and characterization<br />

of the Lactobacillus acidophilus bacteriocin lactacin B. Antimicrob.<br />

Agents Chemother., 26: 328–334.<br />

Buchanan RE, Gibbons NE, Cowan ST, Holt TG, Liston J, Murry RGE,<br />

Niven CF, Ravin AW, Stainer RY (1974). Bergey’s Manual of<br />

Determinative Bacteriology. Eds: Williams and Wilkins Co: Baltimore.<br />

Castla D, Requena T, Gomez R (1996). Antimicrobial activity of lactic<br />

acid bacteria isolated from goats milk and artisanal cheeses:<br />

characteristics of a bacteriocin produced by Lactobacillus curvatus.


2296 Afr. J. Microbiol. Res.<br />

IFPL 105. J. Appl. Bacteriol., 81: 35–41.<br />

Cone DK (1982). A Lactobacillus spp. from diseased female rainbow<br />

trout, Salmo gairdneri Richardson, in Newfoundland, Canada. J. Fish<br />

Dis., 5: 479-485.<br />

Curragh HJ, Collins MA (1992). High levels of spontaneous drug<br />

resistance in Lactobacillus. J. Appl. Bacteriol., 73: 31-36.<br />

Daeschel MA, Fleming HP (1984). Selection of lactic acid bacteria for<br />

use in vegetable fermentations. Food Microbiol., 1: 303–313.<br />

Food and Drug Administration (1988). Nisin preparation: Affirmation of<br />

GRAS status as direct human food ingredient. Federal Register., 53:<br />

11247.<br />

Harlander SK (1993). Regulatory aspects of bacteriocin used. In:<br />

Bacteriocin of LAB, ed by DG Hoover and LR Steinson San Diego,<br />

California. <strong>Academic</strong> Press Inc.<br />

Holzapfel WH, Geisen R, Schillinger U (1995). Biological preservation of<br />

foods with reference to protective cultures, bacteriocins and food<br />

grade enzymes. Inter. J. Food Microbiol., 24: 343-362.<br />

Johnson AP, Uttley AHC, Woodford N (1990). Resistance to<br />

Vancomycin and Teicoplanin: an emerging clinical problem. Cl<br />

Microbiol Rev., 3: 280-291.<br />

Kandler O, Weiss N (1986). In: Bergey's Manual of Systematic<br />

Bacteriology Vol. 2, Baltimore: Williams and Wilkins., pp.1209–1234.<br />

Klaenhammer TR (1993). Genetics of bacteriocins produced by lactic<br />

acid bacteria. FEMS Microbiol Rev., 12: 39-86.<br />

Lee YK (2004). Microbial Biotechnology principles and applications.,<br />

pp.124.<br />

Lowry OH, Rosenberg HJ, Farr AL, Randall RF (1951). Protein<br />

measurement with Foiln phenol reagent. J. Bio. Chem., 193: 265-<br />

275.<br />

Merritt WM, Dean JA, Settle FA (1986). Instrumental method of<br />

analysis. CBS, New Delhi.<br />

Messens W, Neysens P, Vansieleghem W, Vanderhoeven J, De Vuyst<br />

L (2002). Modeling Growth and Bacteriocin Production by<br />

Lactobacillus amylovorus DCE 471 in Response to Temperature and<br />

pH Values Used for Sourdough Fermentations. Appl. Env. Microbiol.,<br />

68: 1431-1435.<br />

Navarro L, Zarazaga M, Saenz J, Ruiz-Larrea F, Torres C (2000).<br />

Bacteriocin production by lactic acid bacteria isolated from Rioja red<br />

wines. J. Appl. Microbiol., 88: 44–51.<br />

Ogden K, Waites MJ, Hammond JRM (1985). Nisin and brewing. J. Inst.<br />

Brew., 94: 233–238.<br />

Ogunbanwo ST, Sanni AI, Onilude AA (2003). Characterization of<br />

bacteriocin produced by Lactobacillus plantarum F1 and<br />

Lactobacillus brevis OG1. Afr. J. Biotechnol., 2: 219-227.<br />

Okafor N, Nzeako BC (1985). Microbial flora of fresh and smoked fish<br />

from Nigerian fresh waters. Food Microbiol., 2: 71-75.<br />

Reinheimer JA, Demkow MR, Candioti MC (1990). Inhibition of coliform<br />

bacgeria by lactic cultures. Austr. J. Dairy Technol., 5-9.<br />

Roberts RM, Gilbert JC, Waldb BR, Wingrove AS (1985). Evidence for<br />

cyclic bromonium ion transfer in electrophilic bromination of alkenes:<br />

Reaction of -alkenyl glycosides with aqueous N-bromosuccinimide.<br />

Mod. Exp. Organic Chem., 52: 7663-7678.<br />

Ross RP, Galvin M, McAuliffe O, Morgan SM, Ryan MP, Twomey DP,<br />

Meaney WJ, Hill C (1999). Developing applications for lactococcal<br />

bacteriocins. Antonie van Leeuwenhoek., 76: 337–346.<br />

Silverstein RM, Bassler GC, Morrill TC (1991). Spectrometric<br />

identification of organic compounds, 5 th ed., John Wiley and Sons,<br />

New York., 512.<br />

Stiles ME, Holzaphel WH (1997). Lactic acid bacteria of foods and their<br />

current taxonomy. Int. J. Food Microbiol., 36: 1 –29.<br />

Tagg JR, Dajani AS, Wannamaker LW (1976). Bacteriocins of Gram<br />

positive bacteria. Bacteriol. Rev., 40: 722-756.<br />

Tahara T, Kanatani K, Yoshida K, Miura H, Sakamoto M, Oshimura M<br />

(1992). Purification and some properties of acidocin 8912, a novel<br />

bacteriocin produced by Lactobacillus acidophilus TK8912. Biosci.<br />

Biotechnol. Biochem., 56: 1212–1215.<br />

Todorov S, Onno B, Sorokine O, Chobert JM, Ivanova I, Dousset X<br />

(1999). Detection and characterization of a novel antibacterial<br />

substance produced by Lactobacillus plantarum ST 31 isolated from<br />

sourdough. Int. J. Food Microbiol., 48: 167-177.<br />

Yakimov MM, Timmis KN, Wray V, Fredrickson HL (1995).<br />

Characterization of a new lipopeptide surfactant produced by<br />

thermotolerant and halotolerant subsurface Bacillus licheniformis<br />

BAS50. Appl. Environ. Microbiol., 61: 1706–1713.


African Journal of Microbiology Research Vol. 5(16), pp. 2297-2303, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.616<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Ephedra alata as biologically-based strategy inhibit<br />

aflatoxigenic seedborne mold<br />

Al-Qarawi, A. A. 1 , Abd_Allah, E. F. 1 * and Hashem Abeer 2<br />

1 Department of Plant Production, College of Food and Agricultural Sciences, King Saud University, Saudi Arabia.<br />

2 Department of Botany and Microbiology, Faculty of Science, King Saud University, Saudi Arabia.<br />

Accepted 30 July, 2011<br />

Investigation has been carried out to evaluate the effect of range plant Ephedra alata as biologicallybased<br />

strategy inhibit growth characters and aflatoxin production (in vitro and in vivo) of Aspergillus<br />

flavus. The aqueous extract of E. alata had significant inhibitory potential against growth as well as<br />

aflatoxin production by aflatoxigenic seedborne mold (Aspergillus flavus). Moreover, it has been found<br />

that, the addition of 1 and 2% (w/w) of plant powder material of E. alata to corn grains and soybean<br />

seeds respectively decreased the aflatoxin contamination and improve their nutritional value (total<br />

nitrogen content, fiber content, total lipids content and ash content) under storage conditions. The<br />

results observed here suggest employment of E. alata as an alternative non-chemical mean to control<br />

aflatoxin contamination of fodder in Saudi Arabia.<br />

Key words: Biocontrol, Aflatoxins, Aspergillus flavus, Ephedra alata, nutritional value, Saudi Arabia.<br />

INTRODUCTION<br />

Aflatoxins are secondary metabolites produced by many<br />

strains of Aspergillus flavus, known to be potent<br />

carcinogens as well as hepatotoxic agents and pose<br />

serious hazards to human, animal health and milk<br />

production in many countries including Saudi Arabia.<br />

Aspergillus flavus are widely distributed in agricultural<br />

commodities like maize (Giorni et al., 2007) and soybean<br />

(Gupta and Venkitasubramanian, 1975). The death of<br />

thousands of camels in Saudi Arabia may have been<br />

caused by the consumption of these aflatoxinscontaminated<br />

fodders (Bokhari, 2010). Bioremediation of<br />

mycotoxins has been carried out by bacteria (Abd_allah<br />

and Ezzat, 2004), fungi (Abd_allah and Ezzat, 2005) and<br />

friendly chemicals such as chitosan (Abd_Allah and<br />

Hashem, 2006). Recently, impacts of natural products of<br />

plant origin have been introduced as non-chemical mean<br />

for biocontrol and bioremediation of mycotoxins in<br />

agricultural commodities (Parsaeimehr et al., 2010).<br />

Ephedra, a medicinal plant belonging to the Ephedraceae<br />

is a genus of non-flowering seed plants belonging to the<br />

Gnetales, the closest living relative of the angiosperm<br />

(Friedman, 1996). The antimicrobial potential of some<br />

*Corresponding author. E-mail: eabdallah@ksu.edu.sa.<br />

Epedhra species such as E. altissima Defs (Tricker et al.,<br />

1987), E. transitorai (Al-Khalil, 1998), E. nebrodensis<br />

(Cottiglia et al., 2005), E. major (Bagheri et al., 2009) and<br />

E. breana (Feresin et al., 2001) has been recognized.<br />

Ephedra alata is a range plant with medicinal application,<br />

belonging to ephedra native of Asia including Saudi<br />

Arabia (Abourashed et al., 2003). Additionally, the foliage<br />

of E. alata has acceptable aroma and used as food stuff<br />

during animal grazing in Saudi Arabia (Al-Taisan et al.,<br />

2010).<br />

The objective of the current study was to describe the<br />

antifungal activity of E. alata (aqueous extract of shoot<br />

system) grown in Saudi Arabia, against growth<br />

characters and aflatoxin production of local seedborne<br />

isolate of Aspergillus flavus in vitro and on both maize<br />

grains and soybean seeds under storage conditions.<br />

Also, the relevance of nutritional value of stored<br />

agricultural commodities (maize grains and soybean<br />

seeds) was studied in relation to storage treatments.<br />

MATERIALS AND METHODS<br />

Experimental mold<br />

Local aflatoxin producing isolate similar to Aspergillus flavus link<br />

was isolated from soybean seed samples collected from Derab


2298 Afr. J. Microbiol. Res.<br />

Agriculture Research Station, Riyadh, Saudi Arabia. The mold was<br />

identified using current universal keys of identification (Domsch et<br />

al., 1993) by Dr. Abeer Hashem, Botany and Microbiology<br />

Department, Faculty of Science, King Saud University, Riyadh,<br />

Saudi Arabia.<br />

Plant extract and antifungal evaluation<br />

Fresh aerial parts (shoot system) of E. alata Decne were collected<br />

from wildlife Research and Development at Thumama, Riyadh,<br />

Saudi Arabia. Plant materials were dried in dark at room<br />

temperature (25±1°C) and powdered. Known weight of powdered<br />

material was extracted with aqueous ethanol (10:90, v/v) at 30°C<br />

for overnight and filtered through double layers of Whatman no. 1<br />

filter paper. The extract was completed to known volume using<br />

aqueous ethanol as above. The extract was filtered using 0.22<br />

micron cellulose acetate filter units (Whatman, Dassel, Germany)<br />

for sterilization. The antifungal activity of plant extract was<br />

determined by disc method (Roberts and Selitrenikoff, 1988), agar<br />

plate method (Bobbarala et al., 2009) and broth culture flask using<br />

glucose-ammonium nitrate salt medium (Brain et al., 1961). The<br />

production and germination of conidia were estimated according to<br />

the methods described by Wilson et al. (2004) and Ghaly et al.<br />

(1998) respectively.<br />

Mycelial dry weight<br />

At the end of incubation period, the culture broths were filtered<br />

through pre-weighed filter papers (Whatman no. 1). The filter<br />

papers with mycelial growth washed carefully with distilled water<br />

followed by drying at 80°C up to two successive constant weights<br />

were obtained. The net dry weight of mycelia was then determined.<br />

Aflatoxin analysis<br />

Replicated samples (each 50 ml of culture growth medium or 100 g<br />

of both maize grains and soybean seeds) were extracted using<br />

chloroform and cleaned up as described by Dutton and Westlake<br />

(1985). Column chromatography (AOAC, 1970) was carried out on<br />

silica gel G-60 mesh 0.02- 0.2 (BDH Chemicals, Poole, UK).<br />

Chemical and biological confirmatory tests to aflatoxin B1 were<br />

carried out according to AOAC (1970) and Madhyastha et al.<br />

(1994), respectively. The sensitive strain of Bacillus subtilis was<br />

used throughout for biological confirmatory test (Abd_Allah and<br />

Ezzat (2004). Thin layer chromatography (TLC) plates (20 x 20 cm)<br />

coated with 0.5-mm-thick silica gel DG (Kieselgel-DG, Riedel- De<br />

Haen, Seelze-Hannover, Germany) were used for chromatographic<br />

separation of aflatoxin B1 followed with elusion of aflatoxin B1 spots<br />

using benzene: acetonitrile (98:2 [v/v]). Quantitative determination<br />

of aflatoxin B1 was done spectrophotometrically according to<br />

Nabney and Nesbitt (1965). Standard aflatoxin B1 (Sigma) was<br />

used as reference in all experiments.<br />

Storage experiments<br />

Storage experiment was carried out in polyethylene pouches (200 x<br />

150 mm) each containing 100 g of blend consisted of powdered E.<br />

alata, mixed well with powdered maize grains or soybean seeds to<br />

give final concentration of 0.5, 1.0 and 2.0% (w/w). The pouches<br />

were inoculated with A. flavus (10 6 spores/pouch). Control pouches<br />

were used as reference. The moisture content was expressed as<br />

percentage of net weight (Abd_Allah and Hashem, 2006). The<br />

storage experiment continued for six months at room temperature<br />

(25 ± 1°C) in dark.<br />

Estimation of nutritional values<br />

The nutritional values (total lipid content, ash content, fiber content,<br />

total nitrogen content) of all plant materials were estimated<br />

according to AOAC (1995).<br />

Statistical analysis<br />

All experiments were repeated at least three times and treatment<br />

means were compared using Least Significant Difference (LSD)<br />

analysis according to Daniel (1987).<br />

RESULTS<br />

Thin layer chromatographic examinations indicated that,<br />

Rf -values and color of spots developed from both<br />

chloroform extract of A. flavus and standard aflatoxin B1<br />

were the same before and after chemical confirmatory<br />

tests. The first conformity test (addition of HCl) indicated<br />

the presence of one blue spot (under UV light 366 mm)<br />

with Rf -value 0.66. The second conformity (addition of<br />

conc. HCl + Acetanhydride) test indicated the presence of<br />

two spots with Rf -values were 0.10 and 0.42<br />

respectively. Bioautographic examination (for both<br />

chloroform extract and standard aflatoxin B1) against<br />

Bacillus subtilis (Biological indicator) indicated the<br />

presence of one inhibitor zone with Rf value was 0.78.<br />

Preliminary experiment was carried out (in vitro) to<br />

demonstrate the antifungal potential of the plant (E. alata)<br />

extract against A. flavus using disc diffusion plate<br />

bioassay (Figure 1). The results in Table 1 revealed that<br />

the gradual concentrations of plant extract caused<br />

significant inhibition in both radial growth rate by percent<br />

of 41.55, 67.98, 88.83 and conidial production (Figure 2)<br />

by percent of 13.82, 50.00 and 90.42 of A. flavus,<br />

respectively .<br />

Data in Table 2 shows the inhibitory effect of plant<br />

extract on the number of germinated conidia especially<br />

after 6 h (absolutely inhibition was observed). It was<br />

noted that, the prolongation of incubation period<br />

decrease the inhibitory effect potential of plant extract at<br />

all concentrations used (Table 2).<br />

The results in Table 3 indicated clearly that all concentrations<br />

of plant extract induced significant inhibition in<br />

both mycelial growth (Figure 3) and aflatoxin B1<br />

production by percent of 33.49, 58.90, 79.76 and 65.75,<br />

83.90, 100 respectively. It is worthy to mention that such<br />

inhibitory effect of plant extract was more intense towards<br />

aflatoxin B1 production compared with mycelial growth at<br />

the same concentrations.<br />

A storage experiment was carried out to investigate the<br />

antagonistic potential of E. alata against aflatoxin<br />

production by A. flavus on maize grains and soybean<br />

seeds (in vivo) with special reference to their nutritional<br />

value. The obtained data indicated that employment of E.<br />

alata at concentrations 0.5 and 1.0% caused significant<br />

inhibition in the accumulation of aflatoxin B1 on maize<br />

grains and soybean seeds (Table 4) by percent of 63.81,


Figure 1. The antifungal activity of plant (E. alata) extract (arraw)<br />

against radial growth of A. flavus (A) using disc assay method.<br />

Table 1. Effect of different concentrations of E. alata (w/v) on radial growth rate (cm day -1 ) and conidial<br />

production ([conidia mm -2 ] X 10 4 ) of A. flavus grown at 28±°C for 96 h in dark.<br />

Treatment<br />

(E. alata w/v)<br />

Radial growth rate<br />

(cm day -1 )<br />

Conidial production<br />

(conidia mm -2 X 10 4 )<br />

Control 1.362 9.382<br />

E. alata 0.5% 0.796 8.102<br />

E. alata 1.0% 0.436 4.708<br />

E. alata 2.0% 0.152 0.584<br />

LSD at: 05 0.066 0.475<br />

Figure 2. Effect of different concentrations (A: control; B: 0.5%; C: 1.0%; D:<br />

2.0%) of plant (E. alata) extract on conidial production of A. flavus.<br />

Al-Qarawi et al. 2299


2300 Afr. J. Microbiol. Res.<br />

Table 2. Effect of different concentrations of E. alata (w/v) on number of germinated conidia (out of 50 conidia) after different<br />

incubation periods (hours).<br />

Treatment<br />

(E. alata w/v)<br />

Number of germinated conidia (out of 50 conidia) after different incubation periods (h)<br />

6 h 12 h 24 h<br />

Control 20.67 49.33 50.00<br />

E. alata 0.5% 11.00 42.83 49.17<br />

E. alata 1.0% 6.17 36.67 48.67<br />

E. alata 2.0% 0.00 32.17 47.17<br />

LSD at: 05 4.7884 3.4099 2.4237<br />

Table 3. Effect of different concentrations of E. alata (w/v) on mycelial dry weight (g 50 ml -1 culture medium) and<br />

aflatoxin B1 production (µg 50 ml -1 culture medium).<br />

Treatment<br />

(E. alata w/v)<br />

Mycelial dry weight<br />

(g 50 ml -1 culture medium)<br />

Aflatoxin B1 production<br />

µg 50 ml -1 culture medium µg g -1 dry weight<br />

Control 0.50154 351.6 701.04<br />

E. alata 0.5% 0.33354 120.4 360.97<br />

E. alata 1.0% 0.21616 56.6 261.84<br />

E. alata 2.0% 0.1015 0.0 0.0<br />

LSD at: 05 0.0331 22.192<br />

Figure 3. Effect of different concentrations [w/v] (A, control, B, 0.5%, C, 0.1%, D, 0.2%) of plant (E. alata) extract on<br />

mycelial growth of A. flavus.<br />

84.89 and 59.57, 89.72, respectively. Absolute inhibition<br />

of aflatoxin B1 accumulated in both maize and soybean<br />

grains was observed accompany with employment of<br />

2.0% (w/w) E. alata (Table 4).<br />

The results show that A. flavus caused significant<br />

deterioration in the nutritional value of maize grains<br />

(Table 5) and soybean seeds (Table 6). Such deterioration<br />

in the nutritional value of agricultural commodities<br />

was aware as significant decrease in their total lipids,<br />

crude fibre and crude protein accompanied with a<br />

significant increase in ash content (Table 5 and 6). The<br />

employment of powdered E. alata succeeded to


Al-Qarawi et al. 2301<br />

Table 4. Effect of different concentrations of E. alata (w/w) on aflatoxin B1 production (µg kg -1 ) on both maize<br />

grains and soyabean seeds stored under biological stress of A. flavus.<br />

Treatment<br />

Aflatoxin B1 production (µg /kg<br />

(E. alata w/w)<br />

-1 )<br />

Maize grains Soyabean seeds<br />

control 212.22 131.20<br />

E. alata 0.5% 76.79 53.04<br />

E. alata 1.0% 32.06 13.48<br />

E. alata 2.0% 0.0 0.0<br />

LSD at: 05 23.096 17.137<br />

Table 5. Effect of different concentrations of E. alata (w/w) on nutritional value of maize grains stored under biological stress of<br />

A. flavus.<br />

Treatment<br />

( E. alata w/w)<br />

Total lipids*<br />

Nutritional value of maize grains Z<br />

Crude fiber*<br />

Ash content* Crude protein**<br />

Control (maize before storage) 4.7752 6.3021 4.2205 2.8240<br />

Maize + A. flavus 2.2577 1.0217 6.3914 1.8387<br />

E. alata 0.5% 3.6204 2.4314 5.3093 2.1400<br />

E. alata 1.0% 4.1111 4.7984 5.4991 2.7304<br />

E. alata 2.0% 4.1139 5.3666 5.6747 3.0224<br />

LSD at: 05 0.3725 0.5438 0.1567 0.3331<br />

*= Total lipids, crude fiber and ash content expressed as percentage (%).<br />

**= Crude protein = total nitrogen (mg/g dry wieght) X 6.25.<br />

Z= Total lipids, crude fiber, ash content and crude protein of E. alata (shoot system) were 1.0425, 4.2463, 7.7599 and 3.1341,<br />

respectively.<br />

Table 6. Effect of different concentrations of E. alata (w/w) on nutritional value of soyabean seeds stored under biological stress<br />

of A. flavus.<br />

Treatment<br />

(E. alata w/w)<br />

Total lipids*<br />

Nutritional value of maize grains Z<br />

Crude fiber*<br />

Ash content* Crude protein**<br />

Control (maize before storage) 18.5986 4.3731 6.4366 34.1012<br />

Maize + A. flavus 15.4252 1.9727 7.5799 12.2231<br />

E. alata 0.5% 16.3723 2.5918 6.8635 18.0028<br />

E. alata 1.0% 17.7983 3.5118 6.4536 24.6835<br />

E. alata 2.0% 18.2606 4.0598 6.3337 30.7081<br />

LSD at: 05 0.7713 0.2725 0.6477 11.735<br />

*= Total lipids, crude fiber and ash content expressed as percentage (%).<br />

**= Crude protein = total nitrogen (mg/g dry wieght) X 6.25.<br />

Z = Total lipids, crude fiber, ash content and crude protein of E. alata (shoot system) were 1.0425, 4.2463, 7.7599 and 3.1341,<br />

respectively.<br />

protect the maize grains (Table 5) and soybean seeds<br />

(Table 6) against the destroyer effect of A. flavus. The<br />

constrictive role of E. alata was directly proportional with<br />

its employed concentrations.<br />

DISCUSSION<br />

Aflatoxins are highly toxic secondary metabolites<br />

produced by certain strains of Aspergillus flavus caused<br />

many diseases for animals as well as humans. In our<br />

investigation, the chemical and biological confirmatory<br />

tests demonstrated the production of aflatoxins B1 by our<br />

experimental mold (A. flavus). Aflatoxins contamination of<br />

foodstuffs and agricultural commodities is a worldwide<br />

problem in tropical and subtropical regions where climatic<br />

conditions and storage practices are conducive to fungal<br />

growth and mycotoxin production (Thompson and Henke,<br />

2000). Many strategies including employment of biological<br />

products from plant origin are being investigated to


2302 Afr. J. Microbiol. Res.<br />

manage deterioration of agricultural commodities<br />

including animal fodder. In the same connection,<br />

investigations of antifungal potential of Ephedra were<br />

carried out recently (Bagheri et al., 2009; Parsaeimehr et<br />

al., 2010). In our results, the aqueous extract of E. alata<br />

was found to be effective in controlling the growth rate<br />

and conidial production by A. flavus. The antifungal<br />

potential of E. alata (aqueous extract) was extended to<br />

inhibit number of germinated conidia, mycelial growth<br />

(mycelial dry weight) and aflatoxin B1 production by A.<br />

flavus in variable percentages. In the same concept, the<br />

results of storage experiment indicated that application of<br />

plant (E. alata) powder was accompanied with significant<br />

decrease in contamination of both maize grains and<br />

soybean seeds with aflatoxin. The antimicrobial potential<br />

of E. alata noted here in the present study has been attributed<br />

to presence of Cis-314-methanoproline (Caveney<br />

et al., 2001), Citronellol (Rosato et al., 2007) and<br />

Heptadecane (Bagheri et al., 2009) which are recorded<br />

as photochemicals possess antimicrobial activity found in<br />

Ephedra.<br />

In geneal, oil seeds introduced into animal feeds as<br />

source of fiber, lipids and fat soluble vitamins, however<br />

the fungal contamination lead to significant deterioration<br />

in their nutritional value due to substrate utilization by<br />

molds. Previous studies indicated the decrease in fat,<br />

protein and fiber contents of many seeds and grains<br />

involved sorghum and soybean infected with different<br />

seedborne fungi and this was accompanied with an<br />

increase in ash content (Fapohunda and Olajuyiabe,<br />

2006; Ezekiel et al., 2010). In this regard, fiber content<br />

with available protein and fat formed ready substrate for<br />

fungal activities including production of hydrolytic<br />

enzymes (Mellon et al., 2007). The results of our study<br />

recorded similar significant decrease in protein, fiber, and<br />

lipids accompanied with an increase in ash content of<br />

both maize grains and soybean seeds due to the<br />

inoculation with A. flavus. The application of plant (E.<br />

alata) powder succeeded to decrease the deterioration of<br />

the nutritional value (fat, protein, fiber, ash contents) of<br />

both maize grains and soybean seeds due to infection<br />

with A. flavus. Regarding the justification manner, our<br />

results showed E. alata possess highly antagonistic<br />

potential against growth and aflatoxin production of A.<br />

flavus in agree with Bagheri et al. (2009) and<br />

Parsacimehr et al. (2010).<br />

Furthermore, with respect to E. alata, it has been<br />

shown that range plant contain suitable contents of<br />

protein and fiber (Nawwar et al., 1985) able to<br />

compensate the decrease in nutritional value of maize<br />

grain and soybean seeds due to mold contamination. Our<br />

results provide evidence for the first time to application of<br />

E. alata as naturally additive control aflatoxin contamination<br />

of fodder in addition to improve its nutritional<br />

value. This may be a novel non-chemical strategy to<br />

protect thousands of camels in Saudi Arabia against the<br />

mystery of fodder contamination with aflatoxins, the main<br />

reason for camels' death in our country.<br />

ACKNOWLEDGMENT<br />

The authors wish to acknowledge Research Centers in<br />

both college of Science and college of food sciences and<br />

agriculture, King Saud University, Riyadh, Saudi Arabia.<br />

At the same time, we acknowledge Deanship of Scientific<br />

Research, King Saud University, Riyadh, Saudi Arabia.<br />

REFERENCES<br />

Abd_Allah EF, Ezzat SM (2004). Role of lipid metabolism through<br />

bioremediation of fusaric acid in germinating peanut seedlings.<br />

Phytoparasitica, 32: 38-42.<br />

Abd_Allah EF, Ezzat SM (2005). Natural occurrence of citrinin in rice<br />

grains and its biocontrol by Trichoderma hamatum. Phytoparasitica,<br />

33: 73-84.<br />

Abd_Allah EF,Hashem Abeer (2006). Seed mycoflora of Lens<br />

esculenta and their biocontrol by chitosan. Phytoparasitica, 34: 213-<br />

218.<br />

AOAC [Association of Official Analytical Chemists] (1970). Official<br />

Methods of Analysis (11 th ed.), Section 26.019 (a), 429. AOAC,<br />

Washington, D.C.<br />

AOAC [Association of Official Analytical Chemists] (1995). Official<br />

Methods of Analysis of AOAC International. 16 th edition, vol. 2,<br />

Arlington, Virginia.<br />

Abourashed EA, EL-Alfy Abir T, Khan IA, Walker L (2003). Ephedra in<br />

perspective- a current review. Phytother. Res., 17: 703-712.<br />

AL-Khalil S (1998). Transtorine, a new quinoline alkaloid from Ephedra<br />

transitoria. J. Natl. Prod., 61: 262–263.<br />

Al-Taisan Wafaa A, AL-Qarawi AA, Alsubiee Moodi S (2010). Effect of<br />

water stress by polyethylene glycol 8000 and sodium chloride on<br />

germination of Ephedra alata Decne seeds. Saudi J. Biol. Sci., 17:<br />

253-257.<br />

Bagheri G, Bigdeli M, Shams GM, Razzaghi AM (2009). Inhibitory<br />

Effects of Ephedra major Host on Aspergillus parasiticus Growth and<br />

Aflatoxin Production. Mycopathologia, 168: 249-255.<br />

Bobbarala V, Chadaram RK, Vadlapudi V, Katikala PK (2009).<br />

Medicinal plants as alternative biocontrol agents in the control of<br />

seed borne pathogen Macrophomina phaseolina. J. Pharm. Res., 2:<br />

1045-1048.<br />

Bokhari FM (2010). Implications of fungal infections and mycotoxins in<br />

camel diseases in Saudi Arabia. Saudi J. Biol. Sci., 17: 73-81.<br />

Brain PW, Dowkins AW, Grove JF, Hemming DL, Norris GLF (1961).<br />

Phytotoxic compounds produced by Fusarium equiseti. J. Bot., 12: 1-<br />

12.<br />

Caveney S, Charlet DA, Freitag H, Maier-Stolte M and Starratt A<br />

(2001). New observations on the secondary chemistry of world<br />

Ephedra (Ephedraceae). Am. J. Bot., 88: 1199-1208.<br />

Cottiglia F, Bonsignore L, Casu L, Deidda D (2005). Phenolic<br />

constituents from Ephedra nebrodensis. Natl. Prod. Res., 19: 117-23.<br />

Daniel WW (1987). Biostatistics: A foundation for Analysis in the Health<br />

Science. 4 th ed., John Wiley and Sons, New York, NY., pp. 292-293.<br />

Domsch KH, Gams W, Anderson TH (1993). Compendium of Soil<br />

Fungi. <strong>Academic</strong> Press, London, pp. 860.<br />

Dutton MF, Westlake K (1985). Occurrence of mycotoxins in cereals<br />

and animal feedstuffs in Natal, South Africa. J. Assoc. Off. Anal.<br />

Chem., 86: 839-842.<br />

Ezekiel CN, Kolawole DO, Olufuwa OA, Odu IA (2010). Shelf-life and<br />

pattern of deterioration in firm Tofu. Acta Satrch J. Life Phys. Sci., 3:<br />

40-47.<br />

Feresin GE, Tapia A, Lopez SN, Zacchino SA (2001). Antimicrobial<br />

activity of plants used in traditional medicine of Sun Juan Province,<br />

Argentine. J. Ethnopharmacol., 78: 103-107.<br />

Fapohunda S, Olajuyigbe OO (2006). Studies on stored cereals degradation<br />

by Alternaria tenuissima. Acta Botanica Mexicna, 77: 31-40.<br />

Friedman WE (1996). Introduction to biology and evolution of the<br />

Gnetales. Int. J. Plant Sci., 157: S1-S2.<br />

Ghaly MF, Ezzat SM, Sarhan MM (1998). Use of propolis and ultragriseofulvin<br />

to Inhibit aflatoxigenic fungi. Folia Microbiol., 43:156-160.


Giorni P, Magan, N, Pietri A, Bertuzzi T, Battilani P (2007). Studies on<br />

Aspergillus section Flavi isolated from maize in northern Italy. Int. J.<br />

Food Microbiol., 113: 330–338<br />

Gupta SK, Venkitasubrmanian TA (1975). Production of aflatoxin on<br />

soybeans. Appl. Microbiol., 29: 834-836.<br />

Madhyastha MS, Marquardi RR, Masi A, Borsa J, Froshlich AA (1994).<br />

comparison of toxicity of different mycotoxins to several species of<br />

bacteria and yeast: Use of Bacillus brevis in a disc diffusion assay. J.<br />

Food Protect, 57: 48-53.<br />

Mellon JE, Cotty PJ, Dowd MK (2007). Aspergillus flavus hydrolases:<br />

their roles in pathogenesis and substrate utilization. Appl. Microbiol.<br />

Biotechnol., 77: 497–504.<br />

Nabney J, Nesbitt BF (1965). A spectophotometric method for<br />

determining the aflatoxins. Analyst, 90: 155-160.<br />

Nawwar MAM, Barakat HH, Buddrus J, Linscheid M (1985). Alkaloidal,<br />

lignan and phenolic constituents of Ephedra alata. Phytochemistry,<br />

24: 878-879.<br />

Parsaeimehr A, Sargsyan E, Javidnia K (2010.) A comparative study of<br />

the antibacterial, antifungal and antioxidant activity and total content<br />

of phenolic compounds of cell cultures and wild plants of three<br />

endemic species of Ephedra. Molecules, 15: 1668-1678.<br />

Al-Qarawi et al. 2303<br />

Roberts WK, Selitrenikoff CP (1988). Plant cell and bacterial chitinase<br />

differ in antifungal activity. J. Gen. Microbiol., 134: 169-176.<br />

Rosato A, Vitali C, De Laurentis N (2007). Antibacterial effect of some<br />

essential oils administered alone or in combination with Norfloxacin.<br />

Phytomedicine, 14: 727–732.<br />

Thompson C, Henke SE (2000). Effect of climate and type of storage<br />

container on aflatoxin production in corn and its associated risks to<br />

wildlife species. J. Wildl. Dis., 36: 172-179.<br />

Tricker AR, Wacker CD, Preussmann R (1987). Nitrosation products<br />

from the plant Ephedra altissima and their potential endogenous<br />

formation. Cancer Lett., 35: 199-206.<br />

Wilson RA, Calvo Ana M, Chang Perng-Kuang and Keller Nancy P<br />

(2004). Characterization of the Aspergillus parasiticus 12 -desaturase<br />

gene: a role for lipid metabolism in the Aspergillus–seed interaction.<br />

Microbiol., 150: 2881–2888.


African Journal of Microbiology Research Vol. 5(16), pp. 2304-2310, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.619<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Primary resistance rates of Mycobacterium<br />

tuberculosis complex strains isolated from new<br />

tuberculosis cases: A 6-year observation<br />

Servet Kayhan*, Alper Akgüne , Hikmet Tereci and Ümit Tutar<br />

Samsun Chest Diseases and Thoracic Surgery Hospital, Turkey.<br />

Accepted 30 July, 2011<br />

The aim of the study was to evaluate the automated mycobacteria growth ındicator tube (MGIT) for drug<br />

susceptibility testing of Mycobacterium tuberculosis and to determine resistance patterns. We used<br />

BACTEC MGIT 960 System to determine the susceptibility of M. tuberculosis complex isolates to major<br />

anti tuberculous agents. Patients with single and first positive isolates were enrolled in the study. We<br />

have performed our drug susceptibility study between January 2005 and December 2010 for monitoring<br />

of drug resistance patterns in six years. A total of 1240 (77.16%) of the 1607 isolates were susceptible to<br />

all four of the antimycobacterial agents while 369 (22.96%) were found to be resistant to one or more of<br />

the drugs. The rate of isolates resistant to streptomycin (SM) was 6.84%, the other rates were 17.17% to<br />

isoniazid (INH), 5.28% to rifampicin (RIF) and 4.10% to ethambutol (ETM). Single drug resistance rates<br />

were found to be 12.13% for INH, 0.99% for RIF, 5.6% for SM, and 1.74% for ETM. The ratio of resistant<br />

isolates to all four drugs was 0.74% (n=12) and the prevalence of multidrug resistant isolates was 3.92%<br />

(n=63). It was concluded that resistance to INH and RIF continues almost in a straight line in the present<br />

study. Monitoring of drug resistance patterns is essential for accurate drug regimen in management of<br />

tuberculosis.<br />

Key words: Mycobacterium tuberculosis complex, primary resistance, ısoniazid, rifampicin.<br />

INTRODUCTION<br />

The World Health Organisation estimated the global<br />

burden of tuberculosis disease in 2009 as 9.4 million<br />

incident patients, 14 million prevalent cases and 2.38<br />

million deaths in the 2010 TB report<br />

(WHO/HTM/TB/2010-3; WHO/HTM/STB/2010-2). An<br />

estimated 11–13% of incident cases were HIV-positive.<br />

Among TB patients notified in 2009, an estimated 250,<br />

000 had multidrugresistant TB (MDR-TB). There were an<br />

estimated 440,000 cases of MDR-TB in 2008. The four<br />

countries that had the largest number of estimated cases<br />

*Corresponding author. E-mail: servet-kayhan@hotmail.com.tr.<br />

Tel: +903624400038. Fax: +903624400042.<br />

Abbrevations: MGIT, Mycobacterium growth indicator tube;<br />

MTC, Mycobacterium tuberculosis complex; TB, tuberculosis;<br />

INH, ısoniazid; RIF, rifampicin; ETM, ethambutol; SM,<br />

streptomycin; MDR-TB, multi drug resistant tuberculosis; XDR-<br />

TB, extensively drug-resistant tuberculosis.<br />

of MDR-TB in absolute terms in 2008 were China, India,<br />

the Russian Federation and South Africa. Primary<br />

resistance is the resistance pattern seen in new patients<br />

who have not previously been exposed to anti-TB drugs.<br />

Secondary resistance is the resistance pattern in patients<br />

with a previous history of anti-TB treatment and is due to<br />

ineffective chemotherapy. Fifty eight countries and<br />

territories have reported at least one case of extensively<br />

drug-resistant TB (XDR-TB). XDR-TB is defined as<br />

resistance to isoniazid and rifampicin (that is, MDR-TB)<br />

plus resistance to a fluoroquinolone and, at least, one<br />

second-line injectable agent (amikacin, kanamycin and/or<br />

capreomycin) (Bozkurt et al., 2010; EuroTB, 2007). Surveillance<br />

of primary and secondary resistance patterns is<br />

important in assessing the quality of chemo-therapy<br />

programs over several years and detecting errors in past<br />

treatments respectively. The resistant strains are<br />

characterised by a unique, lipid- containing and rigid core<br />

of the cell wall. The mycobacterial cell wall is less<br />

permeable to hydrophilic molecules than other


Table 1. Concentrations of drugs used in this study.<br />

Drug<br />

Concentration of drug after<br />

reconstitution (µg/mL)<br />

Volume added to MGIT<br />

Tubes for test (µL)<br />

Kayhan et al. 2305<br />

Final concentration in<br />

MGIT tubes (µg/mL)<br />

MGIT Streptomycin 83 100 1.0<br />

MGIT Isoniazid 8.3 100 0.1<br />

MGIT Rifampicin 83 100 1.0<br />

MGIT Ethambutol 415 100 5.0<br />

bacteria. That is why mycobacteria are resistant against<br />

the majority of drugs commonly used against bacteria<br />

(De Rossi et al., 2006). Anti tuberculous agents interfere<br />

with enzymes involved in cell wall biosynthesis (isoniazid,<br />

ethambutol, ethionamide), protein synthesis<br />

(streptomycin, other aminoglycosides, macrolides),<br />

transcription (rifampin), or DNA replication (quinolones).<br />

In M. tuberculosis spontaneous mutations occur at a<br />

frequency of approximately 10 -5 to 10 -8 (Inderlied and<br />

Nash, 1996). Since resistances to various drugs arise<br />

independently, the likelihood of spontaneous mutation to<br />

isoniazid and rifampin, for instance, is one in 10 -14 . The<br />

threat of dual mutations is one of the rationales for why<br />

an anti-tuberculosis therapy should consist of a<br />

combination of drugs (Brennan and Draper, 1994). To be<br />

successful in the management of tuberculosis, rapidly<br />

detecting the susceptibility of MTC strains to first line<br />

drugs is very important (Aydın et al., 2011).<br />

Many studies performed in Turkey confirmed that the<br />

resistance rates and patterns of MTC may show different<br />

characters in different regions (Talay et al., 2003; Ta ova<br />

et al., 1997). We aimed in this study to observe the<br />

regional and current rates of primary drug susceptibilities<br />

for MTC strains in active pulmonary tuberculosis by using<br />

clinical specimens (bronchial fluid and sputum).<br />

MATERIALS AND METHODS<br />

American Center for Disease Control (CDC) recommends<br />

performing susceptibility tests on specimens, within, 28 to 30 days<br />

after they are submitted to a laboratory, as we have done in this<br />

study (CDCP, 1993).<br />

Strains<br />

1607 MTC strains isolated from sputum and bronchial fluid samples<br />

of active pulmonary tuberculous patients who were diagnosed and<br />

treated in Samsun Pulmonary Disease and Chest Surgery Hospital<br />

from 2005 to 2010. We have reproduced positive samples by<br />

Lowenstein- Jehnsen (LJ) culture examination in a microbiology<br />

laboratory; then the single and first positive isolates from each<br />

patient enrolled in the study were used.<br />

Homogenisation and decontamination<br />

The bronchioloalveolar lavage fluids (BAL) and sputum samples of<br />

suspected tuberculosis patients were subjected to homogenisation<br />

and decontamination process by N-Acetyl-L-Cistein (NALC) and<br />

sodium hydroxide (NaOH) mixture and then to LJ culture .<br />

Culture and identification<br />

The resistance characteristics related to the primary antituberculous<br />

drugs (SM, INH, RIF, ETM) of 1607 units of MTC strains which<br />

were produced in Solid (Lowenstein-Jensen) and liquid (BACTEC<br />

12B and the Mycobacteria Growth IndicatorTube (MGIT)) nutrient<br />

media were detected by the BACTEC 960 (Becton Dickinson, USA)<br />

system. After the reproduction process, MTC and nontuberculous<br />

mycobacteria (MOTT) separation was made by a p-nitro- - -<br />

hydroxy-asetilamino-propiofen (NAP) test.<br />

Susceptibility test against primary antituberculous drugs<br />

We used BACTEC MGIT 960 System to determine the susceptibility<br />

of M. tuberculosis Complex isolates to major anti tuberculous<br />

agents. The susceptibility test was performed according to the<br />

manufacturer's recommendations (Becton, Dickinson and Company<br />

USA). This research is the result of several years study but we used<br />

same method (Trade mark; Becton, Dickinson and Company USA)<br />

and same drug concentrations in six years overall susceptibility<br />

tests. So there was not any change in the brand of drugs. Dilutions<br />

of drugs used in this study are shown in Table 1.<br />

Quality control<br />

For quality control of susceptibility tests, the ATCC 27294 (H37Rv)<br />

strain was used. The Rome Supranational Tuberculosis Reference<br />

Laboratory of World Health Organization and Turkey’s Refik<br />

Saydam Hygiene Center Tuberculosis Research Laboratories,<br />

cooperated in the preparation of a quality control program which<br />

was implemented in this study.<br />

Statistical analysis<br />

We used SSPS.15 and minitab.16 programme for statistic analysis<br />

on INH and MDR ratios by years.<br />

RESULTS AND DISCUSSION<br />

We introduced the state of primary resistance in MTC to<br />

four major antituberculous agents in the last six years in<br />

Samsun province (the central city of Turkey’s Middle<br />

Black sea region, with a population of 1.3 million) and we<br />

also retrospectively compared these values with the other<br />

regional and national determinants on drug resistance. In<br />

this study, 16,932 patient specimens were sent to our


2306 Afr. J. Microbiol. Res.<br />

Table 2. The primary resistance rates of Mycobacterium tuberculosis complex isolates according to years.<br />

Type of resistance 2005 2006 2007 2008 2009 2010 Total (n)<br />

Single drug resistance<br />

SM (%) 1(0.32) 5(1.67) 12(5) 7(2.99) 5(2.02) 13(4.69) 43(2.67)<br />

INH (%) 27(8.64) 25(8.38) 32(13.33) 26(11.11) 21(8.46) 33(11.91) 164(10.20)<br />

RIF (%) 1(0.32) 5(1.67) 6(2.50) - 2(0.80) 2(0.72) 16(0.99)<br />

ETM (%) - 7(2.34) 7(2.91) 11(4.7) 2(0.80) 1(0.36) 28(1.74)<br />

Any (total) monodrug resistance<br />

SM(%) 7(2.25) 14(4.70) 28(11.67) 14(5.98) 23(9.27) 24(8.66) 110(6.84)<br />

INH(%) 49(15.80) 39(13.08) 58(24.17) 39(16.66) 41(16.53) 50(18.05) 276(17.17)<br />

RIF(%) 19(6.12) 11(3.69) 20(8.33) 5(2.13) 15(6.04) 15(5.41) 85(5.28)<br />

ETM(%) 5(1.61) 9(3.02) 20(8.33) 23(9.82) 4(1.61) 5(1.80) 66(4.10)<br />

Resistance to 2 drugs<br />

INH-RIF(%) 13(4.19) 3(1.01) 5(2.08) 1(0.42) 4(1.61) 5(1.80) 31(1.93)<br />

INH-SM(%) 3(0.96) 6(2.01) 5(2.08) 1(0.42) 7(2.82) 5(1.80) 27(1.68)<br />

INH-ETM(%) 1(0.32) 2(0.67) 6(2.50) 5(2.13) 1(0.40) 0 15(0.93)<br />

RIF-SM(%) 0 0 0 0 3(1.20) 0 3(0.18)<br />

RIF-ETM(%) 0 0 0 1(0.42) 0 1(0.36) 2(0.12)<br />

SM-ETM(%) 0 0 1(0.42) 0 0 0 1(0.06)<br />

Resistance to 3 drugs<br />

INH-RIF-SM(%) 1(0.32) 3(1.01) 4(1.67) 0 5(2.02) 4(1.44) 17(1.05)<br />

INH-RIF-ETM(%) 2(0.64) 0 0 0 0 1(0.36) 3(0.18)<br />

INH-SM-ETM(%) 0 0 1(0.42) 3(1.28) 1(0.40) 0 5(0.31)<br />

RIF-SM-ETM(%) 0 0 0 0 0 0 0<br />

Resistance to 4 drugs<br />

INH-RIF-SM-ETM(%) 2(0.64) 0 5(2.08) 3(1.28) 0 2(0.72) 12(0.74)<br />

MDR strains<br />

MDR(%) 21(6.77) 6(2.01) 14(5.83) 4(1.70) 9(3.62) 12(4.33) 63(3.92)<br />

Total(n) n:310 n:298 n:240 n:234 n:248 n:277 n:1607<br />

microbiology laboratory. Only the single and first isolates<br />

of each new patient enrolled in the drug susceptibility<br />

study were used. The test was performed on a total of<br />

1607 isolates from January 2005 to December 2010.<br />

Three hundred and eighty nine (24.20%) isolates were<br />

obtained from females and 1218 (75.80%) were obtained<br />

from male patients. All of the isolates were obtained from<br />

respiratory samples consisting of 113 (7.03%)<br />

bronchioloalveolar lavage (BAL) and 1494 (92.7%)<br />

sputum samples. 1240 (77.16%) of isolates showed sensitivity<br />

to all four drugs. The distribution of the samples<br />

studied over the years, and resistance profiles are<br />

presented completely in Table 2.<br />

Because of global increasing in multidrug resistance<br />

rates we used statistical analysis on type of drug and<br />

years to see the changes in our province. Statistic results<br />

for INH and MDR ratios were analysed by SSPS.15 and<br />

minitab.16 programmes (Table 3). INH drug resistance<br />

rates between 2005-2007 differed a 5% significance level<br />

according to the dual rate comparison over the years.<br />

INH drug resistance rates differed between 2005-2008,<br />

2006-2007 and 2007-2008, and these were statistically<br />

significant according to the dual rate comparison over the<br />

years. The relationship between INH drug sensitivity and<br />

years was determined (chi-square, p=0.025). MDR drug<br />

resistance rates differed between 2005-2008, 2006-2007<br />

and 2007-2008 and these were statistically significant<br />

according to the dual rate comparison over the years.<br />

Between 2005 and 2006, MDR resistance rates differed<br />

at a 5% significance level according to the same method.<br />

Due to previously predicted MDR-TB rates of 20% fort<br />

he present year(2011) in the world, resistance testing in<br />

all high risk cases for drug resistance is recommended by<br />

WHO (WHO/HTM/TB/2010,3). MDR-TB diagnosis and


Table 3. Statistical analysis of isoniazid and multidrug resistance of the study (with SSPS.15 and<br />

minitab.16).<br />

Years<br />

INH resistance<br />

MDR resistance<br />

Z account P Z account P<br />

2005-2006 0.94 0.348 2.34 0.019<br />

2005-2007 -2.44 0.015 -0.25 0.806<br />

2005-2008 -0.29 0.775 2.44 0.015<br />

2005-2009 0.01 0.992 1.11 0.268<br />

2005-2010 -0.76 0.447 0.69 0.491<br />

2006-2007 -3.27 0.001 -2.27 0.023<br />

2006-2008 -1.15 0.252 0.20 0.841<br />

2006-2009 -0.87 0.383 -1.13 0.259<br />

2006-2010 -1.66 0.097 -1.59 0.113<br />

2007-2008 2.04 0.042 2.37 0.018<br />

2007-2009 2.34 0.019 1.23 0.219<br />

2007-2010 1.68 0.093 0.86 0.390<br />

2008-2009 0.28 0.779 -1.26 0.207<br />

2008-2010 -0.43 0.667 -1.71 0.088<br />

2009-2010 -0.73 0.465 -0.42 0.676<br />

treatment must be done according to international<br />

guidelines (WHO/HTM/TB/2010-3; WHO/HTM/STB/2010-<br />

2). The World Report 2008 on antituberculosis drug<br />

resistance’ reported global risks for one or more drug<br />

resistance rates 0-56.3% in new cases, 0- 85.9% in<br />

patients treated previously, and 0-68.9% in all cases<br />

(WHO, 2009). According to the Turkish Ministry of<br />

Health’s 2008 report, 18,452 tuberculous patients were<br />

recorded. The incidence of tuberculosis in Samsun<br />

province in 2008 was 25.8 and the case speed was 28.0<br />

in Samsun. 414 new tuberculosis cases were diagnosed<br />

in 2005, 384 in 2006, 327 in 2007 and 346 in 2008<br />

(Bozkurt et al., 2010).<br />

According to the results in the Turkey (2008) TB report,<br />

a total of 4,963 drug susceptibility patient test were<br />

examined and 19.1% were found to be resistant to at<br />

least one drug. The highest rate of resistance among<br />

drugs belonged to isoniazid. Drug susceptibility tests<br />

detected the total multidrug resistant (MDR-TB) ratio to<br />

be 5.3% (263 people), 3% in new cases and 18.6% in<br />

cases having been previously treated (Table 4) (Bozkurt<br />

et al., 2010). We found 15.74% primary resistance to at<br />

least one drug in this study. Mono drug primary<br />

resistance rates for SM, INH, RIF, ETM were 6.84, 17.17,<br />

5.28 and 4.10% respectively. INH primary resistance<br />

rates are higher than the other major drugs in the present<br />

study and much greater than Turkey’s rates. MDR-TB<br />

has been identified as 3.92%. This rate is lower than the<br />

global estimated average ratio (20%) and Turkey’s ratio<br />

(5.3%) but higher than national primaries (3.0%)<br />

(WHO/HTM/STB/2010-2; Bozkurt et al., 2010). Due to a<br />

low ratio of susceptibility testing in the area (4963 tests in<br />

18452 cases 26.89% for 2008), these results for the<br />

country may not reflect the real resistance state in<br />

Kayhan et al. 2307<br />

Turkey; however, this study reflects the real and nearly<br />

exact drug resistance rates of provincial tuberculosis. The<br />

limitation of our study is not having the secondary<br />

resistance rates at the same time.<br />

In recent years, a variety of resistance test studies<br />

against major drugs have been performed in our country<br />

and the comparative results are shown in Table 4.<br />

We have also analysed the rates of primary, secondary<br />

and total resistance in Turkey in Table 5, by using the<br />

2005-2008 data of Turkish Ministry of Health (Bozkurt et<br />

al., 2010). Secondary resistance rates were extremely<br />

higher than primaries. The ratio of drug resistance to INH<br />

was high compared with the other major drugs. In<br />

addition, multiple and single drug resistances in these<br />

years were close to each other on some level in our<br />

country and in our study.<br />

Regional differences in the resistance prevalence<br />

studies were published by authors. Arseven and his<br />

colleagues reported results from the provinces of the<br />

Eastern Black Sea region between the years 1985 and<br />

1990. A total of 564 (40.6%) of the 1388 culture-positive<br />

TB patients were determined to be resistant to at least<br />

one of the following: INH, RIF, SM, and ETM. Drug<br />

resistance rates against INH, RIF, SM and ETM were<br />

29.6, 17.1, 23.3 and 8.8% respectively. They measured<br />

the poly (more than one) drug-resistance rate to be<br />

22.4%, and the rate of MDR strains to be 13% (Arseven<br />

et al., 1995). Saral and his colleagues found the rates of<br />

resistance to INH, RIF, SM and ETM in a study as 24.6,<br />

15.8, 9.9 and 18.8% respectively. In the same study, the<br />

rate of MDR-MTC was reported as 14.7% (Saral et al.,<br />

2007). Aydın et al. (2011) performed a similar study in<br />

Trabzon and found the single drug resistance to INH to<br />

be 6.1%, RIF 0.5% , SM 5.2% and ETM 2.4%. In the


2308 Afr. J. Microbiol. Res.<br />

Table 4. Some study results about primary resistance in Turkey.<br />

Reference City/Region Date<br />

Number of isolates<br />

(susceptibility test studied)<br />

Primary resistance rates of major anti-TB drugs<br />

SM(%) RIF(%) INH(%) EMB(%)<br />

Turkey (2008) Whole country 2005-2008 15735 8.37 6.77 13.37 4.23<br />

Present study Samsun 2005-2010 1607 6.84 5.28 17.17 4.10<br />

Yolsal et al Regional metaanalysis 1984-1989 368 8.8 5.7 14.4 2.2<br />

Yolsal et al Regional metaanalysis 1990-1995 2848 10.1 8.9 8.8 3.0<br />

Do an et al Sivas 1999-2004 316 15.2 4.1 19.9 2.5<br />

Güneri et al Aegean region 1999-2001 387 0 5.7 12.4 0<br />

Çetinkaya et al Elazı 1989-1994 125 16.6 7.6 11.5 1.2<br />

Sürücüo lu et al Manisa 1997-2003 285 13.3 6.0 14.4 8.4<br />

Talay et al. (2003) stanbul 1997-2000 135 13.3 3.0 8.8 2.2<br />

Otkun et al (38) Edirne 1996 44 32.0 11.0 30.0 9.0<br />

Karada et al. (2004) Samsun 2004 50 4.0 4.0 8.0 2.0<br />

Aydın et al. (2011) Zonguldak 2003-2005 99 13.1 2.0 18.2 3.0<br />

Aydın et al. (2011) Trabzon 2005-2010 212 13.7 5.7 17.5 5.7<br />

Korkmaz et al. (2002) Gaziantep 2002-2003 104 2.89 10.58 25 18.27<br />

Korkmaz et al. (2002) Gaziantep 2002-2003 104 2.89 10.58 25.04 25.96<br />

same study, of the 212 isolates, 25 (11.8%)<br />

strains were multiple drug resistant 10(4,7%)<br />

(Aydın et al., 2011). Many studies have been<br />

performed on relevant city analysis of TB for<br />

mono drug and multidrug resistance ( enol et al.,<br />

2004; Uçar et al., 2010). For example Aydın et al.<br />

(2011) analysed 125 strains in the province of<br />

Zonguldak and reported the sensitivity rate to all<br />

drugs as 69.6%, resistance rate to INH as 23.2%<br />

and the rate of multidrug resistance as 8% in 2005<br />

(Aydın et al., 2008). In our country, it has been<br />

reported that INH+RIF resistant strains are 2.7%<br />

in Isparta, 7 and 3% (two studies done at different<br />

times) in Edirne, and 12.8 and 19.6% in<br />

Gaziantep (two different studies) (Yaylı et al.,<br />

2003; Tansel et al., 2003; Balcı et al., 1999; Gani<br />

et al., 2002). Another study reported resistance<br />

rates in our city (Samsun) as 8% to INH, 4% to<br />

RIF, 4% to SM, 4% to ETM, 2% to pyrazinamide<br />

and 4% to MDR-TB strains in 2004 (Karada et<br />

al., 2004).<br />

The prevalence of MTC drug resistance varies<br />

from one part of the world to another (Jaffar et al.,<br />

2005). In the United States, drug-resistant<br />

tuberculosis was detected in 14.2% in 1991 and<br />

10% in 1997 (Bloch et al., 1994; Espinal et al.,<br />

2001). In the United States, isoniazid resistance<br />

was the most prevalant and accounted for 8%.<br />

Isoniazid resistance has ranged from 0% in New<br />

Caledonia to 7.9% in Mozambique, and was 10%<br />

in India (Pereira et al., 2005). An article from<br />

Saudi Arabia reported the fact that the rate of<br />

resistance to isoniazid varied from one part of the<br />

country to another such as: 4.2-7.2% in Riyadh,<br />

6% in Dammam and Taif, 10.3- 28.7% in Jeddah<br />

and 41% in Gizan. A 15-year study found rates of<br />

resistance to INH in different drug concentrations<br />

as follows: 12.5% resistance in INH (1 g/mL),<br />

and 2.9% resistance in INH (5 g/mL) in Dhahran<br />

(Jaffar et al., 2005). The prevalence of MDR-TB<br />

among new TB cases may differ in diverse<br />

geographies such as 14% in Estonia, 9% in<br />

China, Henan province, 9% in Lithuania, 9% in<br />

Russia, Ivanovo province, 5% in Iran and 4.5% in<br />

China, Zhejiang province (Espinal et al., 2001). In<br />

the early stages of tuberculosis treatment, the<br />

most potent bactericidal drug is isoniazid. INH<br />

resistance may also be an indicator of success in<br />

the treatment of tuberculosis. Storla and<br />

colleagues reported a high treatment failure in<br />

areas with high resistance to INH in Bangladesh<br />

(Storla et al., 2007).<br />

Directly observed therapy (DOT) in TB patients<br />

has been successfully completed in some<br />

countries such as the United States and a significant<br />

fall in TB and MDR-TB cases was provided<br />

contrary to the global rising in the number of


Table 5. The rates of primary, secondary and total drug resistance types in Turkey in 2005-2008.<br />

Drugs Resistance type 2005 (%) 2006 (%) 2007 (%) 2008 (%)<br />

INH Primary 9.0 10.7 11.9 11.3<br />

secondary 27.4 23.8 27.6 27.9<br />

Total 11.5 12.6 14.4 13.8<br />

RIF Primary 4.4 4.5 4.9 3.9<br />

Secondary 21.1 19.8 18.7 21.8<br />

Total 6.7 6.7 7.1 6.6<br />

ETM Primary 3.0 3.6 2.8 3.4<br />

Secondary 10.0 13.2 8.3 9.6<br />

Total 4.0 5.0 3.6 4.3<br />

SM Primary 7.0 8.4 7.1 6.5<br />

Secondary 15.2 17.0 13.8 12.9<br />

Total 8.1 9.7 8.2 7.5<br />

MDR-TB Primary 3.1 3.2 2.9 3.0<br />

Secondary 17.7 16.6 15.5 18.6<br />

Total 5.1 5.1 4.9 5.3<br />

multidrug-resistant tuberculosis. WHO declared the fact<br />

that between 1995 and 2009, a total of 41 millionTB<br />

patients were successfully treated in DOT, and up to six<br />

million lives were saved, including two million women and<br />

children.<br />

In conclusion, resistance to INH and RIF continues,<br />

almost in a straight line in Samsun Province. DOT must<br />

not be disregarded due to global rises in drug resistant<br />

MTC strains. Surveillance of the primary resistance<br />

patterns is important in assessing the quality of<br />

chemotherapy programs over several years.<br />

REFERENCES<br />

Arseven O, Eraksoy H, Uzun Y, Sepkin C, Kalaycıoglu A, Ozmenoglu<br />

M, Bolukbası O (1995). The resistance rates of Mycobacterium<br />

tuberculosis strains to antituberculosis drugs in the Eastern Blacksea<br />

Region Turkey. J. Klimik, 8(2): 63-67.<br />

Aydın F, Kaklıkkaya N, Bayramoglu G, Ozkul G, Buruk K, Dinc U, Kose<br />

T, Dede R (2011). Resistance Rates of Mycobacterium tuberculosis<br />

complex strains isolated from Clinical Specimens. Mikrobiyol Bull.,<br />

45(1): 36-42.<br />

Aydın O, Begendik CF, Kulah C, Aktas E, Sumbuloglu V (2008).<br />

Determination of susceptibilities of Mycobacterium tuberculosis<br />

strains isolated in Zonguldak to primary anti-tuberculosis drugs via<br />

BACTEC MGIT 960 system. Mikrobiyol. Bull., 38(2): 61-70<br />

Balcı , Bayram A, Filiz A (1999). Resistance in Mycobacterium<br />

tuberculosis to first line drugs. Tur. J. Infect., 13(4): 521-525.<br />

Bloch AB, Cauthen GM, Onorato IM (1994). Nationwide survey of drugresistant<br />

tuberculosis in the United States. JAMA, 271: 665–671<br />

Bozkurt H, Türkkanı MH, Musaonba ıo lu S, Güllü Ü, Yıldırım A, Baykal<br />

A, Özkara (2010). Warfare against tuberculosis in Turkey, 2010<br />

Turkish Ministry for Health report.<br />

Brennan JP, Draper P (1994). Ultrastructure of Mycobacterium<br />

tuberculosis. In: Bloom BR, ed. Tuberculosis: Pathogenesis,<br />

Protection, and Control. Washington DC: ASM Press, pp. 271–84.<br />

Kayhan et al. 2309<br />

Centers for Disease Control and Prevention (1993). Initial therapy for<br />

tuberculosis in the era of multidrug resistance: recommendations of<br />

the advisory council for the elimination of tuberculosis. MMWR Mortal<br />

Morbid Weekly Rep., 42 (RR-7):1.<br />

De Rossi E, Ainsa JA, Riccardi G (2006). Role of mycobacterial efflux<br />

transporters in drug resistance: an unresolved question. FEMS<br />

Microbiol. Rev., 30(1): 36-52.<br />

EuroTB (2007). Definitions for the WHO/EuroTB Joint Tuberculosis<br />

Data Collection. EuroTB,www.eurotb.org .<br />

Espinal MA, Laszlo A, Simonsen L (2001). Global trends in resistance<br />

to anti-tuberculosis drugs: World Health Organization-International<br />

Union against Tuberculosis and Lung Disease Working Group on<br />

Anti-Tuberculosis Drug Resistance Surveillance. N. Engl. J. Med.,<br />

344: 1294–1303.<br />

Gani O, Zer Y, Balcı , Bayram A, Korkmaz G (2002). Retrospective<br />

analysis of sample examined in Mycobacteriology laboratory.<br />

Mikrobiyol Bul., 32(3-4):225-9.38(2): 61-70.<br />

Inderlied CB, Nash KA (1996). Antimicrobial agents: in vitro<br />

susceptibility testing, spectra of activity, mechanisms of action and<br />

resistance, and assays for activity in biologic fluids. In: Lorian V, ed.<br />

Antibiotics in Laboratory Medicine. 3rd ed. Baltimore: Williams &<br />

Wilkins., pp. 127–175.<br />

Jaffar A, Amal A, Mahmoud S (2005). Susceptibility Pattern and<br />

Epidemiology of Mycobacterium tuberculosis in a Saudi Arabian<br />

Hospital: A 15-Year Study From 1989 to 2003. Chest, 128:5 3229-<br />

3232; doi:10.1378/chest.128.5.3229.<br />

Karada A, Tokaç M, Güvenli A, Sünbül M, Günaydın M, Saniç A<br />

(2004). Resistance Ratio to Major Antituberculosis Drugs of<br />

Tuberculosis Complex Bacilli Isolated from Clinical Samples ANKEM<br />

Derg., 18(4): 189-192.<br />

Pereira M, Tripathy S, Inamdar V (2005). Drug resistance pattern of<br />

Mycobacterium tuberculosis in seropositive and seronegative HIV-TB<br />

patients in Pune, India. Indian J. Med. Res., 121: 235–239.<br />

Saral BÖ, Sucu N, Aktoz Boz G, Erdem M, Köksal (2007). Evaluating<br />

drug resistance in 442 Mycobacterium tuberculosis strains with<br />

BACTEC method. Tur. J. Thorax, 8(3): 174-178.<br />

Storla DG, Rahim Z, Islam MA (2007). Drug resistance of<br />

mycobacterium tuberculosis in the Sunamganj district of Bangladesh.<br />

Scand. J.Infect. Dis., 39: 142-145.<br />

enol G, Kömürcüo lu B, Kömürcüo lu A (2004). Resistance rates to


2310 Afr. J. Microbiol. Res.<br />

anti-tuberculosis drugs in Mycobacterium tuberculosis strains. Tur. J.<br />

Infect., 18(4): 441-445.<br />

Talay F, Altın S, Karasulu L, Kümbetli (2003). Drug Resistance Rates<br />

In Eyup-Istanbul Tuberculous Struggle Dispensary Between 1997-<br />

2000. Van Med. j., 10(1): 9-15.<br />

Tansel Ö, Yüksel P, Kulo lu F, Akata F (2003). Resistance of<br />

Mycobacterium tuberculosis strains to anti-tuberculosis drugs: Two<br />

years results of Trakya University. Tur. J Infect., 17(1): 23-26.<br />

Ta ova Y, Yaman A, Salto lu N, Erdurak Ö, nal S, Dündar H (1997).<br />

The resistance rates of Mycobacterium tuberculosis strains to<br />

antituberculous drugs in Çukurova University Balcalı Hospital Central<br />

Laboratory. Tur. J. Infect., 11(2): 97-101.<br />

Ucar E, Kılıc A, Ceyhan I (2010). Resistance rates to major antituberculosis<br />

drugs in Mycobacterium tuberculosisstrains isolated from<br />

seven different regions of Turkey in the 2003-2006 period. Mikrobiyol<br />

Bul., 44(1): 11-19.<br />

Yaylı G, Sözen H, A alar C (2003). Susceptibilities of Mycobacterium<br />

tuberculosis strains isolated in Isparta Region to antituberculous<br />

drugs Mikrobiyol Bull., 33: 24-30.<br />

WHO (2010) global report on surveillance and response. Geneva,<br />

World Health Organization, 2010 (WHO/HTM/TB/2010-3).<br />

WHO (2010). The Global Plan to stop TB 2011-2015,Geneva, World<br />

Health Organization,2010 (WHO/HTM/STB/2010-2).<br />

WHO (2009). Anti-tuberculosis Drug Resistance in the World. Fourth<br />

Global Report. www.who.int/tb/publications/2008/<br />

drsreport4_26feb08.pdf.


African Journal of Microbiology Research Vol. 5(16), pp. 2311-2315, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.622<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Cytokine genes expression in mice hepatocytes during<br />

malaria infection<br />

Saad Alkahtani 1 *, AL-Farraj S. A. 1 , Saud A. Alarifi 2 , AL-Eissa Mohammed saad 3 and<br />

Al-Dahmash B. 4<br />

1 Department of Biology, Teachers College, King Saud University, Riyadh, Saudi Arabia.<br />

2 Department of Zoology, Science College, King Saud University, Riyadh, Saudi Arabia.<br />

3 Department of Biology, Science College, Hail University, Hail, Saudi Arabia.<br />

4 Department of Medical Laboratory, College of Health Sciences, King Saud University, Riyadh, Saudi Arabia.<br />

Accepted 30 July, 2011<br />

The aim of this study was to investigate the inflammation genes (IL-1 , IL-6, TNF- and iNos) responses<br />

to Plasmodium chabaudi malaria in the liver of female C57BL/6 hepatocytes via mRNA expression due<br />

to infection with P. chabaudi at different time points. Mice were injected intraperitoneally (ip) with 10 6 P.<br />

chabaudi-infected erythrocytes and then scarified at days (0, 1, 4 and 8 respectively). RT-PCR was used<br />

to quantify liver inflammation genes. The levels of IL-1 , IL-6 and TNF were significantly increased at<br />

days 1 and 8. The total iNos were significantly increased at all days after infection. In conclusion,<br />

present data has shown that infection with P. chabaudi stimulated infalammation genes in the liver.<br />

Thus, we suggest the implication of oxidative stress due to outcome of malaria in mice hepatocytes<br />

according to its natural function need to be confirmed with a larger number of samples to be used as a<br />

reliable inflammation detection method.<br />

Key words: Inflammation, malaria, mice, liver.<br />

INTRODUCTION<br />

Despite decades of intense research, malaria remains a<br />

major global health problem with an estimated mortality<br />

of 1-2 million (Alkahtani, 2011). Due to increasing the<br />

resistance of parasites no effective vaccine is available<br />

and all efforts to develop an effective anti-malaria vaccine<br />

have failed to date (Bergmann-Leitner et al., 2009;<br />

Alkahtani, 2010). However, several vaccine candidates<br />

and vaccine platforms have yielded encouraging results<br />

in animal models of malaria, but have not been<br />

sufficiently evaluated in clinical trials. A rather convenient<br />

malaria model system is the Plasmodium chabaudi blood<br />

stage infection in mice, since P. chabaudi shares several<br />

characteristics with P. falciparum, the most dangerous<br />

human malaria species (Hernandez-Valladares et al.,<br />

2005).<br />

Liver plays a very important role in malaria: It is the site<br />

*Corresponding author. E-mail: salkahtani@ksu.edu.sa.<br />

of pre-erythrocytic development of Plasmodium parasites,<br />

and also acts as an effector against malarial blood stages<br />

as recently shown (Krücken et al., 2005b; Wunderlich et<br />

al., 2005). Kupffer cells are able to eliminate parasitederived<br />

hemozoin and even Plasmodium-infected<br />

erythrocytes. As a result of this process, Kupffer cells<br />

become increasingly activated, which manifests itself as<br />

a production of NO (nitrogen oxide), diverse ROS (radical<br />

oxygen species), and the cytokines IL-1 and TNF . In<br />

addition, cytokines have primary roles in physiology,<br />

including neurophysiology, innate and acquired immune<br />

responses and wound healing, as well as pathogenesis.<br />

Some cytokines, such as TNF, LTs and IL-1 are proinflammatory,<br />

whereas others such as IL-4, IL-10 and the<br />

TGF family, are anti-inflammatory, being subsequently<br />

induced to inhibit these changes (Clark et al., 2010). NO,<br />

ROS and the cytokines result in enormous pathological<br />

complications in the liver. Indeed, IL-1 and TNF are<br />

thought to induce the acute phase response and a<br />

dramatic decrease in liver metabolism, both majorly


2312 Afr. J. Microbiol. Res.<br />

based on liver cells (Krücken et al., 2005a; Guha et al.,<br />

2006). Recent results with deficient mice in the<br />

expression of those cytokines indicate that they are in<br />

general more sensible to insults resulting in neural<br />

damage. Some of the actions induced by TNF- and IFNincluding<br />

both beneficial and detrimental, are mediated<br />

by inducible nitric oxide synthase (iNOS) -derived nitric<br />

oxide (NO) production (Munoz-Fernandez and Fresno.,<br />

1998).<br />

The present study was undertaken to investigate if the<br />

inflammation genes responses to Plasmodium chabaudi<br />

malaria in the liver of mice and to detect if these genes<br />

could be a good marker for inflammation and metabolism<br />

instability due to infection with Plasmodium chabaudi in<br />

mice hepatocytes.<br />

MATERIALS AND METHODS<br />

Animals and infection<br />

Normal female C57BL/6 mice 10-14 weeks old were obtained from<br />

the central animal facilities of Heinrich Heine University, Düsseldorf,<br />

Germany, and housed in plastic cages. Mice were bred and<br />

maintained under specified pathogen free conditions. Mice were fed<br />

with standard diet (Wohrlin, Bad Salzuflen, Germany) and water ad<br />

libitum. The experiments were approved by the State authorities<br />

and followed German law on animal protection.<br />

Blood stage malaria<br />

A non-clonal line of P. chabaudi was used (Wunderlich et al., 1988)<br />

exhibiting a very similar, but not identical restriction length<br />

polymorphism pattern to P. chabaudi (Kruecken et al., 2005b).<br />

Erythrocytic stages of P. chabaudi were passaged weekly in NMRI<br />

mice. From these mice, blood was taken and 10 6 P. chabaudiinfected<br />

erythrocytes were injected i.p. in the mice. Parasitemia was<br />

evaluated in Giemsa-stained blood smears. The total number of<br />

erythrocytes was determined in a Neubauer chamber. A total of five<br />

mice were scarified at each day (0, 1, 4 and 8 respectively) by<br />

cervical dislocation. Livers were removed and cut into smaller<br />

pieces and kept at -80°C.<br />

RNA-Isolation<br />

Approximately 250 mg frozen liver was homogenized with an<br />

ultraturrax in 5 ml Trizol (Peqlab Biotechnology, Erlangen,<br />

Germany) for one minute. After mixing with 1 ml chloroform for 15 s,<br />

the suspension was incubated for 15 min at room temperature and<br />

centrifugated at 3.000 x g for 45 min. After isopropanol precipitation<br />

of the supernatant, the pellet was washed twice with 80% ethanol<br />

and air-dried and dissolved in 200 µl RNase-free water. RNA<br />

concentrations were determined at 260 nm, and the purity of RNA<br />

was checked in 1% agarose gel.<br />

Quantitative real-time PCR<br />

All RNA samples were treated with DNase (Applied Biosystems,<br />

Darmstadt, Germany) for at least 1 h and then converted into cDNA<br />

following the manufacturer’s protocol using the Reverse<br />

Transcription Kit (Qiagen, Hilden, Germany). Quantitative real-time<br />

polymerase chain reaction (qRT-PCR) was performed using the ABI<br />

Prism ® 7500HT Sequence Detection System (Applied Biosystems,<br />

Darmstadt, Germany) with SYBR Green PCR Mastermix from<br />

Qiagen (Hilden, Germany). We investigated the genes encoding the<br />

mRNA for following proteins: 18S, Interleukin-1beta (IL-1 ), Tumor<br />

necrosis factor alpha (TNF ), Interleukin-6 (IL-6), Inducible nitric<br />

oxide synthase iNOS. All primers used for qRT-PCR were<br />

commercially obtained from Qiagen. PCR reactions were conducted<br />

as follows: 2 min at 50°C to activate uracil-N-gylcosylase, 95°C for<br />

10 min to deactivate UNG, 40 cycles at 94°C for 15 s, at 60°C for<br />

35 s and at 72°C for 30 s. Reaction specificity was checked by<br />

performing dissociation curves after PCR. For quantification, mRNA<br />

was normalized to 18S rRNA. The threshold Ct value is the cycle<br />

number selected from the logarithmic phase of the PCR curve in<br />

which an increase in fluorescence can be detected above<br />

background. The Ct is determined by subtracting the Ct of 18S<br />

rRNA from the Ct of the target ( Ct = Ct-target – Ct-18S rRNA).<br />

The relative mRNA levels of non-infected mice are described as a<br />

ratio of target mRNA copy to 18S rRNA copy = 2 – Ct . The fold<br />

induction of mRNA expression on days (0, 1, 4 and 8 p.i. was<br />

determined using the 2 - ct -method (- ct = ct day 0 p.i. - ct day<br />

8 p.i.).<br />

Blood analysis<br />

In sera, IL-1 , IL-6, TNF were measured using ELISA kits (R&D<br />

Systems, Minneapolis, USA) according to the manufacturer's<br />

protocols. Total iNOS was analyzed using a commercially available<br />

kit (R&D Systems).<br />

Statistical analysis<br />

Two-tailed Student’s t-test and Fisher’s exact test were used for<br />

statistical analysis.<br />

RESULTS<br />

Characteristics of P. chabaudi infection<br />

The parasitemia of mice with 10 6 P. chabaudi-infected<br />

erythrocytes became evident on day 4 and rocketed to<br />

reach its peak (48%) on day 8 and then reduced rapidly<br />

to about 0.4% on day 12 (Figure 1).<br />

Inflammation genes expression<br />

Quantitative real-time PCR was used to detect changes<br />

in mRNA levels of different inflammation genes. The<br />

levels of IL-1 , IL-6 and TNF were significantly (P


-Fold change in mRNA expression<br />

Alkahtani et al. 2313<br />

Figure 1. Parasitemia (48%) of female C57BL/6 mice (n=20) infected with 10 6 P. chabaudi-infected erythrocytes.<br />

Figure 2. RT-PCR of IL-1 , IL-6, TNF and iNOS genes expression in the liver of Plasmodium chabaudi infected<br />

mice. The expression of these genes was measured at different times. The data present are the mean ±SE (n=5).<br />

*: significant value at (P


2314 Afr. J. Microbiol. Res.<br />

Table 1. Serum parameters of control (day 0) and infected (day 8) mice with 10 6 P. chabaudi.<br />

Parameters in blood sera 0 day 8 days p.i. p-value 8d p. i. vs. 0d<br />

Parasitemia 0.00 % 48 % < 0.01<br />

IL-1 [ng/l] 1.17 ± 0.2 4.5 ± 1.3


Clark I, Alleva L, Vissel B (2010). The roles of TNF in brain dysfunction<br />

and disease. Pharmacol. Therap., 128(3): 519-548.<br />

Clarka I, Allevaa L, Cowdenb W (2008). Understanding the role of<br />

inflammatory cytokines in malaria and related diseases. Travel Med.<br />

Infect. Dis., 6: 67-81.<br />

Guha M, Kumar A, Choubey V, Maity P, Bandyopadhyay U (2006).<br />

Apoptosis in liver during malaria: role of oxidative stress and<br />

implication of mitochondrial pathway. The FASEB J., 20: E439-E449.<br />

Guha M, Maity P, Choubey V, Mitra K, Reiter R, Bandyopadhyay U<br />

(2007). Melatonin inhibits free radical-mediated mitochondriadependent<br />

hepatocyte apoptosis and liver damage induced during<br />

malaria infection. J. Pineal Rea., 43(4):372-381.<br />

Häussinger D, Schliess F (2008). Pathogenetic mechanisms of hepatic<br />

encephalopathy. Gut, 57: 1156-1165.<br />

Hernandez-Valladares M, Naessens J, Iraqi FA (2005). Genetic<br />

resistance to malaria in mouse models. Trends Parasitol., 21: 352-<br />

355.<br />

Krücken J, Dkhil MA, Braun JV, Schroetel RMU, El-Khadragy M,<br />

Carmeliet P, Mossmann H, Wunderlich F (2005)b. Testosterone<br />

suppresses protective responses of the liver to blood-stage malaria.<br />

Infect. Immun., 73: 436-443.<br />

Alkahtani et al. 2315<br />

Krücken J, Mehnert LI, Dkhil MA, El-Khadragy M, Benten WPM,<br />

Mossmann H, Wunderlich F (2005)a. Massive destruction of malariaparasitized<br />

red blood cells despite spleen closure. Infect. Immun., 73:<br />

6390-6398.<br />

Munoz-Fernandez M, Fresno M (1998). The role of tumour necrosis<br />

factor, interleukin 6, interferon- and inducible nitric oxide synthase in<br />

the development and pathology of the nervous system. Progress<br />

Neurobiol., 56(3): 307-340.<br />

Nussler A, Silvio M, Liu Z, Geller D, Freewick P, Dorko K, Bartoli F,<br />

Billiar T (1995). Further characterization and comparison of inducible<br />

nitric oxide synthase in mouse, rat, and human hepatocytes.<br />

Hepatology, 21(6): 1552-1560.<br />

Wunderlich F, Dkhil MA, Mehnert LI, El-Khadragy M, Borsch E,<br />

Hermsen D, Benten WPM, Pfeffer K, Mossmann H, Krücken J<br />

(2005).Testosterone-responsiveness of spleen and liver in female<br />

lymphotoxin receptor-deficient mice resistant to blood stage<br />

malaria. Microbes Infect., 7: 399-409.<br />

Wunderlich F, Mossmann H, Helwig M, Schillinger G (1988).<br />

Resistance to Plasmodium chabaudi in B10 mice: influence of the<br />

H2-complex and testosterone. Infect. Immun., 9: 2400-2406.


African Journal of Microbiology Research Vol. 5(16), pp. 2316-2324, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.625<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Phylogenetic analysis of the nematicidal actinobacteria<br />

from agricultural soil of China<br />

Xu Chuan Kun 1 , Lou Xiao Jun 1 , Xi Jia Qin 2 , Gu Lei 3 , Duan Chang Qun 1 , Mo Ming He 1 *,<br />

Zhang Ke Qin 1 , Yang Fa Xiang 4 and Fang Dun Huang 5<br />

1 Laboratory for Conservation and Utilization of Bio-Resources and Key Laboratory for Microbial Resources of the<br />

Ministry of Education, Yunnan University, Kunming, 650091, Yunnan Province, P. R. China.<br />

2 Zhengzhou Tobacco Research Institute of China Netherlands Trade Company (CNTC), Zhengzhou, 45000, P. R. China.<br />

3 Yunnan University Secondary School, Kunming, 650091, Yunnan Province, P. R. China.<br />

4 Yunnan Yunye Fertilizer Company Limited, Kunming, 650217, P. R. China.<br />

5 Yunnan Academy of Tobacco Agricultural Science, Yuxi 653100, P. R. China.<br />

Accepted 30 July, 2011<br />

The purpose of this study was to assess the diversity and biocontrol potential of nematicidal<br />

actinobacteria from agricultural soils. Two hundred soil samples were collected from 20 provinces<br />

(autonomous regions or municipalities) of China, and 4000 actinobacteria isolates were obtained. Of the<br />

4000 isolates evaluated, 533 (13.3% of total) and 488 (12.2%) respectively showed nematicidal<br />

properties to target nematodes Panagrellus redivivus and Bursaphelenchus xylophilus with nematicidal<br />

activities (NA) of more than 30%. The sum of strains with NAs of 90-100%, 80-90%, 60-80% and 30-60%<br />

was 55, 100, 127 and 251 to P. redivivus, while 37, 85, 111 and 255 to B. xylophilus, respectively. The<br />

most active isolates showed high host selectivity, in which only 101 isolates (5.1% of the total) were<br />

toxic to the both targets with NAs>30%. Additionally, 69 of the 101 isolates were randomly selected for<br />

species diversity analysis. Phylogenetic analysis placed the 69 actinobacteria in three families<br />

(Streptomycetaceae, Pseudonocardiaceae and Nocardiaceae) of the Actinobacteria with sequence<br />

similarity of 97.4-100%. The largest group was Streptomycetaceae, containing 58 isolates (84.1% of the<br />

total) that showed 97% to 100% sequence identity to 28 species of the genera Streptomyces (57<br />

isolates, 27 species) and Kitasatospora (1, 1). The Pseudonocardiaceae group contained 7 isolates<br />

(10.1% of the total) showing 98.5-99.7% homology to Amycolatopsis lurida (4 isolates) and A.<br />

niigatensis (3). The Nocardiaceae group included 4 isolates (5.8% of the total) with 99-99.6% sequence<br />

identity to one species N. fluminea.<br />

Key words: Nematicidal actinobacteria, nematode, biocontrol, phylogenetic analysis.<br />

INTRODUCTION<br />

Plant parasitic nematodes cause damages to a variety of<br />

agricultural crops throughout the world. Only the rootknot<br />

nematodes (Meloidogyne spp.) cause about US$100<br />

billion loss annually to a wide variety of crops worldwide<br />

(Oka et al., 2009). Currently, the application of chemical<br />

nematicides and fumigants is still the main strategy to<br />

*Corresponding author. E-mail: minghemo@yahoo.com.cn. Tel:<br />

+86871 5031396. Fax: +86871 5034838.<br />

control these pathogens. Although chemical nematicides<br />

are effective, easy to apply, and show rapid effects, they<br />

have begun to be withdrawn from the market in some<br />

developed countries owing to concerns about public<br />

health and environmental safety problems (Schneider et<br />

al., 2003). At present, several successful biocontrol<br />

agents mainly using nematophagous fungi have been<br />

used widely for nematode diseases (Tikhonov et al.,<br />

2002). As antagonists of parasitic nematodes,<br />

nematophagous fungi exhibited their biocontrol traits by


predation, parasitism or poisonous effects.<br />

Extracellular enzymes of nematophagous fungi, especially<br />

the serine protease, served as the vital virulence<br />

factor in the process of infection (Huang et al., 2004). A<br />

few species of antagonistic bacteria have been<br />

developed into agents against plant parasitic nematodes,<br />

e.g. Bacillus thuringiensis and Pasteuria penetrans (Chen<br />

and Dickson, 1998; Javed et al., 2008). B. thuringiensis<br />

exhibited toxic effects against invertebrate by its<br />

parasporal inclusion and has been used widely (Wei et<br />

al., 2003; Meadows et al., 1989). P. penetrans is an<br />

obligate parasite of root knot nematodes (Meloidogyne<br />

spp.), but its fastidious life cycle and the obligate nature<br />

of parasitism have inhibited its mass culturing and<br />

deployment in field conditions. Actinobacteria, however,<br />

have so far received relatively little attention as potential<br />

biocontrol agents against plant-parasitic nematodes.<br />

Actinobacteria is a group of gram positive bacteria with<br />

a G+C content of over 55%. More than 70% of bioactive<br />

compounds are produced from these microorganisms<br />

and they have shown significant applications in pharmacy,<br />

industry, agriculture and environmental protection.<br />

Members of this group are best known for their ability to<br />

produce lytic enzymes, various secondary metabolites,<br />

including antibiotics. For example, avermectins includes a<br />

series of macrocyclic lactone derivatives produced by<br />

Streptomyces avermitilis that belongs to a new family of<br />

potent antihelminthic agents (Burg et al., 1979). Since the<br />

early 1980s, the avermectins and their derivatives have<br />

been used widely in the world to control parasitic<br />

nematodes and pests. The discovery and the huge commercial<br />

market of avermectins promoted an increasing<br />

number of researchers to study the biocontrol potential of<br />

actinobacteria. For example, Mishra et al. (1987) found<br />

that metabolites from 15 isolates of aerobic<br />

actinobacteria (screened from 502 actinobacteria) were<br />

toxic to the free-living nematode Panagrellus redivivus<br />

(Mishra et al., 1987).<br />

A species of the genus Streptomyces isolated from<br />

nematode suppressive soil inhibited the reproduction of<br />

Caenorhabditis elegans in the laboratory test and<br />

reduced tomato root galling caused by M. incognita in the<br />

greenhouse experiment (Dicklow et al., 1993). Some<br />

nematicidal actinobacteria were observed in eggs and<br />

cysts of Heterodera glycines (Nour et al., 2003) and H.<br />

trifolii (Hay and Skipp, 1993). Sun et al. (2006) isolated<br />

30 actinobacteria from root-knot samples and found that<br />

47% of them were virulent to eggs and juveniles of M.<br />

hapla.<br />

These investigations indicated that nematicidal<br />

actinobacteria are abundant in agricultural environments.<br />

However, no study has been performed to analyze the<br />

diversity and phylogenetic relationships of nematicidal<br />

actinobacteria from a wide range of agricultural soils. The<br />

objective of this study was to estimate the relative<br />

biocontrol potential of actinobacteria isolated from 20<br />

provinces (municipalities or autonomous regions) of<br />

China. We also presented a phylogenetic analysis to<br />

Kun et al. 2317<br />

characterize these nematode-antagonistic microorganisms.<br />

MATERIALS AND METHODS<br />

Soil sampling<br />

A total of 200 agricultural soil samples were collected from 20<br />

provinces (municipality or autonomous regions) of China (Figure 1).<br />

In each province (municipality or autonomous region), 10 samples<br />

were collected. For each sample, approximately 1 kg of soil was<br />

sampled randomly from the top layer (2-15 cm) over an area of<br />

more than 5 m 2 per field. Soil samples were spread out to air dry at<br />

room temperature for 2-3 days and sieved through a 2 mm sieve,<br />

then stored in glass bottles at 4°C until used.<br />

Isolation of actinobacteria<br />

To isolate the soil actinobacteria, 1 g of each dry sample was<br />

suspended in 10 m1 sterile distilled water and diluted 1000-fold. 0. l<br />

ml of the dilutions was spread on ISP5 medium (ISP5 medium: Lasparagine<br />

1g, glycerol 10g, K2HPO4 1 g, trace salts 1 ml, agar 18<br />

g, pH 7.2, added to 1000 ml with ddH2O, where the trace salts<br />

including FeSO4•7H2O 0.2 g, MnCl2•2H2O 0.1 g, ZnSO4•7H2O 0.1 g,<br />

added to 100 ml with ddH2O). The plates were incubated at 28°C<br />

for 2 weeks and actinobacteria were purified from single colonies<br />

using the same medium.<br />

Nematode inocula<br />

In this study, a free-living nematode, Panagrellus redivivus, and a<br />

pinewood nematode, Bursaphelenchus xylophilus, were used as<br />

target hosts for assaying the in vitro nematicidal activity of the<br />

actinobacteria. P. redivivus was cultured on autoclaved oatmeal as<br />

previously described (Walker and Barrett, 1991) and B. xylophilus<br />

was fed with a fungus as described by Dong et al. (2004). The<br />

juveniles were separated from media through the Baermann funnel<br />

technique (Gray, 1984), and suspended in the water for use.<br />

Nematicidal activity assay of actinobacteria<br />

Each purified isolate was inoculated into 50 ml of ISP5 liquid<br />

medium in a 150 ml flask. After incubation of 2 weeks at 28°C, 200<br />

rpm, the culture suspension without actinobacteria was obtained by<br />

sterile filtration and used for nematicidal activity assay. Briefly, 1 ml<br />

of the culture suspension was added into a well of a 24-well cell<br />

culture plate, and 0.1 ml nematode suspension (approximate 150<br />

juveniles) was added and mixed. After incubation at 28°C for 24 h,<br />

the mobile (live) and immobile juveniles were recorded by counting<br />

>150 individuals under a microscope. Those immobile juveniles<br />

were taken as dead when they could not revive within 12 h after<br />

being transferred to fresh WA and subsequently to tap water. Each<br />

treatment was replicated three times, and the experiment was run<br />

three times.<br />

Sequence generation and phylogenetic analysis<br />

Genomic DNA of actinobacteria was extracted using a bacterial<br />

genomic DNA extraction kit (BioTeke Corporation, China,<br />

Cat#:DP2001) and their 16S rRNA genes were amplified by PCR<br />

using the universal primer combination 27f (5 AGA GTT TGA TCC<br />

TGG CTC AG 3 ) and 1492r (5 GGT TAC CTT GTT ACG ACT T 3 ).<br />

The amplified products were purified using the Agarose gel DNA


2318 Afr. J. Microbiol. Res.<br />

Figure 1. Location of the 20 sampling sites (∗) in this study distributed on the map of China, which including two<br />

municipalities (BJ and SH), six autonomous regions (GX, NM, NX, XJ and XZ), and twelve provinces<br />

(Abbreviation meanings on the map, BJ: Beijing, GS: Gansu, GD: Guangdong, GX: Guangxi, GZ: Guizhou, HN:<br />

Hainan, HeB: Hebai, HeN: Henan, HLJ: Heilongjiang, HuN: Hunan, JL: Jilin, JS: Jiangsu, LN: Liaoning, NM:<br />

Neimenggu, NX: Ningxia, SD: Shandong, SX: Shanxi, SH: Shanghai, SC: Sichuan, XZ: Xizang).<br />

purification kit (TakaRa, code DV805A) and submitted to Beijing<br />

Genomics Institute for sequencing. The resulting sequences of 16S<br />

rRNA gene were compared with those available in the GenBank<br />

using the BLAST network service to determine their phylogenetic<br />

affiliation. Multiple alignments and sequence evolutionary distance<br />

calculations were carried out using CLUSTAL X version 2.0<br />

(Thompson et al., 1997). Phylogenetic analysis was performed<br />

using the MEGA software packages (Kumar et al., 2004), with gaps<br />

treated as missing data. Clustering was performed using the<br />

Neighbour-Joining method (Saitou and Nei, 1987). Bootstrap<br />

analysis was used to evaluate the tree topology of the Neighbour-<br />

Joining data by performing 1,000 resamplings (Felsenstein, 1985).<br />

The 16S rRNA gene sequences have been deposited in the<br />

GenBank database under accession numbers GQ357927-<br />

GQ357995.<br />

Data analysis<br />

Nematicidal activity (NA) was calculated using the formula: NA =<br />

IN/SN×100%, where IN represents the number of immobile<br />

nematodes and SN represents the sum of all nematodes counted<br />

(SN 100). Data were analyzed using the analysis of variance<br />

(ANOVA), and means were compared by the test of least significant<br />

difference (LSD) at P=0.05 using SPSS 11.0 for Windows (SPSS<br />

Inc., Chicago, USA).<br />

RESULTS<br />

Nematicidal activity of actinobacteria<br />

From each soil sample, 20 strains of actinobacteria were<br />

selected for nematicidal bioassay and these strains were<br />

selected mainly based on the differences of colony<br />

features, or randomly picked if colony features were<br />

similar, which resulting in a total of 4000 strains from 200<br />

samples for virulence test. Of the 4000 isolates tested,<br />

533 (13.2% of total) exhibited nematicidal activities with<br />

NA values more than 30% against the free-living<br />

nematode P. redivivus. These isolates showed


Isolate sum<br />

Isolate sum<br />

���<br />

� �<br />

���<br />

� �<br />

���<br />

§ ¤<br />

§¦�<br />

¡ ¤<br />

¡¦�<br />

�<br />

�<br />

¤<br />

�<br />

¡£¢<br />

17<br />

¡¦�<br />

�<br />

¤ ¥<br />

¨ �<br />

§ ¨<br />

Figure 2. Statistics of nematicidal actinobacteria from different sampling sites with different nematicidal<br />

activity (NA) to nematodes P. redivivus (A) and B. xylophilus (B).<br />

significant differences on their NAs against P. redivivus,<br />

which consisted of 55 isolates with NAs of 90-100%, 100<br />

with NAs of 80-90%, 127 with NAs of 60-80%, and 251<br />

with NAs less than 60% but more than 30% (Figure 2).<br />

Bioassay of the 4000 actinobacteria to plant-parasitic<br />

nematode B. xylophilus also was performed and 488<br />

isolates (12.2% of total) showed nematicidal virulence<br />

with NAs of 30-100%. The numbers of active isolates with<br />

NAs of 90-100%, 80-90%, 60-80% and 30-60% were 37,<br />

85, 111 and 255, respectively (Figure 2).<br />

Among the 4000 actinobacteria evaluated, the most<br />

active isolates showed the highest host selectivity to P.<br />

redivivus or B. xylophilus. Only 101 isolates (2.5% of<br />

total) were toxic to both targets with NA>30%. Among the<br />

101 strains, 4 (SDYM18, SDYM7, NMYC10 and NMCG3)<br />

showed the most potency to produce nematicidal<br />

metabolites with high NAs of more than 80% against both<br />

nematodes. Of the 155 actinobacteria strains with NAs ><br />

80% to P. redivivus, only 18 displayed similar NAs to both<br />

targets without statistically significant differences (P<br />

>0.05), while 54 isolates had no nematicidal activity<br />

against B. xylophilus. Similarly, of the 125 strains with<br />

¡¦¥<br />

¡ ¡<br />

¢<br />

© ¢<br />

§¦¡<br />

¡¦¨<br />

¤ ¥ ¤ � ¤ ¤<br />

§ ¨ § ¨<br />

¡ ¡<br />

§ §<br />

¡¦©<br />

©<br />

¡£¢<br />

¡£� ¡£¡<br />

¥ �<br />

¤<br />

© � ©<br />

� � ¨ ¨ § § ¨<br />

¡£¥<br />

¡¦�<br />

¢<br />

� �<br />

¡¦�<br />

¡£§<br />

§¦�<br />

�<br />

¤ � ¤ ¤ � ¨ ¨<br />

¡£¡<br />

� �<br />

¤ ¤ ¥ ¤ ¥<br />

� � ¨ § § §<br />

BJ GD GS GX GZ HeB HeN HLJ HN HuN JL JS LN NM NX SC SD SH SX XZ<br />

8<br />

A<br />

B<br />

13<br />

�<br />

9<br />

26<br />

15<br />

17<br />

6<br />

8<br />

9<br />

Soil origin<br />

25<br />

8<br />

13<br />

9<br />

14<br />

21<br />

���£¨¦�¦�£�¦¥¦�£� �<br />

���£¥¦�¦�£�¦©¦�£� �<br />

���£©¦�¦�£�¦¢¦�£� �<br />

���£¢¦�¦�£�¦¡¦�£�¦� �<br />

�<br />

� �<br />

�<br />

�<br />

� �<br />

� � � �<br />

� � � � � �<br />

� � � �<br />

� �<br />

� � � � � � � � � � � � � � � � � � � �<br />

� � � � � � � � �<br />

BJ GD GS GX GZ HeB HeN HLJ HN HuN JL JS LN NM NX SC SD SH SX XZ<br />

Soil origin<br />

� �<br />

� �<br />

�<br />

9<br />

��� �£�¦�£� � �£� �<br />

���¦� �¦�£� � �£� �<br />

���¦� �¦�£� � �£� �<br />

���¦� �¦� � �¦�£� �<br />

Kun et al. 2319<br />

NAs>80% to B. xylophilus, the NAs of 11 isolates to the<br />

two tested nematodes were not significantly different<br />

(P>0.05) while 41 isolates had no nematicidal activity<br />

against P. redivivus.<br />

The four potential strains (SDYM18, SDYM7, NMYC10<br />

and NMCG3) causing high mortality against both hosts<br />

were assayed further after diluting the culture suspension<br />

by 2, 5 and 10 folds. Our results indicated that the<br />

original culture broths of the four actinobacteria exhibited<br />

powerful virulence (85.2% NA 100%) to both P. redivivus<br />

and B. xylophilus, but the former was more susceptible<br />

than the latter (P


2320 Afr. J. Microbiol. Res.<br />

Table 1. Nematicidal activity (NA) of four potential actinobacteria.<br />

Strain Nematode<br />

SDYM18<br />

SDYM7<br />

NMYC10<br />

NMCG3<br />

Mean NA (%)±SD under different diluent<br />

Origin broth 2 fold 5 fold 10 fold<br />

P. redivius 100±0 a 93.2±1.22 80.8±0.83 64.2±0.85<br />

B. xylophilus 95.7±0.72 b 89.3±0.52 78.2±1.04 50.6±0.91<br />

P. redivius 99.4±1.31 a 72.6±0.87 57.7±0.88 44.3±1.21<br />

B. xylophilus 88.3±0.89 c 71.7±1.11 51.6±0.62 37.2±0.82<br />

P. redivius 99.2±1.11 a 80.5±0.85 68.7±0.93 34.3±1.12<br />

B. xylophilus 83.2±1.14 d 79.8±1.21 50.8±1.08 34.8±0.94<br />

P. redivius 99.6±1.42 a 80.3±1.13 67.5±0.89 50.3±1.34<br />

B. xylophilus 85.2±0.26 c 75.6±0.92 56.8±1.22 41.3±0.82<br />

Means in the “Origin broth” column followed by the same letter do not differ significantly at p 0.05, according to multiple<br />

comparisons.<br />

amplified using primer pair of 27f and 1492r, and<br />

submitted for sequencing. The resulting sequences,<br />

about 1380 nucleotides, were compared with those<br />

available in GenBank using the BLAST network service<br />

(Table 2), and a phylogenetic tree was constructed to<br />

determine their approximate taxonomic affiliation (Figure<br />

3). Phylogenetic analysis placed the 69 strains in three<br />

families (Streptomycetaceae, Pseudonocardiaceae and<br />

Nocardiaceae) of the Actinobacteria with sequence<br />

similarity of 97-100% (Figure 3 and Table 2).<br />

The largest group was Streptomycetaceae and it consisted<br />

of 58 isolates (84.1% of the total) showing 97 to<br />

100% sequence identity to 27 species of the genera<br />

Streptomyces and 1 species of Kitasatospora. Among the<br />

Streptomyces strains, the isolates phylogenetically<br />

related to species of S. althioticus (8 isolates), S.<br />

flavotricini (7) and S. diastatochromogenes (6)<br />

dominated, with a total of 21 isolates (36.8% of the<br />

Streptomyces group). There were 13 isolates (accounting<br />

to 22.8% of Streptomyces members) respectively<br />

affiliated to S. xanthophaeus (4 isolates), S. lateritius (3),<br />

S. exfoliatus (3) and S. ciscaucasicus (3). Six isolates<br />

respectively identified as S. showdoensi (2 isolates), S.<br />

corchorusii (2) and S. viridochromogenes (2). The remaining<br />

17 isolates were respectively identified belonging<br />

to 17 different species of the genus Streptomyces.<br />

The Pseudonocardiaceae group contained 7 isolates<br />

(10.1% of the total) respectively showing 98.5-99.7%<br />

sequence identity to species of Amycolatopsis lurida (4<br />

isolates) and A. niigatensis (3). The last group,<br />

Nocardiaceae, only included 4 isolates (5.8% of the total)<br />

with 99-99.6% identity to a species N. fluminea.<br />

Of the four isolates showed strong nematicidal activity,<br />

NMYC10 and NMCG3 respectively showed 98.9 and<br />

99.3% sequence identity to S. diastatochromogenes and<br />

S. violaceochromogenes, while SDYM18 and SDYM7<br />

were assigned to the same species A. lurida with the<br />

homology more than 99%.<br />

DISCUSSION<br />

In this study, 4000 actinobacteria isolated from agricultural<br />

soils of a wide range of geographical regions were<br />

evaluated for their nematocidial activities. The proportions<br />

of strains capable of killing P. redivivus and B. xylophilus<br />

were 13.3 and 12.2% respectively when taking NAs 30%<br />

as a cutoff for virulence activity. These results indicated<br />

that there were a large number of nematocidial<br />

actinobacteria distributed in agricultural soils. Of the 69<br />

active actinobacteria submitted for phylogenetic analysis,<br />

57 (accounted to 82.6%) showed 97 to 100% homology<br />

to 27 species of the genus Streptomyces. Ruanpanun et<br />

al. (2010) isolated 83 actinobacteria from plant-parasitic<br />

nematode infested soils and found the predominant<br />

actinobacteria taxa was Streptomyces (97.6%). Similarly,<br />

Luo et al. (2006) reported that in the 20 actinobacteria<br />

isolated from eggs and females of root-knot nematodes,<br />

16 strains were members of Streptomyces. Most<br />

actinobacteria commonly isolated from soil belonged to<br />

the genus Streptomyces because they are more common<br />

in the environment and tend to have rapid growth rate<br />

and good sporulation compared to other actinobacteria<br />

(Williams and Vickers, 1988). This may explain why the<br />

nematicidal Streptomyces spp. had high occurrence<br />

frequencies. Beside the biocontrol application of S.<br />

avermitilis (Jayakumar, 2009; Wright et al., 1983), the<br />

producer of avermectins, many other species of<br />

Streptomyces also had the potential as biocontrol agents<br />

against parasitic nematodes. CR-43 T , a strain of S.<br />

costaricanus isolated from a nematode-suppressive soil,<br />

exhibited both antinematodal and antifungal activities in<br />

laboratory, greenhouse, and field trials (Dicklow et al.,<br />

1993). An application for a patent for CR-43 T as a


Kun et al. 2321<br />

Table 2. Nematicidal actinobacteria used for phylogenetic analysis in this study and their closest affiliation according to the partical 16S<br />

rRNA gene.<br />

Strain [Accession No.] Sample location The closest relative in the database [Accession No.] Similarity (%)<br />

SDYM9 [GQ357978] Shandong S. lateritius LMG 19372 T [AJ781326] 99.8<br />

BJH10 [GQ357963] Beijing S. lateritius LMG 19372 T [AJ781326] 99.9<br />

SCTS20 [GQ357973] Sichuan S. lateritius LMG 19372 T [AJ781326] 99.9<br />

HNHS7 [GQ357938] Henan S. exfoliatus NBRC 13191 T [AB184324] 99.8<br />

SXYC14 [GQ357967] Shanxi S. exfoliatus NBRC 13191 T [AB184324] 99.6<br />

HNYM7 [GQ357937] Henan S. exfoliatus NBRC 13191 T [AB184324] 99.9<br />

NXFQ8 [GQ357970] Ningxia S. griseoplanus AS 4.1868 T [AY999894] 99.9<br />

NMXM7 [GQ357930] Neimenggu S. kurssanovii NBRC 13192 T [AB184325] 99.9<br />

BJS2 [GQ357965] Beijing S. mauvecolor LMG 20100 T [AJ781358] 99.1<br />

GSPG8 [GQ357950] Gansu S. xanthophaeus NBRC 12829 T [AB184177] 99.7<br />

SCYT16 [GQ357972] Sichuan S. xanthophaeus NBRC 12829 T [AB184177] 99.7<br />

SCLZ7 [GQ357981] Sichuan S. xanthophaeus NBRC 12829 T [AB184177] 99.8<br />

SCL7 [GQ357982] Sichuan S. xanthophaeus NBRC 12829 T [AB184177] 99.9<br />

NXHG4 [GQ357980] Ningxia S. flavotricini NBRC 12770 T [AB184132] 99.9<br />

GSPT14 [GQ357949] Gansu S. flavotricini NBRC 12770 T [AB184132] 99.9<br />

HNDD1 [GQ357943] Henan S. flavotricini NBRC 12770 T [AB184132] 99.9<br />

NXHG3 [GQ357983] Ningxia S. flavotricini NBRC 12770 T [AB184132] 99.8<br />

HNGL19 [GQ357939] Henan S. flavotricini NBRC 12770 T [AB184132] 99.3<br />

NXLJ11 [GQ357979] Ningxia S. flavotricini NBRC 12770 T [AB184132] 99.9<br />

NXFQ2 [GQ357928] Ningxia S. flavotricini NBRC 12770 T [AB184132] 99.9<br />

GSSZ10 [GQ357948] Gansu S. showdoensis NBRC 13417 T [AB184389] 98.2<br />

GSSZ14 [GQ357953] Gansu S. showdoensis NBRC 13417 T [AB184389] 98.2<br />

BJL6 [GQ357961] Beijing S. ciscaucasicus NBRC 12872 T [AB184208] 99.8<br />

GZ22 [GQ357951] Guizhou S. ciscaucasicus NBRC 12872 T [AB184208] 99.7<br />

XZYM3 [GQ357971] Xizang S. ciscaucasicus NBRC 12872 T [AB184208] 99.9<br />

SHJ24 [GQ357969] Shanghai S. longwoodensis LMG 20096 T [AJ781356] 99.3<br />

HaNXJ7 [GQ357947] Hainan S. corchorusii NBRC 13032 T [AB184267] 97.9<br />

HaNYM16 [GQ357944] Hainan S. corchorusii NBRC 13032 T [AB184267] 97.4<br />

CZYS9 [GQ357960] Xizang S. mirabilis NBRC 13450 T [AB184412] 100<br />

SHDQ4 [GQ357977] Shanghai S. cinereoruber subsp. fructofermentans<br />

NBRC 15396 T [AB184647]<br />

BJH1 [GQ357966] Beijing S. phaeochromogenes NBRC 3180 T [AB184738] 99.9<br />

SHLJ2 [GQ357976] Shanghai S. diastatochromogenes ATCC 12309 T [D63867] 98.6<br />

BJS8 [GQ357964] Beijing S. diastatochromogenes ATCC 12309 T [D63867] 99.4<br />

SHLJ5 [GQ357975] Shanghai S. diastatochromogenes ATCC 12309 T [D63867] 99.1<br />

GXLY20 [GQ357952] Guangxi S. diastatochromogenes ATCC 12309 T [D63867] 98.7<br />

NMYC10 [GQ357929] Neimenggu S. diastatochromogenes ATCC 12309 T [D63867] 98.9<br />

SXTD7 [GQ357968] Shanxi S. diastatochromogenes ATCC 12309 T [D63867] 99.3<br />

NMCG3 [GQ357931] Neimenggu S. violaceochromogenes NBRC 13100 T [AB184312] 99.3<br />

JLLJ17 [GQ357927] Jilin S. flavoviridis NBRC 12772 T [AB184842] 99.9<br />

DLHG2 [GQ357958] Liaoning S. viridodiastaticus NBRC 13106 T [AB184317] 99.8<br />

DLDG2 [GQ357959] Liaoning S. longispororuber NBRC 13488 T [AB184440] 99.3<br />

GSL5 [GQ357956] Gansu S. coeruleofuscus NBRC 12757T [AB184840] 99.6<br />

BJL2 [GQ357962] Gansu S. alboflavus NBRC 3438T [AB184775] 99.9<br />

HaHD12 [GQ357942] Hainan S. coelicoflavus NBRC 15399T [AB184650] 99.9<br />

DLTS7 [GQ357957] Liaoning S. viridochromogenes NBRC 3113T [AB184728] 99.6<br />

JLIM5 [GQ357940] Jilin S. viridochromogenes NBRC 3113T [AB184728] 99.5<br />

HaNYM8 [GQ357945] Hainan S. malaysiensis ATB-11T [AF117304] 99.9<br />

98.8


2322 Afr. J. Microbiol. Res.<br />

Table 2. Contd.<br />

HaNXJ11 [GQ357946] Hainan S. glauciniger NBRC 100913 T [AB249964] 99.4<br />

JLGC14 [GQ357941] Jilin S. variabilis NBRC 12825 T [AB184884] 99.9<br />

JLYM14 [GQ357933] Jilin S. althioticus NRRL B-3981 T [AY999791] 98.3<br />

JLFQ8 [GQ357936] Jilin S. althioticus NRRL B-3981 T [AY999791] 97.5<br />

GSPG5 [GQ357954] Gansu S. althioticus NRRL B-3981 T [AY999791] 97.6<br />

JLYM15 [GQ357932] Jilin S. althioticus NRRL B-3981 T [AY999791] 97.8<br />

JLFQ12 [GQ357935] Jilin S. althioticus NRRL B-3981 T [AY999791] 97.6<br />

GSPG4 [GQ357955] Gansu S. althioticus NRRL B-3981 T [AY999791] 97.6<br />

JLYM2 [GQ357934] Jilin S. althioticus NRRL B-3981 T [AY999791] 97.8<br />

SHN26 [GQ357974] Shanghai S. althioticus NRRL B-3981 T [AY999791] 97.4<br />

BJY6 [GQ357985] Beijing N. fluminea S1 T [AF277204] 99<br />

SXYM10 [GQ357986] Shanxi N. fluminea S1 T [AF277204] 99.4<br />

SXS5 [GQ357987] Shanxi N. fluminea S1 T [AF277204] 99.6<br />

NMBC12 [GQ357988] Neimenggu N. fluminea S1 T [AF277204] 99.3<br />

DLYT2 [GQ357984] Liaoning K. arboriphila HKI 0189 T [AY442267] 99.7<br />

HaNHS4 [GQ357989] Hainan A. niigatensis LC11 T [AB248537] 98.5<br />

HaNHS8 [GQ357992] Hainan A. niigatensis LC11 T [AB248537] 98.5<br />

HaNHS12 [GQ357991] Hainan A. niigatensis LC11 T [AB248537] 99.1<br />

NXQZ2 [GQ357995] Ningxia A. lurida DSM 43134 T [AJ577997] 99.3<br />

SDYM18 [GQ357994] Shandong A. lurida DSM 43134 T [AJ577997] 99.4<br />

GSPG7 [GQ357993] Gansu A. lurida DSM 43134 T [AJ577997] 99.7<br />

SDYM7 [GQ357990] Shandong A. lurida DSM 43134 T [AJ577997] 99.7<br />

A: Amycolatopsis, K: Kitasatospora, N: Nocardia, S: Streptomyces.<br />

biocontrol agent for nematodes was filed by Research<br />

Corporation Technologies, Tucson, Ark, under exclusive<br />

license from the University of Massachusetts at Amherst<br />

(Esnard et al., 1995). Several strains of Streptomyces<br />

with antifungal activities also significantly reduced rootlesion<br />

nematode population densities in roots of both<br />

susceptible and resistant alfalfa varieties grown in field<br />

(Samac and Kinkel, 2004). Several species of<br />

Streptomyces, e.g. S. dicklowii (US Patent 5549889), S.<br />

cyaneogriseus (US Patent 5030650), have been patented<br />

as nematicidal biopesticides (http://www.patentstorm.us).<br />

Many other actinomycete genera also produced biologically<br />

active secondary metabolites of medical importance<br />

and showed promising biological activity including<br />

parasitism and antibiosis (Goodfellow and O’Donnell,<br />

1989). Rickards et al. (1998) reported that a strain belonged<br />

to the genera Amycolatopsis or Amycolata could<br />

produce the cyclic decapeptide antibiotic quinaldopeptin<br />

with nematicidal activity. A strain of Streptoverticillium<br />

albireticuli was found to show strong nematicidal activity<br />

against Caenorhabditis elegans and several fungal<br />

pathogens. This species exhibited hyphal growth on both<br />

external and internal surfaces of C. elegans and<br />

ultimately killed the nematode (Park et al., 2002). Of the 4<br />

isolates with strong nematicidal activity in this study,<br />

NMYC10 and NMCG3 showed 98.9 and 99.3% homology<br />

to S. diastatochromogenes and S. violaceochromogenes<br />

respectively, while the other two (SDYM18 and SDYM7)<br />

were assigned to A. lurida. To isolate a wider spectrum of<br />

actinobacteria, pretreating soils such as dry-heating<br />

(Nonomura and Ohara, 1969), using Streptomyces’<br />

specific lytic actinophage (Kurtböke et al., 1992), and<br />

applying selective media supplemented with antibiotics<br />

(Williams et al., 1993) could all be employed.<br />

In this study, we used a free-living nematode and a<br />

plant parasitic nematode as target for bioassay. Among<br />

the 533 isolates and 488 isolates respectively showed<br />

nematicidal activity to P. redivivus and B. xylophilus, only<br />

101 isolates showed nematicidal properties to both target<br />

hosts. The host selectivity or sensitivity between<br />

actinobacteria and nematodes suggested that the<br />

nematicidal pathways or microbial actions were diverse.<br />

To determine the control targets of a nematicidal<br />

biopesticide, the host specificity should be taken into<br />

account. Additionally, the nematicidal actinobacteria in<br />

this study were screened based on the bioassay using<br />

culture broth. The metabolite composition of the four<br />

potential strains and their control effects in fields warrant<br />

further studies.<br />

ACKNOWLEDGEMENTS<br />

This work was funded jointly by “National Key Sciences<br />

and Technology Program for Water Solutions”<br />

(2009ZX07102-004-04-01; 2012ZX07102-003), NSFC


Kun et al. 2323<br />

Figure 3. Phylogenetic dendrogram obtained by neighbour-joining method of 16S rRNA gene sequences. Numbers at nodes<br />

are bootstrap values based on 1000 resamplings. Bar, 1% sequence divergence. Bifidobacterium psychraerophilum T16 T<br />

(AY174108) and Bifidobacterium mongoliense DSM 21395 T (AB433856) are used as outgroups. GenBank Accession<br />

Numbers for each strain are indicated in parentheses adjacent to each strain name.


2324 Afr. J. Microbiol. Res.<br />

(30970100), ZhengZhou Tobacco Research Institute<br />

(122009CZ0420), Yunnan Corporation of CNTC<br />

(2011YN03), National Development and Reform<br />

Commission, and Department of Science and Technology<br />

of Yunnan Province.<br />

REFERENCES<br />

Burg RW, Miller BM, Baker EE, Birnbaum J, Currie SA, Hartman R,<br />

Kong YL, Richard L, Monaghan RL, Olsonm G, Putter I, Tunac JB,<br />

Wwllick H, Stapley EO, Oiwa R, Omura S (1979). Avermectins, new<br />

family of potent anthelmintic agents: producing organism and<br />

fermentation. Antimicrob. Agents Ch., 15: 361-367.<br />

Chen ZX, Dickson DW (1998). Review of Pasteuria penetrans: biology,<br />

ecology, and biological control potential. J. Nematol.,30: 313-340.<br />

Dicklow MB, Acosta N, Zuckerman BM (1993). A novel Streptomyces<br />

species for controlling plant-parasitic nematodes. J. Chem. Ecol., 19:<br />

159-173.<br />

Dong JY, Zhao ZX, Cai L, Liu SQ, Zhang HR, Duan M, Zhang KQ<br />

(2004). Nematicidal effect of freshwater fungal cultures against the<br />

pine-wood nematode Bursaphelenchus xylophilus. Fungal Divers.,<br />

15: 125-135.<br />

Esnard J, Potter TL, Zuckerman BM (1995). Streptomyces costaricanus<br />

sp. nov., isolated from nematode-suppressive soil. Int. J. Syst.<br />

Bacteriol., 45(4): 775- 779.<br />

Felsenstein J (1985). Confidence limits on phylogenies: an approach<br />

using the bootstrap. Evolution, 39: 783-789.<br />

Goodfellow M, O’Donnell AG (1989). Search and discovery of<br />

industrially significant actinomycetes. In: Baumberg S, Hunter IS,<br />

Rhodes PM. (eds), Microbial Products: New Approaches. Cambridge<br />

University Press, Cambridge, UK, pp. 343-383.<br />

Gray N (1984). Ecology of nematophagous fungi: comparison of the soil<br />

sprinkling method with the Baermann funnel technique in the isolation<br />

of endoparasites. Soil Biol. Biochem., 16: 81-83.<br />

Hay FS, Skipp RA (1993). Fungi and actinomycetes associated with<br />

cysts of Heterodera trifolii Goffart (Nematoda: Tylenchida) in pasture<br />

soils in New Zealand. Nematologica, 39: 376-384.<br />

Huang XW, ZhaoNH, Zhang KQ (2004). Extracellular enzymes serving<br />

as virulence factors in nematophagous fungi involved in infection of<br />

the host. Res. Microb., 155(10): 811-816.<br />

Javed N, El-Hassan S, Gowen S, Pemproke B, Inam-ul-Hap M (2008).<br />

The potential of combining Pasteuria penetrans and neem<br />

(Azadirachta indica) formulation as a management system for rootknot<br />

nematodes on tomato. Eur. J. Plant Pathol., 120: 53-60.<br />

Jayakumar J (2009). Streptomyces avermitilis as a biopesticide for the<br />

management of root knot nematode, Meloidogyne incognita in<br />

tomato. Karn. J. Agric. Sci., 22: 564-566.<br />

Kurtböke DI, Murphy NE, Sivasithamparam K (1992). Use of<br />

bacteriophage for the selective isolation of thermophilic<br />

actinomycetes from composted eucalyptus bark. Can. J. Microbiol.,<br />

39: 46-51.<br />

Kumar S, Tamura K, Nei M (2004). MEGA3: Integrated software for<br />

molecular evolutionary genetics analysis and sequence alignment.<br />

Brief. Bioinformatics, 5: 150-163.<br />

Luo HL, Sun MH, Xie JP, Liu ZH, Huang Y (2006). Diversity of<br />

actinomycetes associated with root-knot nematode and their potential<br />

for nematode control. Acta Mirobiol. Sinica, 46(4): 598-601.<br />

Meadows J, Gill SS, Bone LW (1989). Factors influencing lethality of<br />

Bacillus thuringiensis kurstaki toxin for eggs and larvae of<br />

Trichostrongylus colubriformis (nematode), J. Parasitol., 75: 191-194.<br />

Mishra SK, Keller JE, Miller JR, Heisey RM, Nair MG, Putnam AR<br />

(1987). Insecticidal and nematicidal properties of microbial<br />

metabolites. Indian J. Microbiol., 2: 267-276.<br />

Nonomura H, Ohara Y (1969). Distribution of actinomycetes in soil (VI).<br />

A culture method effective for both preferential isolation and<br />

enumeration of Microbispora and Streptosporangium strains in soil<br />

(part 1). J. Ferm. Tech., 47: 463-469.<br />

Nour SM, Lawrence JR, Zhu H, Swerhone GDW, Welsh M, Welacky<br />

TW, Topp E (2003). Bacteria associated with cysts of the soybean<br />

cyst nematode (Heterodera glycines). Appl. Environ. Microbiol., 69:<br />

607-615.<br />

Oka Y, Shuker S, Tkachi N (2009). Nematicidal efficacy of MCW-2, a<br />

new nematicide of the fluoroalkenyl group, against the root-knot<br />

nematode Myloidogyne javanica. Pest Manag. Sci., 65: 1082-1089.<br />

Park JO, El-Tarabily KA, Ghisalberti EL, Sivasithamparam L (2002).<br />

Pathogenesis of Streptoverticillium albireticuli on C. elegans and its<br />

antagonism to soil-borne fungal pathogens. Lett. Appl. Microbiol., 35:<br />

361-365.<br />

Rickards RW, Rothschild JM, Lacey E (1998). Structure of<br />

actinotetraose hexatiglate, a unique glucotetraose from an<br />

actinomycete bacterium. J. Antibiot., 52(12): 1093-1098.<br />

Ruanpanun P, Tangchitsomkid N, Hyde KD, Lumyong S (2010).<br />

Actinomycetes and fungi isolated from plant-parasitic nematode<br />

infested soils: screening of the effective biocontrol potential, indole-3acetic<br />

acid and siderophore production. World J. Microbiol. Biotech.,<br />

0959-3993 (Print) 1573-0972 (Online).<br />

Saitou N, Nei M (1987). The neighbor-joining method: a new method for<br />

reconstructing phylogenetic trees. Mol. Biol. Evol., 4: 406-425.<br />

Samac DA, Kinkel LL (2004). Suppression of the root-lesion nematode<br />

(Pratylenchus penetrans) in alfalfa (Medicago sativa) by<br />

Streptomyces spp. Plant Soil, 235: 35-44.<br />

Schneider SM, Rosskopf EN, Leesch JG, Chellemi DO, Bull CT,<br />

Mazzola M (2003). Research on alternatives to methyl bromide: preplant<br />

and post-harvest. Pest Manag. Sci., 59: 814-826.<br />

Sun MH, Li G, Shi YX, Li BJ, Liu XZ (2006). Fungi and actinomycetes<br />

associated with Meloidogyne spp. eggs and females in China and<br />

their biocontrol potential. J. Inv. Pathol., 93: 22-28.<br />

Tikhonov VE, Lopez-Llorca LV, Salinas J, Jansson HB (2002).<br />

Purification and characterization of chitinases from the<br />

nematophagous fungi Verticillium chlamydosporium and V.<br />

suchlasporium. Fungal Gen. Biol., 35: 67-78.<br />

Thompson JD, Gibson TJ, Plewniak F, Jeanmougin F, Higgens DG<br />

(1997). The Clustal X windows interface: flexible strategies for<br />

multiple sequence alignment aided by quality analysis tools. Nucleic<br />

Acids Res., 24: 4876-4882.<br />

Walker J, Barrett J (1991). Cystathionine b-synthase and gcystathionase<br />

in helminths. Parasitol. Res., 77: 709-713.<br />

Wei J, Hale K, Carta L, Platzer E, Wong C, Fang S, Aroian V (2003).<br />

Bacillus thuringiensis crystal proteins that target nematodes. PNAS,<br />

100: 2760-2765.<br />

Williams ST, Vickers JC (1988). Detection of actinomycetes in the<br />

natural environment. In: Okami Y, Beppu T, Ogawara H (eds.),<br />

Biology of Actinomycetes. Japan Scientific Societies Press, Tokyo,<br />

pp. 265-270.<br />

Wright DJ, Birtle AJ, Corps AE, Dybas R (1983). Efficacy of avermectins<br />

against a plant parasitic nematode. Ann. Appl. Biol., 103: 465-470.


African Journal of Microbiology Research Vol. 5(16) pp. 2325-2328, 18 August 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.638<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

A new recommended disinfectant for dental<br />

instruments<br />

Jamileh bigom Taheri 1 , Mahin Bakhshi 1 , Sedigheh Bakhtiari 1 *, Bahare Nazemi 2 , Fateme<br />

Fallah 3 , Sahand Rezaie 5 , Hamed Mortazavi 1 and Somayyeh Azimi 4<br />

1 Department of Oral Medicine, Faculty of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.<br />

2 Department of Pediatric Dentistry, Faculty of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.<br />

3 Department of Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.<br />

4 Department of Oral Medicine, Faculty of Dentistry, Qazvin University of Medical Sciences, Qazvin, Iran.<br />

5 General Practitioner, Faculty of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.<br />

Accepted 30 July, 2011<br />

Enduro hand sanitizer is a new disinfecting agent that has recently been introduced as an effective<br />

antibacterial, antifungal and antiviral agent. Considering the importance of infection control in dentistry,<br />

this study aimed to evaluate the antimicrobial effects of the foam and its ability to disinfect dental<br />

surfaces and instruments. An experimental study was designed to evaluate the microbial load of 50<br />

samples which were obtained randomly from dental surfaces and instruments before and after<br />

application of the disinfecting foam. All samples were cultured in blood agar and nutrient agar culture<br />

media and incubated at 37°C for 24 h. Colony count was subsequently performed after an additional 24<br />

h. Furthermore, to evaluate the efficacy of the foam on different bacterial species, a blank disc was<br />

coated with foam and approximated with the bacteria. The clear zone around each disc was measured<br />

and reported (mm) after 48 h. The significance of differences between the data retrieved before and<br />

after applying the foam was determined using the non parametric Chi-square test. None of the species<br />

were colonized after the application of the foam. It was also shown to significantly reduce the<br />

colonization of resistant bacterial strains and the standard microbial species (P


2326 Afr. J. Microbiol. Res.<br />

species/strains. Its antimicrobial effect remains for two<br />

hours after spraying the agent. Moreover, due to its nonalcoholic<br />

nature, the subcutaneous adipose tissue is<br />

preserved which makes Enduro hand sanitizer foam<br />

suitable for daily hand washing usage without causing<br />

excessive dryness and skin irritability. Longevity and<br />

biocompatibility of this potent antimicrobial agent, makes<br />

it superior to other disinfectants (Enduro hand sanitizer<br />

wwwoiltechnics.co.uk/Animalhygiene/Sanitizers/index).<br />

Hence, this study was performed to assess the efficacy of<br />

this product in disinfecting dental instruments.<br />

MATERIALS AND METHODS<br />

This study was a joint project conducted in the department of oral<br />

medicine and Microbiology, Department of Shahid Beheshti<br />

University of Medical Sciences, Tehran, Iran in 2009-2010. The disinfecting<br />

foam (Enduro Hand Sanitizer) was a product of Biotechnic<br />

Company in England. Fifty samples were obtained from moist<br />

dental instruments (surgical forceps, elevator and periodontal<br />

scaling instruments) using a swab before and after the application<br />

of the foam and were placed in BHI broth. The BHI was subsequently<br />

incubated at 37°C for 24 h. The samples were cultures on<br />

nutrient blood agar (simple agar) and the plates were re-incubated<br />

for an additional 24 h. Positive plates marked bacterial growth. In<br />

order to confirm the presence of bacteria and identify the bacterial<br />

strains in contaminated samples, lamellas were obtained from the<br />

positive plates and subjected to Gram staining after drying and<br />

fixing. For this purpose, each lamella was thoroughly covered with<br />

crystal violet dye for 1 min. and then rinsed out. Each specimen<br />

was then covered with lugol solution immediately for 1 min, washed<br />

out and immersed into alcohol for 20 to 30 s. Fuchsine was<br />

ultimately poured on the lamellas and washed after 1 min. The<br />

specimens were stored in a dry place and observed under ×100<br />

magnification to verify the presence and type of bacterial strains.<br />

In order to evaluate the effect of the foam on MRSA, Bacillus<br />

Subtilis and Entrobacteriacea, these bacteria were cultured on<br />

Muller-Hinton agar. A blank disk was coated with foam and three<br />

antibiogram discs comprising of Penicillin, Ciprofloxacin and<br />

Tetracycline were subsequently placed next to each other on the<br />

agar plate. The plates were incubated at 37°C for 24 h and the clear<br />

zone around the blank discs on the culture plates were measured<br />

and recorded. Clear zones with a diameter of 10 mm or more would<br />

indicate the effectiveness of the foam. Furthermore colony count<br />

was performed to determine CFU with standard experimental<br />

strains. The following bacteria with standard ATCC were used as<br />

control species to verify the growth of contaminating samples in<br />

agar plates:<br />

1. Bacillus ATCC 6633.<br />

2. Staph Aureus 2923.<br />

3. Entrococcus 13047.<br />

4. Staph Epidermis ATCC 12228.<br />

5. Candida ATCC 2091.<br />

The significance of differences between the data retrieved before<br />

and after applying the foam was determined using the non<br />

parametric Chi-square test (P


Table 1. Colony count of standard bacterial species before administration of<br />

Enduro hand sanitiser foam.<br />

Bacterial species Number of positive samples<br />

Stap.h epidermidis 36<br />

Pseudomonas 1<br />

Streptococcus 5<br />

Bacillus 15<br />

Entrococcus 5<br />

Lactobacillus 3<br />

Table 2. Colony count of standard bacterial species after administration of Enduro<br />

hand sanitiser foam.<br />

Bacterial species Number of positive samples<br />

Staph. epidermidis 5<br />

pseudomonas 0<br />

streptococcus 0<br />

Bacillus 1<br />

Entrococcus 0<br />

Lactobacillus 0<br />

anti microbial effects on different bacterial, viral and<br />

fungal species and effectively diminishes microbial contamination<br />

on dental surfaces (Enduro hand sanitizer). The<br />

results of the present study demonstrated that Enduro<br />

hand Sanitizer can effectively eliminate all pathogen<br />

microorganisms from dental instruments including surgical<br />

forceps and periodontal scalers. The importance of<br />

this data is that the tested pathogens are among the<br />

extremely contagious species which if disinfected effectively<br />

between every patient, would significantly prevent<br />

the spread of diseases and infection. Furthermore, the<br />

diameter of clear zone around the blank disk immersed in<br />

the foam was reported 15 mm for Bacillus, 12 mm for<br />

MRSA and 8 mm for Entroccocus. This indicates the<br />

sensitivity of common oral pathogens such as S.<br />

Epidermidis, Bacillus subtitles and candida species to the<br />

new product and further confirms its efficacy in<br />

eliminating the microbial load in dental environments.<br />

However, further evaluations are required to assess the<br />

efficiency of the foam in clinical settings and on different<br />

bacterial and viral species.<br />

The present study failed to evaluate the antiviral effect<br />

of this product however, according to the information<br />

provided by the manufacturers, the foam seems to render<br />

satisfactory results against Avian Flue H5N1 infection<br />

according to UK DEFRA standard. It has further been<br />

claimed that the foam can destroy spores of Aspergillus<br />

niger and Clostridum difficile (EN 1650 and EN 13704<br />

standards respectively). On the other hand, considering<br />

its strong anti bacterial effect against gram positive as<br />

well as gram negative resistant strains of bacteria, the<br />

antiviral effect of the foam may also be justified.<br />

Taheri et al. 2327<br />

Furthermore, due to the fact that HIV and HBV are highly<br />

sensitive microorganisms compared to vegetative<br />

bacterial pathogens, the veridical effect of this new agent<br />

may be conceived (Enduro hand sanitizer). Nonetheless,<br />

it appears that the antiviral and antifungal effect of the<br />

foam should be further studied.<br />

This study demonstrated strong bactericidal effects of<br />

the product however; the important point is the sensitivity<br />

of specific species such as Bacillus Subtilis to Enduro<br />

Hand Sanitizer. This bacterial strain is capable of<br />

producing spores and long lasting infections if not treated<br />

effectively. According to in vitro studies, the efficacy of<br />

disinfecting agents may be altered by the substances in<br />

culture media and thus need to be reported with cautious<br />

(Sen et al., 2009). The basic shortcoming of this study is<br />

therefore hidden within its design that is, investigating the<br />

efficacy of the new foam in controlled laboratory<br />

conditions on specific isolated microorganisms. Hence, it<br />

appears that future robust clinical trials should be<br />

designed to further study the antimicrobial effects of the<br />

Enduro hand sanitizer foam in accurate clinical settings.<br />

It is also important to note that despite the acceptable<br />

results retrieved from different disinfecting agents<br />

irrespective of the form (foam or solution), all of them are<br />

solely capable of reducing the microbial load on the outer<br />

surfaces of the instruments; the inner chamber of the air<br />

motor and the inner wall of the air and water sprays<br />

which are major sources of infection transmission, are not<br />

thoroughly cleaned. Therefore, researchers recommend<br />

the combination of disinfection, lubrication and heat<br />

sterilization of the air motors and hand pieces between<br />

every patient as an ideal measure to minimize the risk of


2328 Afr. J. Microbiol. Res.<br />

cross contamination. In other words, researchers believe<br />

that despite the satisfactory results retrieved from<br />

different disinfecting solutions and foams, these products<br />

are solely used to reduce the microbial load prior to heat<br />

sterilization. Furthermore with constant change in the<br />

bacterial flora and the emergence of new highly resistant<br />

species of bacterial pathogens in health care environments,<br />

there is always a great concern regarding the<br />

resistance of microorganisms to the available antibiotics<br />

and disinfectants. Therefore, there is still room for further<br />

studies on new products and benefitting from successful<br />

experiences of leading companies.<br />

Conclusion<br />

The present study provided an overall evaluation of the<br />

Enduro hand sanitizer foam. Considering the fact that the<br />

manufacturer has provided a list of sensitive microorganisms<br />

to this product, studies should be designed to<br />

assess bacterial and viral pathogens independently.<br />

Once the efficacy of the foam is firmly documented, it can<br />

be widely used in dental environments.<br />

ACKNOWLEDGMENTS<br />

This article was based on a undergraduate thesis by Dr<br />

Rezaie, which was successfully completed under the<br />

supervision of Dr Taheri with the close cooperation of the<br />

Oral Medicine department of dental school of Shahid<br />

Beheshti University of Medical School.<br />

REFERENCES<br />

Bonten MJ (2002). Infection in the intensive care unit: Preventive<br />

stratrgies. Curr. Opin. Infect. Dis., 15: 401-405.<br />

CDC Guide line for infection in dental health care setting (2003).<br />

December, 14; 1-16. Available at http://www.cdc.gov/oral health<br />

/infection control /guidelines/index.htm<br />

Enduro hand sanitizer. Available at:<br />

http//wwwoiltechnics.co.uk/Animalhygiene/Sanitizers/index.html<br />

Oosthuysen J, Potgieter E, Blignaut E (2010). Compliance with infection<br />

control recommendations in South African dental practices: a review<br />

of studies published between 1990 and 2007. Int. Dent. J., 60: 181-<br />

189.<br />

Rauter S, Sigge A, Wiedeck H, Trantmann M (2002). Analysis of<br />

transmission pathway of pseudomonas aeroginos between patient<br />

and tap water outlets. Crit. Care Med., 30: 2222-2228.<br />

Sen B, Akdeniz BG, Denizci AA (2009). The effect of ethylenediaminetetraacetic<br />

acid on Candida albicans .Oral Surg. Oral Med. Oral<br />

Pathol. Oral Radiol. Endod., 90: 651-655.<br />

Silverman SJR (1987). Infection disease control and the dental office:<br />

Aids and other transmissible disease. Int. Dent. J., 37(2):108-113.<br />

Smith A, Creanor S, Hurrell D, Bagg J, McCowan M (2009).<br />

Management of infection control in dental practice. J. Hosp. Infect.,<br />

71: 353-358.<br />

Yüzbasioglu E, Saraç D, CanbazS, Saraç S, Cengiz S (2009). A survey<br />

of cross-infection control procedures: knowledge and attitudes of<br />

Turkish dentists. J. Appl. Oral Sci., 17: 565-569.<br />

Zanetti F, Vannini S, Bergamaschi A, Baldi E, Stampi S (2004).<br />

Infection control in dental health care setting:results of a survey on<br />

current disinfection practices. Ig Sanita Publ., 60: 229-242.


African Journal of Microbiology Research Vol. 5(16), pp. 2329-2337, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.642<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

The functional roles of the residue tyrosine at position<br />

26 in staphylococcal enterotoxin C2<br />

Hongbo Wang 1,2 , Junyi Zhou 3 , Mingkai Xu 1 *, Huiwen Zhang 1 and Chenggang Zhang 1<br />

1 Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China.<br />

2 Graduate School of Chinese Academy of Science, Beijing 100039, China.<br />

3 Shenyang Pharmaceutical University, Shenyang 110016, China.<br />

Accepted 30 July, 2011<br />

Bacterial superantigen Staphylococcus aureus enterotoxin C2 (SEC2) is a very potent activator of T<br />

cells. Previous crystal studies on SEC2 showed that residue Tyr26 which is located near the T cell<br />

receptor (TCR) binding sites could potentially participate in T cell activating. In SEC1, a superantigen<br />

highly homologous to SEC2, the residue Val26 played a critical role in stimulating certain V expansion.<br />

Moreover, residues 20, 22 and 26 in SEC1 determined its serologic cross-reaction. In order to<br />

investigate the potential roles of Tyr26 in SEC2, two SEC2 mutants SEC2(Y26V) and SEC2(Y26A) were<br />

constructed by site-directed mutagenesis. The activities and toxicities of the two mutants were<br />

determined both in vivo and in vitro. Results showed that both SEC2(Y26V) and SEC2(Y26A) remained<br />

potent immune stimulating activities compared with native SEC2. SEC2(Y26V) had a decreased<br />

pyrogenic effect on rabbit model. Both of the two mutants had significantly decreased binding affinities<br />

to anti-SEC2 IgG. Our result indicated that Tyr26 is a critical site for the serological characters of SEC2<br />

rather than for its superantigen activity. This may also provide an insight in constructing a novel mutant<br />

to substitute native SEC2 in cancer immunotherapy avoiding being neutralized by anti-SEC2 IgG in<br />

vivo.<br />

Key words: Staphylococcal enterotoxin C2, Site-directed mutagenesis, superantigen, serological, property.<br />

INTRODUCTION<br />

Microbial superantigens are a family of enterotoxins that<br />

share the ability to trigger excessive and aberrant<br />

activation of T cells (Llewelyn and Cohen, 2002). Unlike<br />

conventional antigens, SAgs bind to invariant regions of<br />

MHC class molecules outside the antigen-binding<br />

groove and are presented as unprocessed proteins to T<br />

lymphocytes expressing appropriate motifs on the<br />

variable domain of the chain(V ) of the T cell receptor<br />

(TCR) (Müller-Alouf et al., 2001). As a consequence,<br />

SAgs can activate an unusually high percentage of T<br />

lymphocytes and initiate massive releasing of cytokines<br />

which plays a pivotal role in T cell-mediated immune<br />

responses. The best characterized SAgs are<br />

Staphylococcal enterotoxins (SEs) and Streptococcal<br />

*Corresponding author. E-mail: mkxu@iae.ac.cn. Tel: +86 24<br />

8397 0380. Fax: +86 24 8397 0381.<br />

pyrogenic exotoxins (Spes) to date. Staphylococcal<br />

enterotoxins (SEs), a series of serological types of heat<br />

stable enterotoxins, produced by S. aureus, are the<br />

leading causes of gastroenteritis resulting from consumption<br />

of contaminated food (Balaban and Rasooly,<br />

2000). These toxins share common phylogenetic relationship,<br />

structure, function, and sequence homology. SECs<br />

are a group of highly conserved proteins with significant<br />

immunological cross-reactivity (Marr et al., 1993).<br />

According to minor epitope differences, SECs were<br />

further classified into three subtypes (C1-C3), which differ<br />

in only several amino acid residues near the N terminus<br />

(Hovde et al., 1990).<br />

Staphylococcal enterotoxin C2 (SEC2) can effectively<br />

activate the immune system and result in tremendous<br />

releasing of cytokines such as tumor necrosis factors-<br />

(TNF- ), interleukin-2 (IL-2), and interferon- (INF- ).<br />

This attribute makes SEC2 an effective ingredient for<br />

cancer immunotherapy in clinic in China (Chen, 2007).


2330 Afr. J. Microbiol. Res.<br />

But as an enterotoxin produced by S. aureus, SEC2 can<br />

cause toxic shock syndrome (TSS) at certain concentration,<br />

which limits its clinical application. Moreover, anti-<br />

SEC2 antibodies in patients’ serum could neutralize<br />

injected SEC2 and impair its effects. In Sweden, a mutant<br />

SEA has been fused to the Fab-fragment of tumortargeted<br />

monoclonal antibody for treatment of solid<br />

tumors in phase II trial. But toxicity and anti-SEA antibody<br />

were also the limiting factors for therapy (Shaw et al.,<br />

2007). For construction of novel superantigen variants<br />

with decreased toxicity and low reactivity to human anti-<br />

SEC2 antibodies, it is necessary to give insight into the<br />

key residues in SEC2.<br />

In SEC1, valine in position 26 (Val26) was likely to<br />

interact with TCR directly and cause certain V expansion<br />

(Deringer et al., 1996). Previous researches on<br />

antigenic uniqueness indicated that residues 20, 22 and<br />

26 were critical for forming SECl- and SEC2-specific<br />

epitopes (Turner et al., 1992). While in SEC2, the<br />

crystallographical data showed that tyrosine in position<br />

26 (Tyr26) might locate in putative TCR binding site<br />

(Schad et al., 1997; Fields et al., 1996), and it was likely<br />

to expose on the molecular surface and influence<br />

molecular interaction between SEC2 and TCR (Turner et<br />

al., 1992; Papageorgiou et al., 1995).<br />

SEC2 and SEC1 are different in only seven residues<br />

near the N terminus (Bohach and Schlievert, 1989), while<br />

their superantigen activities exhibited some differences<br />

(Hovde et al., 1994; Wang et al., 2009a). Based on the<br />

high homology between SEC1 and SEC2, we<br />

hypothesize that the residue of Tyr26 might be important<br />

to the superantigen activities and serological characters<br />

of SEC2.<br />

In order to investigate the potential roles of Tyr26 in<br />

SEC2, in this study, two SEC2 mutants SEC2(Y26V) and<br />

SEC2(Y26A) have been constructed and their<br />

superantigen activities and toxicities were determined<br />

both in vivo and in vitro. Our results showed that the<br />

Tyr26 was not function-related with the immune stimulating<br />

activities of SEC2, but important for the serological<br />

characters of SEC2. The mutant SEC2(Y26V) with potent<br />

immune stimulating activities showed decreased toxicity<br />

and significantly reduced binding by SEC2-specific IgG,<br />

which suggested that the newly constructed mutant<br />

protein could be used as a potentially powerful candidate<br />

for cancer immunotherapy in the future.<br />

MATERIALS AND METHODS<br />

Plasmids, bacterial strains and cell line<br />

Expression vector pET28a(+) from Novagen (Darmstadt, Germany)<br />

was used to transform SEC2 mutant gene into the host. E. coli<br />

BL21(DE3) from Novagen was cultured in Luria–Bertani (LB)<br />

medium for expressing SEC2 mutant protein. Murine hepatoma<br />

Hepal-6 cell line used in this study was purchased from American<br />

Type Culture Collection (ATCC) and maintained in RPMI 1640<br />

medium with 10% (v/v) fetal bovine serum (Gibco, USA).<br />

Recombined plasmid pET-28a-SEC2 which contains the entire<br />

SEC2 cDNA (Accession number AY450554) was constructed and<br />

conserved in our lab (Xu and Zhang, 2006).<br />

Chemicals and enzymes<br />

All chemicals are analytical grade. Pfu DNA polymerase and<br />

restriction enzymes were purchased from Takara Biotechnology Co<br />

(Dalian, China); isopropyl-D-thiogalactopyranoside (IPTG) and<br />

methylthiazol tetrazolium (MTT) were from Sigma Chemical Co (St<br />

Louis, MO, USA); Ni-NTA His-Bind resin was from Qiagen<br />

(Germany); the DNA Gel Extract kit and Mini-preparation of plasmid<br />

kit were from BioDev-Tech Co (Beijing, China).<br />

Experimental animals<br />

Female BALB/c mice (6-8 weeks old, 20-25 g) and adult New<br />

Zealand white rabbits weighing 2.0-2.5 kg were purchased from the<br />

Experimental Animal Center, China Medical University (Shenyang,<br />

China).<br />

All experimental animals were maintained under specificpathogen-free<br />

conditions on a 12 h light/dark cycle. All experiments<br />

were carried out under the guiding principles for the care and use of<br />

laboratory animals approved by Animal Care Committee of China<br />

Medical University.<br />

Site-directed mutagenesis<br />

We constructed two mutant SEC2, SEC2(Y26V) and SEC2(Y26A),<br />

by converting the Tyr at position 26 to Val and Ala respectively. The<br />

conceptual ideas behind this substitution design were as follows:<br />

we introduce Val into position 26 of SEC2 because valine is the<br />

residue in position 26 in SEC1, furthermore we introduce Ala into<br />

position 26 of SEC2 to diminish the effect of Tyr in this position.<br />

The SEC2 mutant genes were constructed by sequence overlap<br />

extension. All the primers related with over-lap PCR were listed in<br />

Table 1. The PCR-generated fragments containing substituted<br />

oligonucleotides were digested by EcoR I and Xho I, and ligated<br />

into plasmid pET28a(+) digested with the same enzymes. The<br />

recombined expression plasmid was transformed into E. coli<br />

BL21(DE3) and positive clone was identified by DNA sequencing.<br />

Expression and purification of mutant protein<br />

For expressing SEC2 mutant protein, the positive clone of<br />

transformed E. coli BL21(DE3) was incubated in LB for 10 h. The<br />

culture was then inoculated at a ratio of 1:100 (v/v) into LB medium<br />

supplemented with 50 g kanamycin ml -1 and incubated at 37°C<br />

with vigorous shaking until the OD600 reached 0.7. Then 1.0mM<br />

IPTG was added into the culture and the incubation was continued<br />

for 4 h more with vigorous shaking at 30°C. The cells were<br />

harvested by centrifugation at 5,000 rpm and resuspended with icecold<br />

loading buffer (50 mM NaH2PO4, 500 mM NaCl, 10 mM<br />

imidazole, pH 7.9). The cells were disrupted by sonication on ice<br />

followed by centrifugation (10,000 rpm, 30 min). The supernatants<br />

were collected and loaded onto a Ni-saturated chelating Sepharose<br />

column pre-equilibrated with loading buffer. After removing nonspecifically<br />

binding protein with washing bufferl (50 mM NaH2PO4,<br />

500 mM NaCl, 40 mM imidazole, pH 7.9), SEC2 mutant protein was<br />

released from the column by washing buffer (50 mM NaH2PO4, 500<br />

mM NaCl, 250 mM imidazole, pH 7.9) and dialysed against PBS<br />

(137 mM NaCl, 2.7 mM KCl, 4.3 mM Na2HPO4, 1.4 mM KH2PO4,<br />

pH 7.4) at 4°C for 48 h to remove the high concentration of


Table 1. Primers for amino acid substitution by over-lap PCR.<br />

Primers Amino acid substitution Primer sequence a<br />

P-1 5 -CGGAATTCGAGAGTCAACCAGA-3<br />

P-2 26Y V 5 -CATCATATAATACTTTCATATTACCCATCG-3<br />

P-3 5 -GGGTAATATGAAAGTATTATATGATGATC-3<br />

P-4 26Y A 5 -CATCATATAATGCTTTCGTATTACCCATCG-3<br />

P-5 5 -GGGTAATACGAAAGCATTATATGATGATC-3<br />

P-6 5 -TCGCTCGAGTTATCCATTCTTTGTTG-3<br />

a The primers were designed for special amino acid substitution. The sequences of mutant residues are stressed in<br />

boldface. The recognition sites of EcoR I and Xhol I are underlined.<br />

imidazole. The purity of SEC2 mutant protein was estimated by<br />

SDS-PAGE and the concentration of mutant protein was<br />

determined by Bradford assay using BSA as the standard.<br />

Stimulating murine T-cell proliferation assay<br />

To investigate the activity of stimulating T cell proliferation by<br />

mutant SEC2, splenocytes from 6- to 8-week-old BALB/c female<br />

mice were collected as experimental target. The mutant SEC2 and<br />

the native SEC2 dissolved in 1640 medium were filled into the wells<br />

of 96-well microtitre plates in four different concentrations (10, 100,<br />

1000, and 10,000 ng/ml). There were three repeated wells at each<br />

concentration. RPMI 1640 medium with 10% FBS was served as<br />

negative control. Murine T cells were seeded into the experimental<br />

and negative control wells at a density of 8×10 5 cells per well.<br />

The proliferation index (PI) was determined by MTT assay. The<br />

splenocytes were incubated at 37°C with 5% CO2 for 72 h. The<br />

incubation was continued for 4 h more after 25 l MTT (5 mg/ml,<br />

dissolved in PBS) had been added to each well. Then the T cells<br />

were collected by centrifugation at 1,000 rpm for 10 min. 120 l<br />

DMSO was added into each well to redissolve the pellet on the<br />

bottom. To fully dissolve the formazan produced by T cells, the<br />

plate was vigorously shaking for 20 min. The absorbance was<br />

measured with a microplate reader at 570 nm, using a reference<br />

wavelength of 630 nm. The final absorbance is the difference<br />

between these two readings. The proliferation index (PI) was<br />

calculated with the following equation:<br />

PI=Abs value of the experimental well / Abs value of the negative<br />

control well.<br />

Antitumor activity<br />

To further investigate the superantigen activity of mutant SEC2, the<br />

inhibitory effects on tumor cell was determined by MTT assay.<br />

Splenocytes from 6- to 8-week-old BALB/c female mice were<br />

served as effector cells and Hepal-6 cell as target. Native SEC2<br />

and the mutants diluted with RMPI 1640 containing 10% FBS were<br />

separately filled into the wells of 96-well microtitre plate at 10ng/ml,<br />

100 and 1,000 ng/ml in triplicate. In experimental wells, effector<br />

cells (5×10 5 cells/well) and target cells (2.5×10 4 cells/well) were<br />

mixed in the present of native SEC2 or the mutants in different<br />

concentrations. Wells contained protein and lymphocytes only were<br />

served as lymphocytes releasing control. Wells contained Hepal-6<br />

cells alone were served as unsettled tumor cell control. The blank<br />

wells were RMPI 1640 with 10% FBS only, and BSA was served as<br />

negative control.<br />

After incubation for 72 h at 37°C with 5% CO2, 25 l MTT was<br />

added into each well, and the cells were incubated for 4 h more.<br />

Wang et al. 2331<br />

The cells were collected by centrifugation and redissolve with<br />

DMSO. The absorbance was measured with a microplate reader at<br />

570 nm, using a reference wavelength of 630 nm. Tumor growth<br />

inhibition (%) was calculated with the equation: 100 - [(Abs value in<br />

protein-treated cells well - Abs value in lymphocytes-releasing well)<br />

/ (Abs value in unsettled tumor cells control wells - Abs value in<br />

blank control wells)] ×100.<br />

Rabbit model assay<br />

Rabbit model was used to compare the febrile response of SEC2<br />

and the mutants in vivo. Each of the experimental animals was<br />

fixed and the rectal temperature was measured for at least 90 min<br />

before injection. Qualified rabbits used in this assay must have<br />

stable body temperatures ranging from 38.6 to 39.5°C. Native<br />

SEC2 and the mutants dissolved in PBS were injected into rabbits<br />

at the does of 10 g/kg (body weight). Three animals were injected<br />

with each protein. PBS was served as negative. Rectal temperatures<br />

of rabbits were measured with indwelling rectal thermometers<br />

right after pyrogen administration continually for 3 h. The rectal<br />

temperature change ( T) of rabbit was accepted as an index of<br />

pyrogen effect in vivo, and it was calculated by subtracting the<br />

temperature immediately before injection from the temperature at<br />

each time point after the pyrogen injection.<br />

Proteolysis susceptibility assays<br />

To investigate whether the trypsin-resistance of SEC2 mutants<br />

were affected by amino acid substitution, purified mutanted proteins<br />

and native SEC2 (100 g/ml) were respectively incubated at 37°C<br />

with trypsin at a trypsin/protein ratio of 1:25 (w/w). After desired<br />

periods of time, the enzymolysis was terminated by boiling in SDS-<br />

PAGE sample buffer for 5 min and analyzed by SDS-PAGE.<br />

Detection of mutant SEC2 by ELISA<br />

ELISA assay was carried out in accordance with the instruction of<br />

the kit (National institute for the control of pharmaceutical and<br />

biological products, Beijing, China). Briefly, the polystyrene plates<br />

were coated with 100 l of 5 g/ml rabbit anti-SEC2 IgG diluted in<br />

carbonate buffer overnight. The excessive IgG which did not bind to<br />

the wells was washed off by PBS containing 0.05% Tween 20<br />

(PBS-T20), then unabsorbed sites were blocked with 1% gelatin in<br />

PBS for 30 min at 37°C. Mutant SEC2 and the native dissolved in<br />

PBS-T20 buffer were added into the wells separately. The plate<br />

was incubated for 2 h at 37°C. Then the wells were emptied and<br />

washed by PBS-T20 buffer. Rabbit anti-SEC2 IgG-HRP conjugate<br />

was added, and the plate was incubated for 1.5 h at 3 7°C. The


2332 Afr. J. Microbiol. Res.<br />

Figure 1. Results of murine T-cell proliferation by SEC2 mutants and native SEC2.<br />

Purified proteins of SEC2 mutants or the native were incubated with murine splenocytes<br />

for 72 h before the proliferation effects were determined by MTT assay. Value on the yaxis<br />

represents the average proliferation index (PI)± SD of triplicate values. Each<br />

purified protein was tested in at least three separate assays.<br />

uncombined enzyme-linked antibody was washed off by PBS-T20,<br />

and 100 l substrate (0.04% ortho-phenylene-diamine) was added<br />

into the wells and incubated for 30 min at 37°C. Reactions were<br />

terminated by addition of 50 l 2 mol/L H2SO4, and the absorbance<br />

was measured at 450 nm.<br />

Statistical analysis<br />

Results are presented as the mean ± SD. Statistical analysis was<br />

performed using Student’s t test. A P-value < 0.05 was considered<br />

statistically significant.<br />

RESULTS<br />

Construction and expression of the mutant SEC2<br />

To construct SEC2 mutant protein, over-lap PCR was<br />

employed. The mutant SEC2 genes were successfully<br />

constructed and cloned into the expression vector pET-<br />

28a (+). The recombinant expression vectors were<br />

transformed into E. coli BL21 (DE3) respectively for<br />

expressing mutant proteins. The positive clones were<br />

identified by DNA sequencing.<br />

For expressing SEC2 mutant proteins, the positive<br />

clones with the right mutagenesis introduced were<br />

respectively cultured in LB medium containing 50 mg<br />

kanamycin ml -1 till the OD 600 reached 0.7. The<br />

expressions of mutant proteins were performed by IPTG<br />

inducing at 30°C for 4 h. After purification with Ni-NTA<br />

His Bind Resin, both of the two mutant proteins shown as<br />

a single band on SDS-PAGE were of purity of more than<br />

95%.<br />

Murine T-cell proliferation activity<br />

The activities of stimulating T cell proliferation by SEC2<br />

and the mutants were investigated. Series of ten-fold<br />

dilutions of native SEC2 and the mutants dissolved in<br />

PBS were filled into the wells of a 96-well microtitre plate<br />

with murine T cells. After 72 h incubation, the proliferation<br />

index of murine T cell was determined by MTT assay. As<br />

the experimental data reflected, both native SEC2 and<br />

the mutants could induce murine T cells proliferation in a<br />

dose-dependent manner. The ability to induce T cell<br />

proliferation of SEC2 mutants were similar with that of the<br />

native, without significant differences at any concentration<br />

(P>0.05; Figure 1). This result indicated that the<br />

amino acid substitution at position 26 with Ala or Val had<br />

not significantly affected the stimulating activity of SEC2.<br />

Examination the antitumor activity<br />

To further investigate the superantigen activity of mutant<br />

SEC2, MTT assay was employed to compare the


Figure 2. MTT assay was employed to compare the tumor cell growth inhibition effects between<br />

SEC2 and the mutants. Tumor cells and the splenocytes were mixed and seeded into the wells at<br />

the density of 2.5×10 4 cells/well and 5×10 5 cells/well respectively. Different concentrations of SEC2<br />

or the mutants were incubated with the cells at 37°C for 72 h before the absorbance was<br />

measured. BSA was used as negative control. Data shown are representative of at least three<br />

separate experiments, and the values represent the mean ± standard error of the mean.<br />

anti-tumor activity of SEC2 and the mutants. Hepal-6<br />

cells (target cells) and splenocytes (effector cells) were<br />

incubated in the present of proteins in different concentrations<br />

for 72 h before the absorbance was measured.<br />

All the proteins tested in this study exhibited predominant<br />

anti-tumor abilities compared with negative control<br />

(P0.05). This indicated that the amino<br />

acid substitution in position 26 had little influence on the<br />

superantigen activity of SEC2.<br />

Toxicity<br />

The Rabbit model assay was carried out to investigate<br />

the pyrogenic effect of mutant SEC2. The rectal<br />

temperature of each experimental animal was continually<br />

measured for 3 h after pyrogen injection. The results<br />

showed that both SEC2 and the mutants could induce the<br />

enhancement of temperature in experimental animal<br />

significantly compared with negative control (P0.05). Whereas SEC2(Y26V) exhibited a<br />

decreased ability to induce fever at 2h, 2.5h, and 3h<br />

compared with SEC2. Furthermore, the enhancement of<br />

temperature in experimental animal injected with<br />

SEC2(Y26V) reached its top at 1.5 h after pyrogen<br />

injection. But it is noteworthy that the body temperature of<br />

animal injected with SEC2 or SEC2(Y26A) rose<br />

continually although the experiment.<br />

Susceptibility to trypsin<br />

In order to determine whether the minor decreased<br />

pyrogenic effect of SEC2(Y26V) in the later stage of<br />

pyrogen experiment was resulted from its degradation,<br />

trypsin digestion assay was performed to evaluate the<br />

stability of the two mutants. The result was determined by<br />

15% SDS-PAGE and showed that there was no significant<br />

difference in the trypsin susceptibility between native<br />

SEC2 and the mutants (Figure 4). This result suggested<br />

that the amino acid substitution at position 26 did not<br />

affect the instability of SEC2, and the decreased<br />

pyrogenic activity of SEC2(Y26V) was not resulted from<br />

its degradation.


2334 Afr. J. Microbiol. Res.<br />

Figure 3. The pyrogenicities of SEC2 and the mutants in rabbit model. The body<br />

temperature of experimental rabbit was continually measured for 3h right after<br />

protein injection. PBS solution was served as negative control. The value of y-axis<br />

represents the rise of temperature.<br />

Figure 4. Susceptibilities of SEC2(Y26V), SEC2(Y26A), and<br />

native SEC2 to trypsin were compared. Each protein was<br />

mixed with trypsin at a trypsin/protein ratio of 1:25 (w/w). The<br />

digestive reaction was performed at 37°C for time gradient<br />

and terminated by boiling for 5 min at 100°C. The result was<br />

analyzed by 15% SDS-PAGE.


Figure 5. Comparison the anti-SEC2 IgG binding affinity of native SEC2 and the mutants<br />

through ELISA. The absorbance was measured and calculated as the mean readings of<br />

triplicate samples ± SD.<br />

Evaluating epitope change of mutant SEC2 by ELISA<br />

ELISA assay was employed to investigate whether the<br />

Tyr at position 26 determines the epitope of SEC2. Both<br />

of the two mutants showed significant decreased binding<br />

affinities to SEC2-specific IgG compared with native<br />

SEC2 (P


2336 Afr. J. Microbiol. Res.<br />

side chain of Try26 was completely removed by Ala<br />

substitution in SEC2(Y26A). A reasonable explanation for<br />

this phenomenon is that Try26 was not the key residue<br />

for the immune stimulating activity of SEC2.<br />

Previous crystal structure study on SEC2 conveyed<br />

that residues 20, 22 and 26 potentially interacted with<br />

TCR (Papageorgiou et al., 1995). Although there was no<br />

direct evidence that Tyr26 in SEC2 participated in the<br />

interaction of SEC2 with TCR, some researchers<br />

reported that Val26 in SEC1 recognized the specificity of<br />

TCR V of SEC1. Mutant study in SEC1 showed that<br />

SEC1(V26 Y)with a single amino acid substitution at<br />

position 26 could significantly enhance the expansion of<br />

V 13.1 but decreased the expansion of V 3 (Deringer et<br />

al., 1996).<br />

So, on the basis of the high homology between SEC1<br />

and SEC2, we constructed SEC2(Y26V) and<br />

SEC2(Y26A) to respectively change and diminish the<br />

TCR V recognization ability of SEC2. It was widely<br />

accepted that the affinity of SAgs for TCR can<br />

significantly affect their mitogenic potency (Andersen et<br />

al., 2001). The weak affinity of the TCR-Sag interaction<br />

results in poor T cell response. But in this study, both of<br />

the two mutants exhibited comparable stimulating ability<br />

with SEC2, which suggested that substitution in position<br />

26 could not alter the TCR affinity of SEC2.<br />

In rabbit model assay, SEC2(Y26V) exhibited a<br />

decreased ability to induce fever at 2h, 2.5h, and 3h<br />

compared with SEC2. This promoted us to investigate<br />

whether the decreased toxicity in vivo was due to its<br />

instability resulted from amino acid substitution. Our<br />

result from proteolysis susceptibility assays has exhibited<br />

that both of the two mutants maintained comparable<br />

resistance to trypsin with SEC2, which indicates that the<br />

amino acid substitution in position 26 did not result in the<br />

exposure of additional tryptic cleavage sites. So it is<br />

suggested that the decreased toxicity of SEC2(Y26V)<br />

does not due to its degradation. SEC2(Y26V) has potent<br />

immune stimulating activities compared with SEC2 in all<br />

concentration, which is inconsistent with the previous<br />

theory that the febrile activity of SE is a direct<br />

consequence of T cell stimulation. On the other hand,<br />

macrophages and monocytes stimulated by superantigen<br />

could secrete some endogenous pyretogenic cytokines<br />

such as TNF- and IL-1, which was the major cause of<br />

immune-mediated diseases such as fever (Roggiani et<br />

al., 1997). Further investigation was needed to interpret<br />

it.<br />

Previous researches on antigenic uniqueness indicated<br />

that residues 20, 22 and 26 were critical for forming<br />

SECl- and SEC2-specific epitopes. SEC1 mutants with<br />

substitutions at all three positions reacted only with an<br />

SEC2-specific antibody (Turner et al., 1992). On the<br />

basis of the homology between SEC1 and SEC2, we<br />

hypothesized that Tyr26 was important for the serological<br />

characters in SEC2. The result from ELISA confirmed this<br />

hypothesis that SEC2(Y26V) and SEC2(Y26A) showed<br />

significantly decreased binding affinities to SEC2-specific<br />

IgG. The residue of Val26 in SEC2(Y26V) was homologous<br />

to Val26 in native SEC1, so Val26 could mediate<br />

the cross-reactivity of SEC2(Y26V) with rabbit anti-SEC2<br />

polyclonal IgG used in this study. It could be a possible<br />

explanation for that SEC2(Y26V) showed higher binding<br />

affinity to SEC2-specific IgG than SEC2(Y26A) did. For<br />

the lack of SEC1-specific IgG, we could not determine<br />

whether SEC2(Y26V) and SEC2(Y26A) were able to<br />

induce cross-reactivity with SEC1-specific IgG. However<br />

it was quite sure that the mutation introduced into position<br />

26 influenced the recognition of SEC2 by SEC2-specific<br />

antibodies, which suggested that Tyr 26 was critical for<br />

the antigenic uniqueness of SEC2.<br />

Concluded from our result, the roles of residue at<br />

position 26 in SEC2 are different from those of in SEC1,<br />

which may result from continuous evolution. This slight<br />

difference can display different serological specificity to<br />

avoid the cross-reactivity with antibodies from the host.<br />

In summary, we have investigated the potential roles of<br />

Tyr26 in SEC2 by site-directed mutagenesis. Our results<br />

indicated that Tyr26 is not important for the immune<br />

stimulating activities, but a critical site for the serological<br />

characters of SEC2. A low toxic mutant SEC2(Y26V)<br />

constructed in this study with potent superantigen<br />

activities and significantly decreased binding affinities to<br />

anti-SEC2 IgG has the potentiality to be a novel mutant to<br />

substitute native SEC2 in cancer immunotherapy.<br />

ACKNOWLEDGEMENT<br />

This work was supported by a grant from the National<br />

Science and Technology Major Specific Projects of China<br />

for "Significant Creation of New Drugs" (2009ZX09103-<br />

692).<br />

REFERENCES<br />

Andersen PS, Geisler C, Buus S, Mariuzza RA, Karjalainen K (2001).<br />

Role of the T Cell Receptor Ligand Affinity in T Cell Activation by<br />

Bacterial Superantigens. J. Biol. Chem., 276: 33452-33457.<br />

Balaban N, Rasooly A (2000). Staphylococcal enterotoxins. Int. J. Food<br />

Microbiol., 61: 1-10.<br />

Bohach GA, Schlievert PM (1989). Conservation of the biologically<br />

active portions of staphylococcal enterotoxins Cl and C2. Infect.<br />

Immun., 57: 2249-2252.<br />

Chen TZ (2005). The basis and criteria for the effective dose of SEC2<br />

oral liquid. Prog. in Microbiol. Immunol. China, 33: 51-58.<br />

Chen TZ (2007). The future development direction of HAS. Prog. in<br />

Microbiol. Immunol. China, 35: 49-50.<br />

Deringer JR, Ely RJ, Stauffacher CV, Bohach GA (1996). Subtypespecific<br />

interactions of type C stapholoccal enterotoxins with the Tcell<br />

receptor. Mol. Microbiol., 22: 523-534.<br />

Fields BA, Malchiodi EL, Li H, Ysern X, StauVacher CV, Schlievert PM,<br />

Karjalainen K, Mariuzza RA (1996). Crystal structure of a T-cell<br />

receptor beta-chain complexed with a superantigen. Nature, 384:<br />

188-192.<br />

Hovde CJ, Hackett SP, Bohach GA (1990). Nucleotide sequence of the<br />

staphylococcal enterotoxin C3 gene: sequence comparison of all<br />

three type C staphylococcal enterotoxins. Mol. Gen. Genet., 220:<br />

329-333.<br />

Hovde CJ, Marr JC, Hoffmann ML, Hackett SP, Chi YI, Crum KK,<br />

Stevens DL, Stauffacher CV, Bohach GA (1994). Investigation


of the role of the disulphide bond in the activity and structure of<br />

staphylococcal enterotoxin C1. Mol. Microbiol., 13: 897-909.<br />

Llewelyn M, Cohen J (2002). Superantigens: microbial agents that<br />

corrupt immunity. Lancet infect. Dis., 2: 156-162.<br />

Marr JC, Lyon JD, Roberson JR, Luphe MR, Davis WC, Bohach GA<br />

(1993). Characterization of novel type C staphylococcal enterotoxins:<br />

biological and evolutionary implications. Infect. Immun., 61: 4254-<br />

4262.<br />

Müller-Alouf H, Carnoy C, Simonet M, Alouf JE (2001). Superantigen<br />

bacterial toxins: state of the art. Toxicon, 39: 1691-1701.<br />

Papageorgiou AC, Acharya KR, Shapiro R, Passalacqua EF, Brehm<br />

RD, Tranter HS (1995). Crystal structure of the superantigen<br />

enterotoxin C2 from Staphylococcus aureus reveals a zinc-binding<br />

site. Structure, 3:769-779.<br />

Pastacaldi C, Lewis P, Howarth P (2010). Staphylococci and<br />

staphylococcal superantigens in asthma and rhinitis: a systematic<br />

review and meta-analysis. Allergy, 66: 549-555.<br />

Roggiani M, Stoehr JA, Leonard BA, Schlievert PM (1997). Analysis of<br />

toxicity of streptococcal pyrogenic exotoxin A mutants. Infect.<br />

Immun., 65: 2868-2875.<br />

Schad EM, Papageorgiou AC, Svensson LA, Acharya KR (1997). A<br />

structural and functional comparison of staphylococcal enterotoxins A<br />

and C2 reveals remarkable similarity and dissimilarity. J. Mol. Biol.,<br />

269: 270-280.<br />

Shaw DM, Connolly NB, Patel PM, Kilany S, Hedlund G, Nordle O,<br />

Forsberg G, Zweit J, Stern PL, Hawkins RE (2007). A phase study of<br />

a 5T4 oncofoetal antigen tumor-targeted superantigen (ABR-214936)<br />

therapy in patients with advanced renal cell carcinoma. Br. J. Cancer,<br />

96: 567-574.<br />

Wang et al. 2337<br />

Strandberg KL, Rotschafer JH, Vetter SM, Buonpane RA, Kranz DM,<br />

Schlievert PM (2010). Staphylococcal superantigens cause lethal<br />

pulmonary disease in rabbits. J. Infect. Dis., 202: 1690-1697.<br />

Sundstedt A, Celander M, Ohman MW, Forsberg G, Hedlund G (2009).<br />

Immunotherapy with tumor-targeted superantigens (TTS) in<br />

combination with docetaxel results in synergistic anti-tumor effects.<br />

Int. Immunopharmacol., 9: 1063-1070.<br />

Turner TN, Smith CL, Bohach GA (1992). Residues 20, 22, and 26<br />

Determine the Subtype Specificities of Staphylococcal Enterotoxins<br />

Cl and C2. Infect. Immun., 60: 694-697.<br />

Wang XG, Xu MK, Cai YM, Yang HL, Zhang HW, Zhang CG (2009a).<br />

Functional analysis of the disulphide loop mutant of staphylococcal<br />

enterotoxin C2. Appl. Microbiol. Biotechnol., 82: 861-871.<br />

Wang XG, Xu MK, Zhang HW, Liu J, Li X, Zhang CG (2009b).<br />

Enhancement of superantigen activity and antitumor response of<br />

staphylococcal enterotoxin C2 by site-directed mutagenesis. Cancer<br />

Immunol. Immunother., 58: 677-686.<br />

Xu MK, Zhang CG (2006). Gene Expression and Function Study of<br />

Fusion Immunotoxin Anti-Her-2-scFv-SEC2 in E.coil. Appl. Microbiol.<br />

Biotechnol., 70: 78-84.


African Journal of Microbiology Research Vol. 5(16), pp. 2338-2348, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.136<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Prevalence study of cytomegalovirus (CMV) infection<br />

among foreign manpower in Jeddah Saudi Arabia<br />

N. A. Redwan 1 , M. M. M. Ahmed 1,2 * and M. S. H. AL Awfi 1<br />

1 Department of Biological Sciences, Faculty of Science, P. O. Box 80203, King Abdulaziz University, Jeddah, 21589,<br />

Saudi Arabia.<br />

2 Nucleic Acids Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI),<br />

Mubarak City for Scientific Research and Technology Applications, Alexandria, Egypt.<br />

Accepted 25 June, 2011<br />

Human cytomegalovirus (HCMV) is a species-specific DNA virus of the Herpetoviridae family.<br />

Cytomegalovirus (CMV) is more widespread in developing countries and in areas of low socioeconomic<br />

conditions. It causes high morbidity and mortality. After primary infection CMV is not<br />

eradicated but establishes life-long infection in its host. CMV dispersed and become dormant or latent<br />

in multiple end organs, and can later be reactivated by a number of different stimuli, including<br />

immunosuppresion and inflammation. To determine CMV prevalence in a sample of the foreign<br />

manpower population in Jeddah region, Saudi Arabia, we tested serum samples for CMV-specific<br />

immunoglobulin G from participants aged 20 to 60 years (n = 514) by enzyme linked immunosorbent<br />

assay (ELISA). The prevalence of CMV infection was 80.7% in studied population. CMV prevalence<br />

differed significantly by sex (p


through the epithelium of the upper alimentary,<br />

respiratory, or genitourinary tracts.<br />

However, the initial infection of epithelial cells is not<br />

essential, as demonstrated by infections from blood<br />

transfusions and organ transplantation. CMV is spread<br />

through the body with the aid of leukocytes and vascular<br />

endothelial cells (Halpern et al., 2004). Dissemination of<br />

CMV through the blood is typically followed by infection of<br />

the ductal epithelial cells. CMV spreads through the body<br />

quickly into many sites. Historical in-vitro studies<br />

characterize CMV as a slowly replicating virus, but in a<br />

recent study based on changes in viral load in humans,<br />

CMV has been shown to replicate quickly with a doubling<br />

time of approximately one day during active infection<br />

(Gerna et al., 2004). CMV infects a wide range of tissues<br />

and cell types: it has been found in salivary glands, lung,<br />

liver, pancreas, kidney, ear, eye, placenta, alimentary<br />

tract, heart, ovaries, pituitary, brain, skin, thyroid,<br />

esophagus, prostate, testes, and adrenals (Reddehase et<br />

al., 2002).<br />

In the vast majority of healthy individuals the immune<br />

system quickly reaches homeostasis with CMV. While the<br />

immune system of healthy individuals is usually able to<br />

prevent CMV from producing clinical manifestations, the<br />

immune system very rarely completely eliminates CMV<br />

from the body - the viral genome remains in a latent<br />

stage waiting for reactivation. While the latent stage of<br />

CMV infection has been researched extensively, the<br />

identity of cell types that harbor latent CMV, the ranges of<br />

genes expressed during latency, and the mechanisms of<br />

reactivation continue to be controversial (Jarvis and<br />

Nelson, 2002).<br />

A crucial part of the immune system defense against<br />

CMV is the development of CMV specific antibodies.<br />

Antibodies bind to CMV, thereby inhibiting its ability to<br />

infect new cells and marking it for removal from the body.<br />

The first type of antibody to develop in response to CMV<br />

is Immunoglobulin M (IgM), which develops within a few<br />

days following primary infection. While CMV IgM remains<br />

detectable for six to nine months, medium to high levels<br />

of CMV IgM can be detected during the first three months<br />

of a primary infection. IgM can also be detected during<br />

some secondary infections, both reactivation and<br />

reinfection, and is therefore not a valid marker of primary<br />

infection. The second antibody type to respond to CMV is<br />

IgG. This antibody develops within 1 to 2 weeks after<br />

infection and, once developed, can be detected throughout<br />

life. Consequently, IgG is commonly used and widely<br />

accepted measure of previous CMV infection (Drew,<br />

1988).<br />

CMV seropositivity rates are higher in females, older<br />

people, those of lower socioeconomic status (SES) and<br />

residents of developing countries. A recent study also<br />

shows a relationship between ethnicity and seroprevalence<br />

that is independent of factors such as SES.<br />

Worldwide, seroprevalence in adults in the general<br />

population varies from 40 to 90% (Dowd et al., 2009).<br />

Redwan et al. 2339<br />

Tests for antibodies in an individual's serum are called<br />

serologic tests. If antibodies are not detected in the<br />

serum, the individual is seronegative. For<br />

Immunoglobulin G (IgG), this is commonly interpreted to<br />

mean that the individual's immune system has never<br />

encountered the virus and they have never been infected<br />

with CMV. Individuals are called seropositive if antibody<br />

to the virus is detectable in their blood. This means that<br />

the individuals have been previously infected with CMV. If<br />

upon an individual's first CMV IgG test they are seronegative<br />

and on a later test are seropositive, the individual<br />

is said to have undergone seroconversion, indicating the<br />

occurrence of a primary infection.<br />

The aim of this study was a prevalence of infected<br />

CMV in worker population in Jeddah region, Saudi<br />

Arabia. In addition, find out the relationship of our result<br />

with age, gender and ethnicity.<br />

MATERIALS AND METHODS<br />

Serum samples collection<br />

A total of 514 serum samples were tested for CMV seroprevalence<br />

using enzyme linked immunosorbent assay (ELISA). Samples were<br />

collected from 4 diagnostic laboratories throughout Jeddah,<br />

including: the Medical Laboratory of Ghulail Dispensary, Medical<br />

Laboratory of Badr Adeen Dispensary, Medical laboratory of AL-<br />

Quds dispensary and Medical laboratory of University street<br />

hospital. These laboratories supplied remnant sera from samples<br />

that had been submitted for serological testing and would otherwise<br />

have been discarded. Sera from subjects who were known to be<br />

infected with human immunodeficiency virus, hepatitis B virus and<br />

hepatitis C virus were excluded. Sera were identified at the referring<br />

laboratory by the sex of the subject, age or date of birth, date of<br />

collection, and nationality. The samples were coded by date of<br />

collection, nationality/territory of origin, sample number and<br />

referring laboratory. All serum samples were stored at -20°C until<br />

use.<br />

Study population<br />

Serum samples were collected from foreign workers between 20<br />

and 60 years of age and stratified into the following age groups: 20<br />

to 24, 25 to 29, 30 to 34, 35 to 39, 40 to 44, 45 to 49, 50 to 54 and.<br />

55 to 60, Serum samples were not available for less than 20 year of<br />

age. Approximately equal numbers of males and females were<br />

tested. Also, approximately equal numbers of seven nationalities<br />

including: Egyptian, Yemeni, Sudan, Pakistani, Indian, Bangladesh<br />

and Ethiopian were tested. Prevalence was calculated separately<br />

for each age group, for each nationality, for male and female<br />

separately, and for foreign workers as a whole. The CMV prevalence<br />

for the group aged 61 years and above was assumed to be<br />

the same as for the group aged 55 to 60 years. Sample sizes were<br />

calculated to achieve a 95% confidence interval (CI) of<br />

approximately ±5% for each age group.<br />

Serological testing<br />

Serum samples were tested for HCMV-specific immunoglobulin G<br />

(IgG) using a HCMV IgG enzyme-linked immunosorbent assay<br />

(ELISA) technique using DRG kit (DRG International, Inc., USA).


2340 Afr. J. Microbiol. Res.<br />

Principle of the test<br />

The antigen composed of partially purified and inactivated<br />

cytomegalovirus is bound to the solid phase (8-well strips). The<br />

specific immunoglobulin is bound to the antigen through incubation<br />

with diluted human serum. After washing to eliminate the proteins<br />

which have not reacted, incubation is performed with the conjugate,<br />

composed of human IgG monoclonal antibodies conjugated to<br />

horse radish peroxidase. The unbound conjugate is eliminated, and<br />

the peroxidase substrate added. The blue color which develops is<br />

proportional to the concentration of specific antibodies present in<br />

the serum sample. When the enzymatic reaction is interrupted by<br />

the addition of sulphuric acid solution, the yellow color, which<br />

develops, can be easily read by using a microplate reader (Wisdom,<br />

1976).<br />

Reagent preparation<br />

All reagents should be allowed to reach room temperature (18 to<br />

25°C) before use. Dilute 1 volume of wash buffer with 19 volume of<br />

distilled water. For example, dilute 50 ml of wash buffer into 950 ml<br />

distilled water to prepare 1000 ml of wash buffer which is stable for<br />

1 month at 2 to 8°C mixed well before use.<br />

Sample and controls dilution<br />

Prepare 1:40 dilution of test samples, negative control, positive<br />

control and calibrator by adding 5 l of the samples and controls to<br />

195 l of sample diluents and mixed well. Procedure is as in assay<br />

(ELISA) technique using DRG kit.<br />

RESULTS<br />

Calculation of the result<br />

Qualitative results<br />

The CMV IgG Index of each determination was<br />

calculated by dividing the absorbance value of each<br />

sample by absorbance value of cut-off. Samples with<br />

CMV index less than 0.90 was sero-negative for IgG<br />

antibody to CMV, those with CMV index between 0.91<br />

and 0.99 is equivocal and the samples should be<br />

repeated.<br />

Quantitative results<br />

For quantitative determination of anti-CMV IgG levels of<br />

positive specimens in IU/ml, the O.D of cut-off and<br />

positive calibrators are plotted on the Y-axis of a group<br />

against their corresponding anti-CMV IgG concentration<br />

of 0, 1, 2, 6, 18 IU/ml on the X-axis. The estimates of<br />

levels in patient sera are read off the graph using their<br />

individual O.D. values.<br />

Interpretation of the results<br />

Qualitative results<br />

The results were interpreted according to the<br />

manufacturer’s instructions. Samples with CMV index<br />

less than 0.90 was seronegative for IgG antibody to<br />

CMV, those with CMV index between 0.91 and 0.99 is<br />

equivocal and the samples should be repeated. Samples<br />

with CMV index of 1.00 or greater was seropositive for<br />

IgG antibody to CMV.<br />

Quantitative result<br />

The results were interpreted according to the<br />

manufacturer’s instructions. when the anti-CMV IgG<br />

concentration in the sample is less than 0.8 IU/ml, the<br />

sample was no immune, those with anti-CMV IgG<br />

concentration is more than 1.2 IU/ml, the sample was<br />

immune. If the result is between the two values, in this<br />

case it is advisable to repeat the test in duplicate.<br />

Statistical analysis<br />

The percentages of individuals with positive, negative,<br />

and equivocal results were determined for each age<br />

group and sex. SPSS program version 15 was used for<br />

the analysis and comparison of sero-statuses among age<br />

groups, male and female and each nationality. Ninetyfive-percent<br />

confidence intervals were calculated where<br />

appropriate, and P values of < 0.05 were considered<br />

statistically significant.<br />

Demographic characteristics of the studied<br />

population<br />

Serum samples according to sex divided to two groups'<br />

males (51.4%) and female (48.6%) with their ages varied<br />

from 20-60 years old. Serum samples were stratified<br />

according to age into the following groups: 20 to 24<br />

(12.6%), 25 to 29 (12.6%), 30 to 34 (12.8%), 35 to 39<br />

(12.8%), 40 to 44 (12.4%), 45 to 49 (12.3%), 50 to 54<br />

(12.3%) and 55 to 60 (12.1%) year. Serum samples<br />

were not available for less than 18 years of age. Serum<br />

samples according to nationality were divided to the<br />

following groups: Egyptian (14.6%), Yemen (14.2%),<br />

Sudan (14.4%), Pakistan (14.6%), Indian (14%),<br />

Bangladesh (14.2%) and Ethiopian (14%). According to<br />

race/ethnicity serum samples were divided to African<br />

(43%) and Asian (57%). Approximately equal numbers of<br />

males and females were tested. Also approximately<br />

equal numbers of each nationality and ages groups were<br />

tested. Prevalence of CMV- IgG in each group were<br />

tested using enzyme linked immunosorbent assay and


Table 1. Demographic characteristics of the studied population.<br />

Characteristic<br />

Age(years)<br />

No. of samples<br />

No.<br />

Positives<br />

No.<br />

P value<br />

20-24 12.6 65 53.8 35


2342 Afr. J. Microbiol. Res.<br />

Sex<br />

Figure 1. Prevalence of cytomegalovirus by sex (Males and females).<br />

Table 3. Prevalence of cytomegalovirus by age groups from 20 to 60 years old.<br />

Age group No. of samples<br />

Positive<br />

No. %<br />

Compared to age group 20 - 24<br />

2<br />

(p) (95%CI)<br />

20-24 65 35 50.8 Referent 41.0-61.8<br />

25-29 65 43 66.2 2.051(>0.05) 61.5-72.1<br />

30-34 66 46 69.7 3.486(>0.05) 62.2-77.8<br />

35-39 66 58 87.9 18.418(


Figure 2. Prevalence of cytomegalovirus by age groups from 20-60 years old.<br />

Table 4. Prevalence of cytomegalovirus in females by age groups from 20 to 60 years old.<br />

Age group No. of samples<br />

Positive<br />

No. %<br />

Redwan et al. 2343<br />

Compared to age group 20 - 24<br />

2<br />

(p) (95%CI)<br />

20-24 32 21 65.6 Referent 60.1-69.9<br />

25-29 33 26 78.8 1.406(>0.05) 74.8-83.6<br />

30-34 32 29 90.6 5.851(


2344 Afr. J. Microbiol. Res.<br />

Table 5. Prevalence of cytomegalovirus in males by age groups from 20 to 60 years old.<br />

Age group No. of samples<br />

Positive Compared to age group 20 - 24<br />

No. %<br />

2<br />

(p) (95%CI)<br />

20-24 33 14 42.4 Referent 34.3-50.1<br />

25-29 32 17 53.1 0.746(>0.05) 47.5-60.5<br />

30-34 34 17 50 0.387(


Redwan et al. 2345<br />

Table 6. Prevalence of cytomegalovirus by Nationality: (Egyptian; Yemeni; Sudan; Pakistan; Indian Bangladesh and Ethiopian).<br />

Nationality No. of samples<br />

Positive Compared to age group 20 - 24<br />

No. %<br />

2<br />

(p) (95%CI)<br />

Indian 72 48 66.7 Referent Referent<br />

Egyptian 75 59 78.7 2.671(>0.05) 72.9-85.1<br />

Yemen 73 56 76.7 1.804(>0.05) 73.5-79.5<br />

Sudan 74 65 87.8 9.350(


2346 Afr. J. Microbiol. Res.<br />

Table 7. Prevalence of cytomegalovirus by ethnicity (African and Asian).<br />

Ethnicity No. of sample<br />

No.<br />

Positive 2 (p) (95%CI)<br />

African 221 188 85.1 4.671(


toddlers. Infected infants and children, in particular those<br />

under 30 months old, actively excrete the virus in their<br />

saliva and urine. Thus, one hypothesis to explain the<br />

higher females CMV seroprevalence would be that<br />

women have more contact with children and have more<br />

opportunities for HCMV infection during pregnancy,<br />

delivery, and menstruation.<br />

In the study reported herein, the seroprevalence of<br />

CMV IgG among the studied population varied with<br />

nationality and/or ethnicity ranging from 66.7% in Indian,<br />

78.7% in Egyptian, 76.7% in Yemeni, 87.8% in Sudan,<br />

82.7% in Pakistan, 83.6% in Bangladesh, to 88.9% in<br />

Ethiopian. The prevalence varied significantly with<br />

nationality when all nationalities compared with Indian<br />

nationality as reference (p< 0.05) except for the Egyptian<br />

and Yemeni nationality (p>0.05). In general the seroprevalence<br />

of cytomegalovirus among the African population<br />

(85.1%) varied significantly from Asian population<br />

(77.5%). The differences in the prevalence rate in<br />

different nationalities perhaps could be related to socioeconomic<br />

factors, environmental, and climatic factors.<br />

The result of the present study is consistent to the<br />

results of other researches carried out in different parts of<br />

world. Badami et al. (2009) reported that the ethnicity<br />

appeared to be related to seroprevalence of cytomegalovirus<br />

(CMV) in New Zealand between 2003 and 2006<br />

and it ranging from 93.2% in Pacific Islanders, 54.8% in<br />

Caucasians, 80.4% in Maori, 77.6% in Asian, to 71.4% in<br />

Maori/Caucasian (Badami et al., 2009). The result of<br />

present study is also consistent to the result of another<br />

study carried out by Staras et al. (2006) between the year<br />

1988 and 1994 to determine CMV prevalence in the US<br />

population. CMV seroprevalence differed by race and/or<br />

ethnicity as follows: 51.2% in non-Hispanic white<br />

persons, 75.8% in non-Hispanic black persons, and<br />

81.7% in Mexican Americans. Racial and/or ethnic<br />

differences in CMV seroprevalence persisted when<br />

controlling for household income level, education, marital<br />

status, area of residence, census region, family size,<br />

country of birth, and type of medical insurance (Staras et<br />

al., 2006). Also similar results to our results shown in a<br />

study carried out by Gaytant et al. (2005) to investigate<br />

the incidence of congenital cytomegalovirus infections in<br />

the Netherlands. A significant difference (P


2348 Afr. J. Microbiol. Res.<br />

REFERENCES<br />

Adjei AA, Armah HB, Gbagbo F, Boamah I, Adu-Gyamfi C, Asare I<br />

(2008). Seroprevalence of HHV-8, CMV, and EBV among the general<br />

population in Ghana, West Africa. BMC Infect. Dis., 8:111.<br />

Ahmed SA, Al-Joudi FS, Zaidah AW, Roshan TM, Rapiaah M, Abdullah<br />

YMS, Rosline H (2006). The prevalence of human cytomegalovirus<br />

seroposoitivity among blood donors at the unit of blood transfusion<br />

medicine, hospital University Sains Malaysia. The Southeast Asian J.<br />

Trop. Med. Public Health., 37: 294-296.<br />

Badami KG, Mcquilkan-Bickerstaffe S, Ells JE, Parata M (2009).<br />

Cytomegalovirus seroprevalence and ‘cytomegalovirus-safe’<br />

seropositive blood donors. Epidemiol. Infect., 137: 1776-1780.<br />

Dowd JB, Aiello AE, Alley DE (2009). Socioeconomic disparities in the<br />

seroprevalence of cytomegalovirus infection in the US population:<br />

NHANES III. Epidemiol. Infect., 137: 58–65.<br />

Drew WL (1988). Diagnosis of Cytomegalovirus Infection. Review of<br />

Infect. Dis., 10: S468-S476.<br />

Fowler KB, Stagno S, Pass RF (2003). Maternal Immunity and<br />

Prevention of Congenital Cytomegalovirus Infection. JAMA., 289:<br />

1008-1011.<br />

Gaytant MA, Galama JMD, Semmekrot BA, Melchers WJG, Sporken<br />

JMM, Oosterbaan HP, van Dop PA, Huisman A, Merkus HM,<br />

Steegers EA (2005). The Incidence of Congenital Cytomegalovirus<br />

Infections in The Netherlands. J. Med. Virol., 76: 71–75.<br />

Gerna G, Baldanti F, Revello MG (2004). Pathogenesis of Human<br />

Cytomegalovirus Infection and Cellular Targets, Hum. Immunol., 65:<br />

381-386.<br />

Halpern CT, Hallfors D, Bauer DJ, Iritani B, Waller MW, Cho H (2004).<br />

Implications of Racial and Gender Differences in Patterns of<br />

Adolescent Risk Behavior for HIV and Other Sexually Transmitted<br />

Diseases. Perspectives Sexual Rep. Health, 36: 239-347.<br />

Jarvis MA, Nelson JA (2002). Human cytomegalovirus persistence and<br />

latency in endothelial cells and macrophages, Curr. Opin. Microbiol.,<br />

5: 403–407.<br />

Joseph SA, Beliveau C, Muecke CJ, Rahme E, Soto JC, Flowerdew G,<br />

Johnston L, Langille D, Gyorkos TW (2005). Risk factors for<br />

cytomegalovirus seropositivity in a population of day care educators<br />

in Montreal, Canada. Occupational Med., 55: 564–567.<br />

Kafi S, Eldouma E, Saeed S, Musa H (2009). Seroprevalence of<br />

Cytomegalovirus among blood donors and antenatal women<br />

attending two hospitals in Khartoum State, Sudan J. Med. Sci., 4: 1-5.<br />

Kanapeckien V, Kalibatas J, Redaitien E, eremnych J (2007). The<br />

association between cytomegalovirus infection and aging process,<br />

Medicina (Kaunas), 43: 419-423.<br />

Likitnukul S, Bhatarakosol P, poovorawan Y (2003). Seroprevalence of<br />

cytomegalovirus in children born to HIV 1 infected women. Asian<br />

Pacific journal of allergy and immunology / launched by the Allergy<br />

and Immunology Society of Thailand., 21: 127-130.<br />

Lübeck PR, Doerr HW, Rabenau HF (2009). Epidemiology of Human<br />

Cytomegalovirus (HCMV) in an urban region of Germany: what has<br />

changed?. Med. Microbiol. Immunol., 199: 53-60.<br />

Mussi-Pinhata MM, Yamamoto AY, Figueiredo LT, Cervi MC, Duarte G<br />

(1998). Congenital and perinatal cytomegalovirus infection in infants<br />

born to mothers infected with human immunodeficiency virus, J.<br />

Pediatr., 132: 285–290.<br />

O'Brien TP, Thompson JM, Black PN, Becroft DM, Clark PM, Robinson<br />

E, Wild C, Mitchell EA (2009). Prevalence and determinants of<br />

cytomegalovirus infection in pre-school children, J. Pediatr. Child<br />

Health, 45: 291 – 296.<br />

Ocak S, Duran N, Eskocak AF (2006). Seroprevalence of<br />

Cytomegalovirus Antibodies in Haemodialysis Patients. Turk. J. Med.<br />

Sci., 36: 155-158.<br />

Paschale MD, Agrappi C, TeresaManco M, Paganini A, Clerici P (2009).<br />

Incidence and Risk of Cytomegalovirus Infection during Pregnancy in<br />

an Urban Area of Northern Italy. Infect. Dis. Obstetr. Gynecol., pp. 1-<br />

5.<br />

Reddehase MJ, Podlech J, Grzimek NKA (2002). Mouse models of<br />

cytomegalovirus latenc, J. Clin. Virol., 25: S23-S36.<br />

Seale H, MacIntyre CR, Gidding HF, Backhouse JL, Dwyer DE, Gilbert<br />

L (2006). National Serosurvey of Cytomegalovirus in Australia. Clin.<br />

Vaccine Immunol., 13: 1181–1184.<br />

Staras SAS, Dollard SC, Radford KW, Flanders WD, Pass RF, Cannon<br />

MJ (2006). Seroprevalence of Cytomegalovirus Infection in the<br />

United States, 1988–1994, Clin. Infect. Dis., 43: 1143–1151.<br />

Staras SAS, Flanders WD, Dollard SC, Pass RF, McGowan JE, Cannon<br />

MJ (2008). Cytomegalovirus seroprevalence and childhood sources<br />

of infection: A population-based study among pre-adolescents in the<br />

United States, J. Clin. Virol., 43: 266-271.<br />

Wisdom GB (1976). Enzyme – Immunoassay, Clin. Chem., 22: 1243.


African Journal of Microbiology Research Vol. 5(16), pp. 2349-2352, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

OI: 10.5897/AJMR11.169<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Antibacterial and cytotoxic activity of Eremurus<br />

persicus (Jaub and Spach) Boiss<br />

Mojdeh Hakemi Vala 1 , Jinous Asgarpanah 2 *, Mohammad Hossein Hedayati 3 , Jeilan Shirali 2<br />

and Fatemeh Bagheri Bejestani 4<br />

1 Microbiology Department, Shahid Beheshti University of Medical Sciences, Tehran, Iran.<br />

2 Pharmacognosy Department, Pharmaceutical Sciences Branch and Pharmaceutical Sciences Research Center,<br />

Islamic Azad University (IAU), Tehran, Iran.<br />

3 Pasteur Institute of Iran (Karaj branch), Iran.<br />

4 Microbiology Department, Pharmaceutical Sciences Branch, Islamic Azad University (IAU), Tehran, Iran.<br />

Accepted 30 May, 2011<br />

Traditional medicine has a key role in health care worldwide. Obtaining scientific information about the<br />

efficacy and safety of the plants is one of the researcher's goals. In this research, the flowering aerial<br />

parts of Eremurus persicus were collected from Golpayegan (Isfahan, Iran) in May 2010. The extract was<br />

tested for its antibacterial activity against 4 Gram-positive bacteria strains (Staphylococcus aureus,<br />

Staphylococcus epidermidis and Bacillus cereus, Streptococcus pyogenes) and 5 Gram-negative<br />

bacterial strains (Escherichia coli, Salmonella typhi, Shigella dysantriae, Klebsiella pneumoniae,<br />

Pseudomonas aeruginosa). Its cytotoxic activity was also investigated using MTT assay. Here we<br />

reported the antibacterial activity of E. persicus against S. aureus (MIC = 125 mg/ml), B. cereus (MIC =<br />

15.62 mg/ml), E. coli (MIC = 125 mg/ml), S. typhi (MIC = 31.25 mg/ml), S. dysantriae (MIC = 0.48 mg/ml)<br />

for the first time. Furthermore the cytotoxic potentials of E. persicus on two cell lines, HeLa and Caco-2,<br />

were studied.<br />

Key words: Eremurus persicus, antibacterial activity, cytotoxic activity.<br />

INTRODUCTION<br />

The genus Eremurus (Liliaceae) comprising nearly 50<br />

species, is mainly restricted to central and western Asia<br />

(Chong et al., 2000) and six species are known to exist in<br />

Iran. Eremurus persicus locally called "Serish” is widely<br />

distributed in south, east and west of Iran. The roots are<br />

used as natural glue. The leaves are traditionally used to<br />

relieve constipation and treatment of diabetes, liver and<br />

stomach disorders. Polysaccharides have been reported<br />

as chemical constituents of species in the genus<br />

Eremurus (Chong et al., 2000). Since the literature<br />

survey revealed that there is no any information on the<br />

biological activities of E. persicus, we prompted to investigate<br />

the antibacterial activity of methanolic extract of<br />

flowering aerial parts of this plant by using the cup plate<br />

*Corresponding author. E-mail: asgarpanah@iaups.ac.ir. Tel:<br />

+98 21 22640051. Fax: +98 21 22602059.<br />

method and determine its cytotoxic effect after infection<br />

by two cell lines (HeLa and Caco-2) and further by MTT<br />

assay.<br />

MATERIALS AND METHODS<br />

Plant material<br />

The flowering aerial part of E. persicus was collected from<br />

Golpayegan (Isfahan, Iran) in May 2010 and identified by Dr. Gh.R.<br />

Amin at the Pharmacognosy Department, Faculty of Pharmacy,<br />

Tehran University of Medical Sciences, Tehran, Iran. A voucher<br />

specimen (NO. 197) has been deposited in the herbarium of the<br />

Department of Pharmacognosy, Pharmaceutical Sciences Branch,<br />

Islamic Azad University (IAU), Tehran, Iran.<br />

Extraction and isolation<br />

The dried ground material was extracted by percolator apparatus


2350 Afr. J. Microbiol. Res.<br />

Table 1. IC50 of the methanolic extract of Eremurus persicus and DMSO in cell lines.<br />

Cell line<br />

HeLa<br />

Caco-2<br />

Eremurus persicus extract IC50 (mg/ml)<br />

4.74<br />

5<br />

using methanol (Merck). The extract was concentrated by rotary<br />

evaporator apparatus and the solvent removed to produce a dark<br />

brown gummy solid. The resulting extract was kept in a sterile vial<br />

in a dark and cool place for further tests.<br />

Bacterial strains<br />

Gram-positive bacteria including Staphylococcus aureus (PTCC<br />

1431), Staphylococcus epidermidis (PTCC 1435) and Bacillus<br />

cereus (PTCC 1247), Streptococcus pyogenes (PTCC 1447), and<br />

Gram-negative bacteria including Escherichia coli (PTCC1399),<br />

Salmonella typhi (PTCC 1639), Shigella dysantriae (PTCC 1188),<br />

Klebsiella pneumonia (PTCC 1053), Pseudomonas aeruginosa<br />

(PTCC 1430) were obtained from Persian Type Culture Collection,<br />

Iranian Research Organization for Science and Technology (PTCC,<br />

Iran).<br />

Antibacterial assay<br />

Antibacterial activity of the methanolic crude extract of E. persicus<br />

was investigated against 9 bacterial strains by the cup plate method<br />

(Fazyl Bazzaz et al., 2005). An overnight bacterial culture<br />

containing 1.5 x 10 8 CFU/ml was used to culture on surface of<br />

Muller-Hinton agar plates. The wells were made on agar plates.<br />

1000, 500 and 250 mg, respectively, of the extract were dissolved<br />

in 1 ml DMSO 10% and then filtered. 80 l of each test solution was<br />

added to each well. Following diffusion of solutions into agar, the<br />

plates were incubated at 37°C for 24 h. The diameter of inhibition<br />

zones around each well were determined in comparison with the<br />

well of ciprofloxacin (31.25 mg/ml) which was used as positive<br />

control. A well added by 80 l DMSO 10% instead of the extract<br />

solution, served as negative control too. The experiments carried<br />

out 3 times and the results were presented as mean ± SD.<br />

Minimum inhibitory concentration (MIC)<br />

After confirmation of the antibacterial activity in methanolic crude<br />

extract of E. persicus, MIC of the extract was determine by testing<br />

10 concentrations of the extract against sensitive Gram-positive<br />

and Gram-negative tested bacteria by the micro plate dilution<br />

method. The reconstituted extract was diluted to give concentrations<br />

of 250, 125, 62.5, 31.25, 15.62, 7.81, 3.90, 1.95, 0.97 and<br />

0.48 mg/ml, respectively. The lowest concentration of the extract<br />

that could inhibit the bacterial growth was considered as MIC<br />

(Mehregan et al., 2008). The data was subject to statistical analysis<br />

of SPSS software.<br />

Cytotoxic activity<br />

Cell culture<br />

HeLa (Human cervix carcinoma) and Caco-2 (Human colon<br />

carcinoma) cell lines were obtained from Iranian Cell Bank of<br />

Institute Pasteur of Iran. HeLa cells was grown in a flask and RPMI<br />

DMSO 10% IC50 (v/v)<br />

4.45 %<br />

4.73 %<br />

1640 media (Gibco, Germany) which was supplemented with 10%<br />

fetal bovine serum; FBS (Gibco, Germany) and 20mg/ml gentamicin<br />

(Sigma) was added. In addition, Caco-2 cell line was grown in<br />

DMEM media (Gibco, Germany) which was supplemented with 20%<br />

FBS and 20 mg/ml gentamicin. All plated were incubated in a CO2<br />

incubator at 37°C. 100 µl of 3 x 10 5 cells/ml suspension of HeLa cell<br />

line and 100 µl of 6 x 10 5 cells/ml suspension of Caco-2 cell line<br />

transferred to a 96- well microplate and incubated at the same<br />

condition.<br />

MTT assay<br />

Based on determined Ic50 (Table 1), 17 mg methanolic extract of E.<br />

persicus dissolved in DMSO 1%, separately for HeLa and Caco-2<br />

assay and 100 µl of each concentration was transferred in a row of<br />

separate microplate to make serial dilution. After 72 h incubation in<br />

a CO2 incubator at 37ºC, 20 µl of MTT (5 mg/ml in PBS) was added<br />

and incubated at the same condition for 3 to 5 h for MTT assay.<br />

After Formazan crystallization, 50 µl of isobutyl alcohol was added<br />

and OD (Optical density) was read at 570 nm wave length (Figures<br />

1 to 2).<br />

RESULTS<br />

The inhibitory effects of methanolic extract of E. persicus<br />

against different test organisms are shown in Table 2 (the<br />

p-value mentioned in Table 2 is only for the 1000 mg/ml<br />

concentration of extract). This extract indicated significant<br />

antibacterial activity (growth inhibition zone diameters<br />

ranging from (12.5 to 32 mm) against some Grampositive<br />

bacteria including S. aureus, B. cereus, S.<br />

epidermidis and Gram-negative bacteria including E. coli,<br />

S. typhi and specially S. dysantriae.<br />

Also, the MIC of the extract was determined by using<br />

the micro plate dilution method for Gram-positive<br />

bacteria, S. aureus and B. cereus. In addition, the<br />

methanolic extract could inhibit the growth of other Gramnegative<br />

bacteria at 125 and 15.62 mg/ml, respectively;<br />

the extract could effectively inhibit other test bacteria, E.<br />

coli, S. dysantriae and S. typhi, even at concentrations as<br />

low as 125, 0.48 and 31.25 mg/ml, respectively.<br />

The cytotoxicity effect of methanolic extract of E.<br />

persicus on HeLa cell and Caco-2 cell culture was<br />

evaluated by MTT assay. The result shows that the more<br />

concentration of extract in the cell culture the less OD<br />

was detected. It means, after increasing the concentration<br />

of methanolic extract the viability of cells were<br />

decreased. Similarly, both cell lines showed decrease in<br />

cell viability after the concentration of extract was<br />

increased and decrease in viability begins when 2.125<br />

and 4.25 mg/ml of methanolic extract was added to


Eremurus persicus<br />

Figure 1. Absorbance value versus concentration of E. persicus extract and DMSO in Caco-2 cell line.<br />

Eremurus persicus<br />

Figure 2. Absorbance value versus concentration of E. persicus extract and DMSO in HeLa cell line.<br />

Table 2. Antibacterial activity of E. persicus methanolic extract against various bacteria, as obtained by cup plate method (n=6).<br />

S/No.<br />

1<br />

2<br />

3<br />

4<br />

5<br />

6<br />

Organism<br />

S. aureus<br />

S. epidermidis<br />

B. cereus<br />

E-coli<br />

S. typhi<br />

S. dysantriae<br />

Mean zone of inhibition in mm and standard deviation (SD)<br />

A<br />

B<br />

C<br />

D<br />

Mean ± SD Mean ± SD Mean ± SD Mean ± SD<br />

0.0±0.0<br />

0.0±0.0<br />

0.0±0.0<br />

0.0 ±0.0<br />

0.0±0.0<br />

28.0±0.0<br />

11.0±1.1<br />

0.0±0.0<br />

0.0±0.0<br />

0.0 ±0.0<br />

0.0 ±0.0<br />

31.0±0.0<br />

17.5±0.5<br />

15.0±0.0<br />

13.5±1.6<br />

12.5± 0.5<br />

14.0±1.1<br />

32.0±0.0<br />

60.0±0.0<br />

57.0±3.3<br />

46.0±0.0<br />

50.0±0.0<br />

48.0±2.2<br />

60.0±0.0<br />

P- value*<br />


2352 Afr. J. Microbiol. Res.<br />

Eremurus persicus<br />

Figure 3. Effects of E. persicus extract and DMSO on cell viability in Caco-2 cell line.<br />

Eremurus persicus<br />

Figure 4. Effects of E. persicus extract and DMSO on cell viability in HeLa cell line.<br />

shows more sensitivity than Caco-2 cell line against both<br />

E. persicus extract and DMSO concentrations as positive<br />

control.<br />

DISCUSSION<br />

Since the presence of anthraquinones has been indicated<br />

in the aerial parts of other species of this genus (Chong<br />

et al., 2000), we suggest the strong antibacterial effect<br />

due to anthraquinones because they are more likely to<br />

possess antibacterial activity (Chukwujecwu et al., 2006).<br />

The cytotoxic activity is also related to the anthraxquinones<br />

because it is shown that they are able to induce<br />

opoptosis due to the fact that they are suitable substrate<br />

for one-electron-reducing enzymes and effective redox<br />

cycler which lead to the production of oxygen derived free<br />

radicals that eventually induce opoptotic cell death<br />

(Kagedal et al., 1999). Based on the results of this study,<br />

further in-vivo and ex-vivo confirmatory tests are<br />

recommended.<br />

ACKNOWLEDGMENT<br />

This work was supported by Pharmaceutical Sciences<br />

Branch, Islamic Azad University (IAU), Tehran, Iran and<br />

the authors are thankful for this support. We would like to<br />

thank Mr. M. Asoudeh for collecting the plant material.<br />

REFERENCES<br />

Chong L, Jian G, Peng Zhang Y, Zhong Z (2000). Constituents of<br />

Eremurus chinensis. J. Nat. Pro., 63: 653-656.<br />

Chukwujecwu JC, Coombes PH, Mulholland DA, Vanstaden J (2006).<br />

Emodin, an antibacterial anthraquinone from the roots of Cassia<br />

occidentalis. South Afr. J. Bot., 72(2): 295-297.<br />

Fazly Bazzaz BS, Khajehkaramadin M, Shokooheizadeh HR (2005).<br />

Antibacterial activity of Rheum ribes extract obtained from various<br />

plant parts against clinical isolates of Gram-negative pathogens. Iran.<br />

J. Pharm. Res., 2: 87-91.<br />

Kagedal K, Bironaite D, Ollinger K (1999). Anthraquinone cytotoxicity<br />

and opoptosis in primary cultures of rat hepatocytes. Free. Radic.<br />

Res., 31(5): 419-428.<br />

Mehregan H, Mojab F, Pakdaman SH, Poursaeed M (2008).<br />

Antibacterial activity of Thymus pubescence methanolic extract. Iran.<br />

J. Pharm. Res., 7(4): 291-295.


African Journal of Microbiology Research Vol. 5(16), pp. 2353-2358, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.174<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Optimization of ultrasound-assisted extraction<br />

conditions using orthogonal matrix design to enhance<br />

the antimicrobial activity of extracts from Cichorium<br />

intybus root<br />

Quanzhen Wang 1 *, Haitao Liu 1 , Jinhong Du 1 , Jian Cui 2 , Guo Chen 1 and Yuyan Liu 1<br />

1 Department of Grassland Science, College of Animal Science and Technology, Northwest Agriculture and Forestry<br />

University, 712100 Yangling, Shanxi Province, P. R. China.<br />

2 Department of Plant Science, College of Life Science, Northwest Agriculture and Forestry University, 712100,<br />

Yangling, Shanxi Province, P. R. China.<br />

Accepted 30 July, 2011<br />

Plant-derived compounds used as alternatives to chemical preservatives have been extensively<br />

researched for use as natural medical ingredients or food preservatives. Conditions for the ultrasoundassisted<br />

extraction (UAE) of chicory (Cichorium intybus) root (including type of solvent, impregnation<br />

time, number of sonication steps and ultrasonic power) were optimized to determine the best extract<br />

antibacterial activity by using orthogonal matrix design [L16 (4 5 )]. The combination of 70% ethanol v/v, a<br />

36 h impregnation time, three sonication rounds and 300 W ultrasonic power input provided the best<br />

antimicrobial activity results. Our results demonstrate that solvent composition has the largest effect<br />

on antimicrobial activity. Several extracts demonstrated antibacterial activity against Escherichia coli,<br />

Staphylococcus aureus, Bacillus thuringiensis and Salmonella typhi, and all extracts exhibited weak<br />

activity against Bacillus subtilis. To our knowledge, these results represent the first example of<br />

ultrasound-assisted chicory extracts aimed at increasing its potential for use in food industry.<br />

Key words: Cichorium intybus, antibacterial, ultrasound-assistant extract, optimal conditions, orthogonal matrix<br />

design.<br />

INTRODUCTION<br />

There are a variety of useful compounds from higher<br />

plants that have not yet been identified. Medical and food<br />

industries tend to use natural antimicrobials, such as<br />

plant-derived compounds, as an alternative to chemical<br />

preservatives (Bachir and Mohamed, 2010). These<br />

preferences have led to the search for plant products that<br />

have natural antimicrobial and antioxidant effects. Such<br />

products have been extensively researched and used as<br />

both natural medical ingredients and food additive<br />

agents (Issa-Zacharia et al., 2010; Kumaravel et al.,<br />

2010; Liu et al., 2010; Mini et al., 2010; Somchit et al.,<br />

2010).<br />

*Corresponding author. E-mail: wangquanzhen191@163.com.<br />

Tel: 0086-29-87091953 or 0086-13759942845. Fax: 0086-29-<br />

87092164.<br />

Chicory (Cichorium intybus) is a perennial plant of the<br />

Asteraceae family, which is a native of to the<br />

Mediterranean region and widely grown in Europe,<br />

Western Asia, Egypt and North America. In traditional<br />

Indian medicine, chicory has been used to treat fever,<br />

diarrhea, spleen and liver enlargement, jaundice, gout<br />

and rheumatism (Mulabagal et al., 2009). In Belgium,<br />

France and the United States, chicory root has been<br />

used as a coffee additive for its bitter taste, which is<br />

caused by sesquiterpene lactones (Peters and<br />

Amerongen, 1996; Poli et al., 2002).<br />

A number of studies report that chicory extracts<br />

possess antimicrobial activities. It was reported that<br />

water, ethanol and ethyl acetate extracts from chicory<br />

have antibacterial properties, and that root extracts have<br />

more intensive antibacterial activity than extracts from<br />

whole plants (Petrovic et al., 2004). Moreover, it was<br />

found that the sesquiterpene lactones extracted from


2354 Afr. J. Microbiol. Res.<br />

Table 1. Assignment of levels and condition factors using orthogonal matrix L16 (4 5 ).<br />

Levels<br />

Factors<br />

A (Solvent) B [Impregnation time (h)] C (Sonication repetitions) D [Ultrasonic input power (W)]<br />

I petroleum ether 12 1 200<br />

ii ethyl ether 24 2 300<br />

iii ethyl acetate 36 3 400<br />

iv 70% ethanol v/v 48 4 500<br />

chicory root can inhibit the growth of zoophilic and<br />

anthropophilic dermatophytes (Mares et al., 2005). Many<br />

gastrointestinal foodborne infections in humans, particularly<br />

those in developing countries, are often caused by<br />

enterobacteria (Ajayi and Akintola, 2010). Increasing<br />

concern over pathogenic and spoilage microorganisms in<br />

food is due to the growing prevalence of food borne<br />

disease outbreaks (Rahman and Kang, 2009). Considering<br />

the questionable safety of synthetic food<br />

preservatives, consumers are demanding more natural<br />

and fresh foods with fewer synthetic additives. However,<br />

to increase safety and maintain a long shelf life, food<br />

manufacturers are compelled to use natural or mild preservation<br />

techniques. Therefore, alternative sources of<br />

safe and effective natural preservatives must be explored<br />

(Abbasi et al., 2010; Negi et al., 2008; Hussain et al.,<br />

2009).<br />

Orthogonal matrix design has been used to evaluate<br />

the interaction of different production parameters and<br />

how they affect product recovery. Furthermore, this<br />

method has been used to optimize multiple production<br />

parameters to establish optimum conditions (Hedayat et<br />

al., 1999). At present, ultrasound-assisted extraction<br />

(UAE) is considered a desirable method for organic<br />

compound extraction from different matrices (Khan et al.,<br />

2010). This method can shorten extraction time because<br />

the increased pressure favors penetration and transport<br />

while the temperature decreased may improve solubility<br />

and diffusivity (Pena et al., 2006).<br />

In order to optimize conditions to achieve the best<br />

antibacterial activity for chicory extract use as a natural<br />

food preservative, the solvent, impregnation time, sonication<br />

repetitions and ultrasonic input power were evaluated<br />

by orthogonal matrix design. The L16 (4 5 ) experiment was<br />

adopted and sixteen different extraction groups were<br />

analyzed. The antibacterial activities of extracts derived<br />

using different extraction protocols were investigated.<br />

MATERIALS AND METHODS<br />

Plant materials<br />

Five-year-old Puna chicory roots were freshly harvested in<br />

September 2009 in an experimental field at the Grassland Science<br />

Department at Northwest Agriculture and Forestry University<br />

(Shaanxi Province, China). The plant was identified by associate<br />

professor Quanzhen Wang (Northwest Agriculture and Forestry<br />

University, China). A voucher specimen was deposited in the<br />

Herbarium of the Laboratory of Grassland Science at the Faculty of<br />

Animal Science and Technology (Northwest Agriculture and<br />

Forestry University, China).<br />

Preparation of plant extracts<br />

Four levels for each of four different extraction conditions resulting<br />

in sixteen extraction method combinations (Table 1) were studied<br />

by an L16 (4 5 ) orthogonal matrix design (Hedayat et al., 1999). For<br />

each sample, 100 g dried ground powder (40 meshes) was soaked<br />

in 800 ml of various solvents for different times at room temperature<br />

until extraction was exhausted. The extraction temperatures were<br />

30°C (ethyl ether), 50°C (petroleum ether and ethyl acetate) and<br />

65°C (70% ethanol v/v), respectively. An ultrasonic apparatus (KQ-<br />

500DE, Kunshan Ultrasound Instrument Co., Ltd., China) was used<br />

for accelerated extraction. A beaker was partially submerged in an<br />

isothermal water bath to maintain the extraction temperature for 30<br />

min. After extraction, the resulting mixture was passed through filter<br />

paper (Whatman No. 1.) by vacuum. The filtrate was concentrated<br />

on a rotary evaporator (SHB-III, Zhengzhou Science and Industrial<br />

Foreign Trade Co., Ltd., China) at 45°C and then stored at 4°C for<br />

further use. All samples were redissolved in dimethylsulfoxide<br />

(DMSO) at a concentration of 10 mg/mL and stored at 4°C.<br />

Bacteria<br />

The antibacterial activity of chicory extracts was determined using<br />

the following food-related bacteria obtained from the Department of<br />

Life Science, Northwest Agriculture and Forestry University: E. coli,<br />

Staphylococcus aureus, Bacillus thuringiensis, Bacillus subtilis,<br />

Salmonella typh. Cultures of each strain were maintained on beef<br />

cream-peptone culture media at 4°C (Monadi et al., 2010; Moussa<br />

and Hessan, 2010).<br />

Antibacterial assays of disc diffusion<br />

Antibacterial activity tests were conducted based on the disc<br />

diffusion method (Yan et al., 2009). A suspension of bacteria (2×10 8<br />

CFU/ml) was spread on solid media plates. Sterile paper discs (6<br />

mm diameter) were individually impregnated with solvent at 10<br />

mg/ml. Discs with the solvent used for dissolution were used as<br />

negative controls and the standard reference antibiotic streptomycin<br />

(10 g/disc) was used as a positive control for the bacteria in<br />

question. The plates were incubated at 37°C for 24 h after which<br />

the inhibition zone was measured in millimeters. Each assay in this<br />

experiment was performed in triplicate.<br />

Statistical analysis<br />

All experimental results are expressed the mean of four repeats.


Table 2. The L16 (4 5 ) matrix associated with analytical results.<br />

Group����<br />

Factors and levels<br />

A B C D<br />

Staphylococcus<br />

aureus b<br />

Bacillus<br />

subtilis b<br />

Zone of inhibition in mm a<br />

Bacillus<br />

thuringiensis b<br />

Wang et al. 2355<br />

Salmonella<br />

typhi c<br />

Escherichia<br />

coli c<br />

1 1 1 4 3 10.2 7.2 9.7 8.6 9.7<br />

2 2 1 1 1 11.6 6.5 9.4 8.2 8.1<br />

3 3 1 3 4 8.5 8.5 13.1 12.3 11.1<br />

4 4 1 2 2 12.2 8.9 10.8 7.5 9.5<br />

5 1 2 3 2 12.1 7.1 11.8 10.2 9.2<br />

6 2 2 2 4 11.7 8.1 8.9 10.1 11.5<br />

7 3 2 4 1 9.2 9.6 9.6 11.6 12.0<br />

8 4 2 1 3 9.8 8.2 12.7 8.6 11.7<br />

9 1 3 1 4 11.0 7.9 8.6 10.4 7.6<br />

10 2 3 4 2 9.4 9.8 10.7 14.2 11.2<br />

11 3 3 2 3 8.6 7.5 10.2 10.9 10.9<br />

12 4 3 3 1 11.1 6.7 10.3 12.2 13.6<br />

13 1 4 2 1 8.3 8.7 10.2 7.4 10.2<br />

14 2 4 3 3 8.4 7.1 9.7 9.7 6.9<br />

15 3 4 1 2 9.1 7.5 9.4 11.4 8.1<br />

16 4 4 4 4 11.5 9.1 11.5 7.1 10.1<br />

a Values are means (mm) of four separate treatments. b Gram positive bacteria. c Gram negative bacteria.<br />

Analysis of variance was performed by ANOVA. SAS software<br />

(version 8.2, USA) was used for statistical analysis. Results were<br />

considered statistically significant at p ultrasonic input power (D) > impregnation<br />

time (B) > sonication repetition number (C) (Table 3). The<br />

optimal extraction parameters were defined as the<br />

following: treatment with 70% ethanol v/v (A4), 300 W<br />

ultrasonic input power (D2), 12 h impregnation time (B1)<br />

and two rounds of sonication (C2). The impact of<br />

variables on the inhibition zone against Bacillus subtilis<br />

was measured in the following order: C > D > A > B. The<br />

optimal combination of parameters for this bacterium was<br />

to use four sonication rounds (C4), 300 W ultrasonic input<br />

power (D2), ethyl ether (A2) and a 36 h impregnation time<br />

(B3). The impact of variables on the inhibition zone of B.<br />

thuringiensis was in the following order: A > C > B > D.<br />

The optimal combination was to use ethyl acetate (A3),<br />

three sonication rounds (C3), a 12 h impregnation time<br />

(B1) and 500 W ultrasonic input power (D4). The impact of<br />

variables on the inhibition zone of Salmonella typh was in<br />

the following order: B > A > C > D. The optimal parameter<br />

combination was to use a 36 h impregnation time (B3),<br />

ethyl ether (A2), four sonication rounds (C4), and 300 W<br />

ultrasonic input power (D2). The impact of variables on<br />

the inhibition zone of against E. coli was in the following<br />

order: B > A > C > D. The optimal parameter combination<br />

was found using a 36 h impregnation time (B3), 70%<br />

ethanol v/v (A4), three sonication rounds (C3), and 200 W<br />

ultrasonic input power (D1). According to the results for<br />

antibacterial activities against all five types of bacteria,<br />

we found that extraction using 70% ethanol v/v, a 24 h<br />

impregnation time, three sonication rounds and 300 W<br />

ultrasonic input power is the optimal synthetic<br />

combination.<br />

Variance analyses of the models of all bacteria but B.<br />

thuringiensis were found to be significant (Table 4. Pr <<br />

0.001). Factors A, B and D were significantly effective for<br />

inhibiting S. aureus whereas factors C and D were significant<br />

against Bacillus subtilis (Table 4). Factors A, B and<br />

C consistently inhibited the Gram-negative bacteria S.<br />

typhi and E. coli; however, factor D (ultrasonic input<br />

power) was not significant (Table 4). Solvent type was<br />

found to be the most influential factor; increasing solvent<br />

polarity resulted in better antibacterial activities primarily<br />

because substances such as carbohydrates, proteins,


2356 Afr. J. Microbiol. Res.<br />

Table 3. Averages of levels with factors and ranges.<br />

Factors A B C D A B C D<br />

Staphylococcus aureus Bacillus subtilis<br />

Level i 10.4 10.63 10.38 10.05 7.73 7.78 7.53 7.88<br />

Level ii 10.35 10.70 10.20 10.70 7.88 8.25 8.30 8.33<br />

Level iii 8.85 10.03 10.03 9.25 8.28 7.98 7.35 7.50<br />

Level iv 11.15 9.33 10.08 10.68 8.23 8.1 8.93 8.40<br />

Range a 2.3 1.37 0.35 1.45 0.55 0.47 1.58 0.90<br />

Order 1 3 4 2 3 4 1 2<br />

Bacillus thuringiensis Salmonella typhi<br />

Level i 10.08 10.75 10.03 9.88 9.15 9.15 9.65 9.85<br />

Level ii 9.68 10.75 10.03 10.67 10.55 10.13 8.98 10.83<br />

Level iii 10.58 9.95 11.23 10.58 11.55 11.93 11.10 9.45<br />

Level iv 11.33 10.2 10.38 10.53 8.85 8.90 10.38 9.98<br />

Range a 1.65 0.8 1.2 0.79 2.7 3.03 2.12 1.38<br />

Order 1 3 2 4 2 1 3 4<br />

Escherichia coli<br />

Level i 9.18 9.6 8.87 10.98<br />

Level ii 9.43 11.1 10.53 9.5<br />

Level iii 10.53 10.83 10.2 9.8<br />

Level iv 11.23 8.83 10.75 10.08<br />

Range a 2.05 2.27 1.88 0.58<br />

Order b 2 1 3 4<br />

a R means the average range for four average responses in each level for the inhibition zone against Staphylococcus aureus, Bacillus<br />

subtilis, Bacillus thuringiensis, Salmonella typhi, Escherichia coli, respectively.<br />

b The ordinal numeral for the range sequence of the eight factors in decreasing order.<br />

Table 4. Variance analysis for the model and experimental factors.<br />

Factor<br />

Model<br />

A<br />

B<br />

C<br />

D<br />

Staphylococcus<br />

aureus b<br />

Bacillus subtilis b<br />

Bacillus<br />

thuringiensis b<br />

Salmonella<br />

typhi c<br />

Escherichia coli c<br />

F Value 3.85 3.72 1.87 4.93 4.33<br />

Pr > F 0.0003 0.0005 0.0608 F 0.0003 0.3348 0.0248 0.0007 0.0040<br />

F Value 3.49 0.88 1.14 8.11 6.19<br />

Pr > F 0.0221 0.4567 0.3415 0.0002 0.0011<br />

F Value 0.22 9.74 2.15 3.58 3.89<br />

Pr > F 0.8807 F 0.0113 0.0350 0.4933 0.2560 0.0956<br />

b Gram positive bacteria. c Gram negative bacteria.<br />

alkaloids, tannins, glycosides and amines increase in<br />

abundance with increased solvent polarity. Impregnation<br />

time, sonication repetition number and ultrasonic input<br />

power were found to be subordinate factors. Most


ioactive compounds such as polyphenolics (e.g., tannins<br />

and flavonoids) exist in higher polarity solvents.<br />

Additional compounds found in the extracts may either<br />

enhance or attenuate the effect of phenolic compounds.<br />

The 70% ethanol and acetic ether extracts exhibited<br />

significant antibacterial activity against many of the<br />

bacteria in this study. This antibacterial activity may be<br />

ascribed to polyphenols, tannins and coumarins found in<br />

crude extracts (Kilani et al., 2008). Phenolic compounds<br />

can attack cell walls and membranes by affecting their<br />

permeability, enabling the release of intracellular constituents<br />

and interfering with membrane functionality (Bajpai<br />

et al., 2009). There were differences in chicory extract<br />

antibacterial activity against Gram-positive and Gramnegative<br />

bacteria. Gram-positive bacteria were found to<br />

be more susceptible to chicory extracts than Gramnegative<br />

bacteria, possibly because the hydrophilic cell<br />

walls of Gram-negative bacteria are composed of<br />

lipopolysaccharide (LPS), which inhibits the accumulation<br />

of phenolic compounds in a target cell membrane (Bezic<br />

et al., 2003).<br />

Chicory contains several sesquiterpene lactones,<br />

particularly in the roots (Beek et al., 1990; Peters and<br />

Amerongen 1996; Poli et al., 2002). Poli et al. has<br />

reported the isolation of two guaianolides with different<br />

chemical structures, including 8-deoxylactucin and 11 ,<br />

13-dihydrolactucin, from the root extracts of C. intybus<br />

var. ‘Rosso di Chioggia’ (Picman, 1986). Two pure<br />

isolated lactones possess similar biological activity to<br />

chicory extracts against several phytopathogens, which<br />

cause morphological anomalies that have been observed<br />

using scanning electron microscopy (Mares et al., 2005).<br />

Conclusions<br />

The antibacterial activity of chicory extracts was found to<br />

be affected by the type of solvent used for extraction. The<br />

optimal UAE conditions were obtained to produce the<br />

best chicory root extract antimicrobial activity. It was<br />

concluded that combining 70% ethanol v/v, a 36 h<br />

impregnation time, three sonication rounds and 300 W<br />

ultrasonic input power could produce the highest<br />

antibacterial activity in the extracts. Extracts from chicory<br />

roots can be used as a natural food preservative as well<br />

as an antibacterial agent. To our knowledge, these<br />

results represent the first example of a practical application<br />

for UAE aimed at increasing the potential use of<br />

chicory extracts in food industry. Chicory root<br />

antibacterial activities against additional bacterial strains<br />

should also be studied.<br />

ACKNOWLEDGMENTS<br />

This work was supported by The Ministry of Science and<br />

Technology of the Peoples Republic of China,<br />

International Cooperation Research Project<br />

(2008DFA31650) and Shaanxi Province International<br />

Wang et al. 2357<br />

Cooperation Research Project (2008KW-29). We are<br />

grateful to Prof. Hu Tianming and postgraduate students<br />

Sun Luanzhi and Wu Chunhui.<br />

REFERENCES<br />

Abbasi FM, Ahmad H, Perveen F, Inamullah, Sajid M, Brar DS (2010).<br />

Assesment of genomic relationship between Oryza sativa and Oryza<br />

australiensis. Afr. J. Biotechnol., 9(12): 1312-1316.<br />

Ajayi AO, Akintola TA (2010). Evaluation of antibacterial activity of some<br />

medicinal plants on common enteric food-borne pathogens. Afr. J.<br />

Microbiol. Res., 4: 314-316.<br />

Bachir RG, Mohamed B (2010). Antimicrobial activity determination of<br />

the gum of Pistacia atlantica Desf. Oil. Afr. J. Microbiol. Res., 4:<br />

2457-2460.<br />

Bajpai VK, Al-Reza SM, Choi UK, Lee JH, Kang SC (2009). Chemical<br />

composition, antibacterial and antioxidant activities of leaf essential<br />

oil and extracts of Metasequioa glyptostroboides Miki ex Hu. Food<br />

Chemic. Toxicol., 47: 1876-1883.<br />

Beek TV, Maas P, King B, Leclercq E, Voragen A, Groot AD (1990).<br />

Bitter sesquiterpene lactones from chicory roots. J. Agric. Food<br />

Chem., 38: 1035-1038.<br />

Bezic N, Skocibusic M, Dunkic V, Radonic A (2003). Composition and<br />

antimicrobial activity of Achillea clavennae L. essential oil. Phytother.<br />

Res., 17: 1037-1040.<br />

Hedayat A, Sloane N, Stufken J (1999). Orthogonal arrays: theory and<br />

applications: Springer Verlag.<br />

Issa-Zacharia A, Kamitani Y, Muhimbula H, Iwasaki K (2010).<br />

Antimicrobial effect of slightly acidic electrolyzed water for inactivation<br />

of Salmonella spp. and Escherichia coli on fresh strawberries<br />

(Fragaria L.). Afr. J. Microbiol. Res., 4: 2174-2180.<br />

Khan MK, Abert-Vian M, Fabiano-Tixier AS, Dangles O, Chemat F<br />

(2010). Ultrasound- assisted extraction of polyphenols (flavanone<br />

glycosides) from orange (Citrus sinensis L.) peel. Food Chem., 119:<br />

851-858.<br />

Kilani S, Sghaier MB, Limem I, Bouhlel I, Boubaker J, Bhouri W (2008).<br />

In vitro evaluation of antibacterial, antioxidant, cytotoxic and apoptotic<br />

activities of the tubers infusion and extracts of Cyperus rotundus.<br />

Bioresource Technol., 99: 9004-9008.<br />

Kumaravel K, Ravichandran S, Sritama bose S (2010). In vitro<br />

antimicrobial activity of shrimps haemolymph on clinical pathogens.<br />

Afr. J. Microbiol. Res., 4: 2592-2596.<br />

Liu GQ, Lin QL, Guo-Pu Ren GP, Han WJ, Wang XL, Zhang HY (2010).<br />

Use of response surface methodology to optimize critical medium<br />

components for biomass and extracellular polysaccharide production<br />

by Ganoderma sinense. Afr. J. Microbiol. Res., 4: 2735-2741.<br />

Mares D, Romagnoli C, Tosi B, Andreotti E, Chillemi G, Poli F (2005).<br />

Chicory extracts from Cichorium intybus L. as potential antifungals.<br />

Mycopathologia, 160: 85-92.<br />

Mini NV, Ida B, Seema D, Shital D, Riva DS, Astrida R (2010).<br />

Antimicrobial activity of ten common herbs, commonly known as<br />

‘Dashapushpam’ from Kerala, India. Afr. J. Microbiol. Res., 4: 2357-<br />

2362.<br />

Monadi AR, Mirzaei H, Javadi A, Hosseinzade N, Amjadi Y (2010).<br />

Effect of some probiotics on Salmonella typhi during associated<br />

growth in milk. Afr. J. Microbiol. Res., 4: 2708-2711.<br />

Moussa IM, Hessan AM (2010). Rapid detection of community<br />

acquired- methicillin resistance Staphylococcus aureus recovered<br />

from King Saudi Arabia. Afr. J. Microbiol. Res., 4: 2804-2810.<br />

Mulabagal V, Wang H, Ngouajio M, Nair MG (2009). Characterization<br />

and quantification of health beneficial anthocyanins in leaf chicory<br />

(Cichorium intybus) varieties. European Food Res. Technol., 230: 47-<br />

53.<br />

Negi PS, Jayaprakasha GK, Jena BS (2008). Antibacterial activity of the<br />

extracts from the fruit rinds of Garcinia cowa and Garcinia<br />

pedunculata against food borne pathogens and spoilage bacteria.<br />

Lwt-Food Sci. Technol., 41: 1857-1861.<br />

Pena A, Ruano F, Mingorance MD (2006). Ultrasound-assisted<br />

extraction of pesticides from olive branches: a multifactorial approach<br />

to method development. Analytic. Bioanalytic. Chem., 385: 918-925.


2358 Afr. J. Microbiol. Res.<br />

Peters AM, Amerongen AV (1996). Sesquiterpene lactones in chicory<br />

(Cichorium intybus L) - Distribution in chicons and effect of storage,<br />

Food Res. Int., 29: 439-444.<br />

Petrovic J, Stanojkovic A, Comic L, Curcic S (2004). Antibacterial<br />

activity of Cichorium intybus. Fitoterapia, 75: 737-739.<br />

Picman A (1986). Biological activities of sesquiterpene lactones.<br />

Biochem. Syst. Ecol., 14: 255-281.<br />

Poli F, Sacchetti G, Tosi B, Fogagnolo M, Chillemi G, Lazzarin R<br />

(2002). Variation in the content of the main guaianolides and sugars<br />

in Cichorium intybus var. "Rosso di Chioggia" selections during<br />

cultivation. Food Chem., 76: 139-147.<br />

Rahman A, Kang S (2009). In vitro control of food-borne and food<br />

spoilage bacteria by essential oil and ethanol extracts of Lonicera<br />

japonica Thunb. Food Chem., 116: 670-675.<br />

Somchit MN, Abdul RR, Abdullah A, Zuraini A, Zakaria ZA, Sulaiman<br />

MR, Arifah AK, Mutalib AR (2010). In vitro antimicrobial activity of<br />

leaves of Acalypha indica Linn. (Euphorbiaceae). Afr. J. Microbiol.<br />

Res., 4: 2133-2136.<br />

Yan RW, Yang Y, Zeng Y, Zou G (2009). Cytotoxicity and antibacterial<br />

activity of Lindera strychnifolia essential oils and extracts. J.<br />

Ethnopharmacol., 121: 451-455.


African Journal of Microbiology Research Vol. 5(16), pp. 2359-2366, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.273<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

Optimization of the production of exopolysaccharides<br />

by Bacillus thuringiensis 27 in sand biological soil<br />

crusts and its bioflocculant activity<br />

Z. R. Wang 1.2 , J. P. Sheng 1 , X. L. Tian 1 , T. T. Wu 1 , W. Z. Liu 1 and L. Shen 1 *<br />

1 College of Food Science and Nutritional Engineering, China Agricultural University, Beijing100083, China.<br />

2 School of Food Science, Henan Institute of Science and Technology, Xinxiang 453003, China.<br />

Accepted 30 June, 2011<br />

To improve the yield of exopolysaccharides (EPS) by Bacillus thuringiensis 27 from sand biological soil<br />

crusts in Gurban Tonggut Desert, Xinjiang, China and to analyze its bioflocculant activity, orthogonal<br />

matrix method was used and this method enabled us to obtain maximum EPS production. By studying<br />

the optimal medium condition of beef extract 3 g/l, peptone 10 g/l, maltose 40 g/l, and NaCl 4g/l we<br />

observed that the optimal medium condition was pH 6.0, incolum size 8%, liquid volume 40 ml in 200 ml<br />

flak and temperature 28°C. The maximum EPS production was 20.19 g/l which is about five times more<br />

than that at the basal condition. Furthermore, results obtained indicated that the flocculation activity of<br />

the extracellular polymer can be achieved at over 80.4% in kaolin suspension and this occurred at a<br />

concentration of 0.4 mg/l. This paper describes the optimum condition of exopolysaccharides<br />

production by Bacillus thuringiensis 27 and showed that exopolysaccharides had high bioflocculant<br />

activity. This work provides a scientific foundation to explore new exopolysaccharides and<br />

bioflocculation in sand biological soil crusts. Maximum production of exopolysacchaides under the<br />

optimal medium and condition can be achieved and exopolysaccharides have high bioflocculating<br />

activity in kaolin system.<br />

Key words: Biological soil crusts, exopolysaccharides, flocculation, optimization, Bacillus.<br />

INTRODUCTION<br />

Extreme environments, once thought to be too hostile to<br />

permit survival of living organisms, are the natural habitat<br />

of certain microorganisms. It is now recognized that the<br />

microorganisms living in extreme environments have<br />

different genetic background and metabolic pathway<br />

when compare to general microbiology and their seconddary<br />

metabolites have special function (Kennedy et al.,<br />

2001). Among the secondary metabolites from extreme<br />

environments, polysaccharide for biotechnological applications<br />

has been reported in Literature (Nicolaus et al.,<br />

2004). Therefore a wide search for bacteria that are able<br />

*Corresponding author. E-mail: pingshen@cau.edu.cn. Tel:<br />

8610-62738456. Fax: 8610-62737604.<br />

Abbreviation: EPS, Exopolysaccharide.<br />

to produce new polysaccharides with potentially useful<br />

properties has been undertaken. Sand biological soil<br />

crusts (BSCs) as extreme environment are a unique mininature<br />

landscape in desert district as well as the obvious<br />

sign of fixing mobile dune. They are composed of living<br />

microorganisms, their product of metabolism (mainly<br />

extracellular polysaccharides) and sand granule<br />

(Gundlapally and Garcia-Pichel, 2006).<br />

Exopolysaccharides (EPS) produced by microorganisms<br />

in BSCs are barely reported in the recent research.<br />

Medium condition and other bacteria growth conditions<br />

are important factors for EPS production. There are large<br />

numbers of reports on optimization of EPS production by<br />

statistical optimization techniques (Xu et al., 2003; 2010;<br />

Kaditzky and Vogel, 2008; Hao et al., 2010) Orthogonal<br />

design as one of the important statistical methods has<br />

been successfully applied to improvement of the


2360 Afr. J. Microbiol. Res.<br />

Table 1. Orthogonal matrix table of the L9 (3 4 ).<br />

Factor 1 2 3<br />

Beef extract (g/100ml) 0.2 0.3 0.4<br />

Peptone (g/100ml) 0.5 1.0 1.5<br />

Maltose (g/100ml) 2 3 4<br />

NaCl (g/100ml) 0.4 0.5 0.6<br />

production of primary and secondary metabolites in<br />

cultivation process (Xu et al., 2003).<br />

This work is an attempt to isolated strain producing<br />

EPS from sand BSCs, analysize suitable media and<br />

culture condition for the production of EPS from B.<br />

thuringiensis 27 and examines the bioflocculaton activity<br />

of EPS. To the best of our knowledge, the nutritional<br />

requirements and culture condition of EPS production<br />

form B. thuringiensis 27 isolated from sand BSCs and its<br />

bioflocculation activity has not been demonstrated.<br />

MATERIALS AND METHODS<br />

Bacterial strain<br />

Bacillus thuringiensis 27 was originally isolated from BSCs collected<br />

in the Gurban Tonggut Desert. It was maintained on agar slants<br />

containing (g/l): beef extract 3; peptone 10; NaCl 5 and agar 20 (pH<br />

7.0-7.4). The slants were incubated at 35°C for 24 h and the fully<br />

grown slants were stored at 4°C.<br />

Culture medium<br />

Basal medium, beef extract 3 g, peptone 10 g, NaCl 5 g, water<br />

1000 g, pH 7.0-7.4. Fermentation medium, beef extract 3 g,<br />

peptone 10 g, NaCl 5 g, water 1000 g, pH 7.0-7.4, temperature<br />

35°C.<br />

Single factor experiments of culture requirement<br />

To find the optimal culture requirements, the following factors were<br />

investigated using the one-factor-at-a-time method, including<br />

carbon sources nitrogen sources inorganic ions, initial pH value,<br />

cultivation temperature, inoculum size, liquid volume triangle flask.<br />

All experiments were performed in triplicates (n = 3).<br />

Orthogonal matrix method<br />

The orthogonal L9 (3 4 ) was used to obtain the optimal medium after<br />

the test by the one-factor-at-a-time method. This enables us to<br />

determine which process variables affect the response. A logical<br />

next step is to determine the point in the important factors that<br />

leads to the best possible response (Di et al., 2003; Li et al., 2001).<br />

The levels of components of the culture medium are listed in Table<br />

1. All experiments were performed in triplicate (n = 3). The software<br />

SPSS was used for experimental design, data analysis and model<br />

building. The optimal fermentation conditions for enhanced yield of<br />

EPS and cell growth were obtained by solving the regression<br />

equation using the software Origin 8.0.<br />

Determination of flocculating activity<br />

A kaolin suspension was used to measure the flocculating activity<br />

of the EPS crude extracts. One milliliter of 90 mmol/l CaCl2 and one<br />

milliliter were added into 50 ml 4.0 g/l kaolin suspension. The<br />

mixture was vigorously stirred for 0.5 min, and stand for 4 min. The<br />

optical density (OD) of the clarifying solution was measured with a<br />

spectrophotometer at 550 nm. A control experiment was conducted<br />

in the same manner by replacing distilled water. The flocculating<br />

activity was calculated according to the following equation (Kurane<br />

et al., 1986):<br />

η= (A-B) ×100/A<br />

where; A is the optical density of the control experiment at 550 nm<br />

and B is the optical density of the sample experiment at 550 nm. All<br />

experiments were performed in triplicates (n = 3).<br />

Analytical methods<br />

Samples collected at various culture conditions from shake flasks<br />

were centrifuged at 7000 g for 15 min. The 2 ml resulting<br />

supernatant was precipitated with threefold times 95% ethanol,<br />

stirred vigorously, and left overnight at 4°C. The precipitated<br />

polysaccharides were collected by centrifugation at 7000 g for 15<br />

min, discarding the supernatants, and dissolved with distilled water,<br />

dialyzing for 24 h, adding distilled water to 20 ml. Phenol-Sulfuric<br />

Acid Method was used for determining EPS yield (Pazur et al.,<br />

1994), using glucose solution as a standard reference. The EPS<br />

yield was expressed as gram per liter.<br />

RESULTS<br />

Effect of carbon and nitrogen source on EPS yield<br />

To find out the optimal carbon and nitrogen source for the<br />

EPS production of B. thuringiensis 27 five 2% carbon<br />

sources (glucose, maltose, sucrose, lactose and glycerol)<br />

were separately provided in the basal medium and five<br />

1% nitrogen sources (ammonium chloride, ammonium<br />

sulfate, peptone, beef extract and yeast extract) were<br />

separately instead of peptone employed in the basal<br />

medium. A high level of EPS was obtained when maltose<br />

and glycerol were used as the carbon source. Among the<br />

carbon sources tested, maximum EPS (10.45 g/l) was<br />

obtained in the maltose medium. Based on this study<br />

maltose is a good candidate. The maximum EPS


EPS prduction (g/l)<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Basal medium<br />

Glucose<br />

Frucose<br />

Maltose<br />

Carbon sources<br />

Lactose<br />

Glycole<br />

Figure 1. Effect of carbon and nitrogen sources on EPS production.<br />

production (10.53 g/l) was obtained in the peptone<br />

(Figure 1). So we selected maltose peptone as our<br />

optimal nitrogen source.<br />

Effect of concentration of carbon and nitrogen<br />

source on EPS yield<br />

To find out the suitable concentration of maltose for the<br />

EPS yield B. thuringiensis 27 five different concentration<br />

(1, 2, 3, 4 and 5%) maltose were separately instead of<br />

2% maltose above. The maximum EPS production (10.25<br />

g/l) was obtained when the concentration is 3% (Figure<br />

2). To find out the suitable concentration of nitrogen<br />

source for the EPS yield five different concentration (0.1,<br />

0.2, 0.5, 1.0 and 1.5%) peptone were separately instead<br />

of 1% peptone above. The maximum EPS production<br />

(10.74 g/l) was obtained when the concentration is 1%<br />

(Figure 2).<br />

Effect of inorganic ions and its concentration on EPS<br />

yield<br />

To find out the suitable mineral elements for the EPS<br />

yield of B. thuringiensis 27 six different 0.5% mineral<br />

elements (sodium chloride, magnesium chloride, copper<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

NaNO3<br />

NH4Cl<br />

NH4SO4<br />

Nitrogen sources<br />

peptone<br />

Wang et al. 2361<br />

Yeast extracts<br />

sulfate, zinc chloride, ferric chloride and manganese<br />

chloride) were separately in basal medium. The maximum<br />

EPS production (15.36 g/l) was obtained in sodium<br />

chloride (Figure 3). Besides magnesium chloride, others<br />

depress the EPS production. Based on the result we<br />

selected sodium chloride as our selection.<br />

Further, to find out the suitable concentration of NaCl<br />

for the EPS yield five different concentration (0.3, 0.4,<br />

0.5, 0.6 and 0.7%) sodium chloride were separately<br />

instead of 0.5% sodium chloride above.<br />

The maximum EPS production (15.36 g/l) was obtained<br />

when the concentration is 0.5% (Figure 3). The difference<br />

in EPS yield between the peptone concentrations of 0.5%<br />

and 0.6% was not significant because the cell could not<br />

absorb more mineral element. But considering the<br />

economic factor, we use the 0.5% NaCl as our optimal<br />

option.<br />

Effect of initial pH and temperature on EPS yield<br />

To find out the optimal temperature for and EPS<br />

production, B. thuringiensis 27 was cultivated at various<br />

temperatures ranging from 23 to 45°C. The EPS (12.12<br />

g/l) were observed at 28°C (Figure 4). B. thuringiensis 27<br />

was grown at initial pH 7.0 for polysaccharide production<br />

and optimum EPS production is 14.96 g/l) (Figure 4).


2362 Afr. J. Microbiol. Res.<br />

EPS Production (g/l)<br />

EPS Production (g/l)<br />

Figure 2. Effect of maltose and peptone concentration on EPS production.<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

(g/l)<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

FeCl 3<br />

10 20 30 40 50<br />

ZnCl 2<br />

Maltose Concentration (g/l)<br />

MnCl 2<br />

MgCl 2<br />

Inorganic ions<br />

CuSO 4<br />

Figure 3. Effect of inorganic ions and NaCl concentration on EPS production.<br />

Effect of inoculum size and liquid volume on EPS<br />

yield<br />

To examine the effect of inoculum size, B. thuringiensis<br />

27 was varying the inoculum size (2, 5, 8 and 11%) in<br />

basal medium. The results indicated 8% inoculums size<br />

NaCl<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

1 2<br />

5<br />

10<br />

Pepton Concentration (g/l)<br />

15<br />

3 4 5 6 7<br />

NaCl (g/l)<br />

was fit for EPS production (Figure 5).<br />

To find out the optimal oxygen rate four different culture<br />

medium volumes (25, 40, 55 and 70 ml) added in 200 ml<br />

flask were separately instead of 60 ml basal medium. The<br />

maximum EPS production (14.38 g/l) was obtained when<br />

the culture medium volume is 25 ml in 200 ml flask


(Figure 5).<br />

EPS Production (g/l)<br />

EPS Yield (g/L)<br />

Figure 4. Effect of initial pH value and temperature on EPS production.<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

16<br />

14<br />

12<br />

(g/L)<br />

10<br />

8<br />

6<br />

4<br />

5 6 7 8 9<br />

Initial pH value<br />

25 40 55 70<br />

Liquid Volume (ml/200ml flask)<br />

Figure 5. Effect of liquid volume and inoculum size on EPS production.<br />

Optimization by orthogonal matrix method<br />

According to the orthogonal method, the effect of those<br />

medium on EPS production was analyzed and the results<br />

are shown in Table 2. Based on the magnitude order of R<br />

value (maximum difference), the order of effects of all<br />

factors on EPS production was maltose > peptone ><br />

NaCl > beef extract. In terms of the maximum K-value of<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

23°C 28°C 33°C 38°C 43°C<br />

Temperature<br />

2 5 8 11<br />

Inoculum Size (%)<br />

Wang et al. 2363<br />

each column in Table 3, optimal level of each medium<br />

ingredient for EPS yield was 3 g/l beef extract glucose,<br />

10 g/l peptone, 40 g/l maltose, 4 g/l NaCl.<br />

Based on the magnitude order of R value (maximum<br />

difference), the order of effects of all factors on EPS yield<br />

was maltose >peptone >NaCl >beef extracts. According<br />

to ANOVA (Table 4), the factors, peptone and maltose<br />

had significant effect on EPS yield (P


2364 Afr. J. Microbiol. Res.<br />

Table 2. Results of L9 (34) orthogonal test of exopolysaccharide production of Bacillus strain 27 strain in shake flask culture.<br />

Run 1 2 3 4<br />

Factor Beef extract (g/L) Peptone (g/L) Maltose (g/L) NaCl (g/L) Results (g/L)<br />

1 2 5 20 4 5.63±0.2<br />

2 2 10 30 5 15.98±1.07<br />

3 2 15 40 6 13.24±0.73<br />

4 3 5 30 6 5.83±0.31<br />

5 3 10 40 4 18.08±2.26<br />

6 3 15 20 5 5.59±0.52<br />

7 4 05 40 5 10.54±2.21<br />

8 4 10 20 6 8.57±0.22<br />

9 4 15 30 4 13.48±0.2<br />

Values are means ± SD of triple determinations.<br />

Table 3. Analysis of media on exopolysaccharide production of Bacillus strain. 27 in shake flask culture with<br />

L9 (34) orthogonal test.<br />

Beef extract (g/l) Peptone (g/l) Maltose (g/l) NaCl (g/l)<br />

k1 11.61 7.33 6.59 12.39<br />

k2 9.83 14.21 11.76 10.70<br />

k3 10.86 10.77 13.953 9.21<br />

R 1.78 6.87 7.356 3.18<br />

Ki =Σ exopolysaccharide in thrice experiment at Xi. Values are means ±SD of triple determinations<br />

R = maximum Kix -minimum Kix. Values are means ± SD of triple determinations.<br />

Table 4. Variance analysis of L9 (34) orthogonal experiment on EPS yield.<br />

Sum of squares Degree of freedom Mean square Significance<br />

Beef extract 4.809 2 1.000<br />

Peptone 70.933 2 14.750 *<br />

Maltose 85.611 2 17.802 *<br />

NaCl 15.221 2 3.165<br />

Error 4.81 2<br />

inoculum size 8%, liquid volume 40 ml in 200 ml flask)<br />

was carried on. Consequently, the maximal EPS<br />

production was 20.17 g/l. The EPS yield on optimal<br />

condition was about five times than that at the basal<br />

culture condition.<br />

Bio-flocculating activity<br />

The flocculating activity of the EPS was measure by<br />

using a kaolin suspension. In order to neutralize the<br />

charge, one milliliter of 90 mmol/L CaCl2 were added into<br />

50 ml 4.0 g/l kaolin suspension. The optical density (OD)<br />

of the clarifying solution was measured with a<br />

spectrophotometer at 550 nm. After calculation the<br />

flocculating activity of B. thuringiensis 27 is 80.4%.<br />

DISCUSSION<br />

Carbon source plays a very important role in growth<br />

especially in polysaccharide production. A substantial<br />

change in the polysaccharide production was observed<br />

with different carbon sources. Maltose supported<br />

maximum EPS production which was difference with<br />

other bacillus (Lee et al., 1997a). The difference in EPS<br />

production among the maltose concentration of 3, 4 and<br />

5% was not significant because the cell maybe not<br />

absorb more maltose. It also is report that see that the<br />

EPS production increased as the initial carbon resource<br />

concentration increased from 20 to 100 g/l (Lee et al.,<br />

1997b). Nitrogen sources in the form of proteins and<br />

nucleic acid play an important role in the cell mass.<br />

Peptone as organic nitrogen source is the best nitrogen


source in our work. Additionally, it was found in the study<br />

that organic nitrogen source are more suitable for the<br />

EPS production. It was reported that organic nitrogen<br />

sources were absorbed by the cells easier than the<br />

inorganic ones (Hwang et al., 2003; Gandhi et al., 1998).<br />

Inorganic ions affected EPS production by combined with<br />

enzyme. The result showed NaCl could provide the<br />

maximum EPS production, which did not match those of<br />

other researcher (Lung and Huang, 2010).<br />

Optimum pH for polysaccharide production for bacteria<br />

ranges from 6.0 to 7.5 (Kumar et al., 2007), which exactly<br />

accorded with those of B. thuringiensis 27. Optimum EPS<br />

production (14.96 g/l) was obtained in the neutral pH<br />

range. The maximum EPS production by B. thuringiensis<br />

27 were observed at 28°C, which is comparable to many<br />

kinds of bacillus that have relatively low temperature<br />

optimal (example 20 to 25°C) in their submerged<br />

cultures(Cerning et al., 1992). Among several bacteria<br />

physiological properties inoculum size may play an<br />

important role in biological development (Gancel and<br />

Novel, 1994). Inoculum size and liquid volume may play<br />

an important role in cell reproduction and EPS production<br />

(Chen et al., 2008). Result showed 8% inoculum size and<br />

25 ml in 200 ml flasks was fit for EPS production.<br />

From the single factors experiments the three critical<br />

factors affect the EPS yield were identified. Considered<br />

beef extract as an important nitrogen source it is selected<br />

to further optimize. To investigate the relationship among<br />

various factors the orthogonal experimental design<br />

technique as a mathematical method were used the<br />

orthogonal matrix method was obviously a serviceable<br />

experimental design to simultaneously investigate the<br />

relationship between the effect of medium components<br />

and their optimal concentrations (Gundlapally and<br />

Garcia-Pichel, 2006). In addition, the orthogonal matrix<br />

can comprehensively investigate central composite<br />

design, and thus facilitates economical benefit, experimental<br />

convenience. In our work by orthogonal method<br />

the EPS production arrived to 20.17 g/l, which is not only<br />

about five times than those of the basal medium but more<br />

than those of the maximum result of orthogonal design.<br />

That is means the orthogonal matrix method can be used<br />

for optimizing the EPS production in a submerged<br />

fermentation process. Furthermore it is applied to<br />

optimization of culture media for the production of primary<br />

and secondary metabolites in fermentation processes<br />

(Chen et al., 2008).<br />

Flocculation technology as a kind of effective and quick<br />

method is used for wastewater treatment. And bioflocculation<br />

is a dynamic process resulting from the synthesis<br />

of extracellular polymers by living cells. In recent years,<br />

the use of microbial flocculants has been promoted as a<br />

solution to environmental problems because their<br />

intermediates are harmless and biodegradable. Thus<br />

searching and usage bioflocculation arouse the<br />

interesting of researcher in the world. The flocculating<br />

activity of 27 is 80.4% which is below the bioflocculant p-<br />

KG03 (Yim et al., 2007). To obtain new and better<br />

Wang et al. 2365<br />

bioflocculant, we need screen more strain to obtain<br />

functional EPS in BSCs. Otherwise new usage for<br />

microbiology in the desert and desert biological soil<br />

crusts is proved a scientific basis.<br />

Conclusion<br />

The traditional separation, screening and purification<br />

methods were used, 27 strains of producing EPS were<br />

isolated. Under optimal condition the strain could produce<br />

a large amount of EPS. And the EPS possess a certain<br />

degree of flocculation, the flocculation rate was 80.4%,<br />

which is lower than the previously reported strain isolated<br />

from the soil (flocculation rate can reach 97%). Further<br />

screening and optimization to increase the flocculation<br />

rate still carry on. It is well known that microbial<br />

flocculants as a kind of sewage treatment agent are nontoxic,<br />

harmless, no secondary pollution. In this subject,<br />

the discovery of new sources of microbial flocculant has<br />

brought new interest in dealing with the pollution of water<br />

resource. And new usage for the desert biological soil<br />

crusts is provided a scientific basis.<br />

ACKNOWLEDGEMENT<br />

This work was supported by the Commonweal Industry<br />

Scientific Researcher Project from the Ministry of<br />

Agriculture (No. 200803033).<br />

REFERENCES<br />

Cerning J, Bouillanne C, Landon M, Desmazeaud M (1992). Isolation<br />

and characterization of exopolysaccharides from slime-forming<br />

mesophilic lactic acid bacteria. J. Dairy Sci., 75:692.<br />

Chen W, Zhao Z, Chen SF, Li YQ (2008). Optimization for the<br />

production of exopolysaccharide from Fomes fomentarius in<br />

submerged culture and its antitumor effect in vitro. Bioresour.<br />

Technol., 99: 3187-3194.<br />

Di X, Chan K, Leung HW, Huie CW (2003). Fingerprint profiling of acid<br />

hydrolyzates of polysaccharides extracted from the fruiting bodies<br />

and spores of Lingzhi by high-performance thin-layer<br />

chromatography. J. Chromatogr. A., 1018: 85-95.<br />

Gancel F, Novel G (1994). Exopolysaccharide production by<br />

Streptococcus salivarius ssp. thermophilus cultures. 1. Conditions of<br />

production. J. Dairy Sci., 77: 685-688.<br />

Gandhi HP, Ray RM, Patel RM (1998). Exopolymer production by<br />

Bacillus species. Carbohydr. Polym., 34:323-327.<br />

Gundlapally SR, Garcia-Pichel F (2006). The community and<br />

phylogenetic diversity of biological soil crusts in the Colorado Plateau<br />

studied by molecular fingerprinting and intensive cultivation. Microb.<br />

Ecol., 52: 345-357.<br />

Hao LM, Xing XH, Li Z, Zhang JC, Sun JX, Jia SR, Qiao CS, Wu T<br />

(2010). Optimization of effect factors for mycelial growth and<br />

exopolysaccharide production by Schizophyllum commune. Appl.<br />

Biochem. Biotechnol., 160: 621-631.<br />

Hwang HJ, Kim SW, Choi JW, Yun JW (2003). Production and<br />

characterization of exopolysaccharides from submerged culture of<br />

Phellinus linteus KCTC 6190. Enzyme Microb. Technol., 33: 309-319.<br />

Kaditzky S, Vogel RF (2008). Optimization of exopolysaccharide yields<br />

in sourdoughs fermented by lactobacilli. Eur. Food Res. Technol.,<br />

228: 291-299.


2366 Afr. J. Microbiol. Res.<br />

Kennedy SP, Ng WV, Salzberg SL, Hood L, Dassarma S (2001).<br />

Understanding the adaptation of Halobacterium species NRC-1 to its<br />

extreme environment through computational analysis of its genome<br />

sequence. Genome Res., 11: 1641.<br />

Kumar AS, Mody K, Jha B (2007). Bacterial exopolysaccharides - A<br />

perception. J. Basic Microbiol., 47: 103-117.<br />

Kurane R, Takeda K, Suzuki T (1986). Screening for and characteristics<br />

of microbial flocculants. Agric. Biol. Chem., 50: 2301-2307.<br />

Lee IY, Seo WT, Kim GJ, Kim MK, Ahn SG, Kwon GS, Park YH (1997a).<br />

Optimization of fermentation conditions for production of<br />

exopolysaccharide by Bacillus polymyxa. Bioprocess Eng., 16: 71-75.<br />

Lee IY, Seo WT, Kim GJ, Kim MK, Ahn SG, Kwon GS, Park YH (1997b).<br />

Optimization of fermentation conditions for production of<br />

exopolysaccharide by Bacillus polymyxa. Bioprocess Eng., 16: 71-75.<br />

Li Y, Chen J, Lun SY, Rui XS (2001). Efficient pyruvate production by a<br />

multi-vitamin auxotroph of Torulopsis glabrata: key role and<br />

optimization of vitamin levels. Appl. Microbiol. Biotechnol., 55: 680-<br />

685.<br />

Lung MY, Huang PC (2010). Optimization of exopolysaccharide<br />

production from Armillaria mellea in submerged cultures. Lett. Appl.<br />

Microbiol., 50: 198-204.<br />

Nicolaus B, Moriello VS, Lama L, Poli A, Gambacorta A (2004).<br />

Polysaccharides from extremophilic microorganisms. Orig. Life Evol.<br />

Biosph., 34: 159-169.<br />

Pazur JH, Chaplin MF, Kennedy JF (1994). Em Carbohydrate Analysis:<br />

A Practical Approach. New York: IRL, pp. 93-100.<br />

Xu CP, Kim SW, Hwang HJ, Choi JW, Yun JW (2003). Optimization of<br />

submerged culture conditions for mycelial growth and exo-biopolymer<br />

production by Paecilomyces tenuipes C240. Process Biochem., 38:<br />

1025-1030.<br />

Xu R, Ma S, Wang Y, Liu L, Li P (2010). Screening, identification and<br />

statistic optimization of a novel exopolysaccharide producing<br />

Lactobacillus paracasei HCT. Afr. J. Microbiol. Res., 4: 783-795.<br />

Yim JH, Kim SJ, Ahn SH, Lee HK (2007). Characterization of a novel<br />

bioflocculant, p-KG03, from a marine dinoflagellate, Gyrodinium<br />

impudicum KG03. Bioresour. Technol., 98: 361-367.


African Journal of Microbiology Research Vol. 5(16), pp. 2367-2373, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.893<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

A simple and efficient method that uses low<br />

concentration fetal bovine serum to culture and purify<br />

Schwann cells<br />

Han Feng, Qu Wei*, Jiang Huajun, Fu Chongyang, Lu Ming, Zhang Weiguo and Lv Decheng<br />

Department of Orthopaedic Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.<br />

Accepted 18 April, 2011<br />

To develop a simple and efficient method for the isolation of highly purified Schwann cells (SCs) from<br />

the sciatic nerves of newborn rats, cells were initially cultured in 10% FBS for 6 days, and then cultured<br />

in 2% FBS for 6 days. And we used the immunocytochemical characterization, flow cytometry, RT-PCR,<br />

to observe the condition of SCs. After the 12-day treatment protocol, we obtained a high yield of SCs<br />

with an average purity of 96.9%. Furthermore, the condition of SCs had no change. In our purification<br />

protocol, we had shown that our procedures did not alter the cell cycle and neurokines’ secretion. Our<br />

simple and efficient method yielded pure and healthy SCs and did not require treatment with antimitotic<br />

agents or any special equipment.<br />

Key words: Schwann cells, cell culture, low fetal bovine serum concentration, tissue engineering, fibroblast.<br />

INTRODUCTION<br />

Tissue engineering, the use of cells and engineering<br />

methods to modify biological functions, has already been<br />

applied to diverse clinical fields. In neurology, tissue<br />

engineering appears to have certain advantages over<br />

nerve cell transplantation. There are three critical<br />

meterials of tissue engineering: (i) biodegradable<br />

materials; (ii) trophic factors; and (iii) cells, typically<br />

autologous cells that do not cause antigenic reactions<br />

(Mackinnon and Dellon, 1990; Vacanti et al., 2001). In<br />

peripheral nerve tissue engineering, it is critical to rapidly<br />

obtain large numbers of pure Schwann cells (SCs)<br />

(Fansa and Keilhoff, 2004). SCs are the principal<br />

supporting cells of the peripheral nervous system, and<br />

are crucially involved in the functional recovery of injured<br />

peripheral nerves (Bunge, 1993). SCs play an important<br />

role in nerve regeneration and are at the core of<br />

peripheral nerve tissue engineering, a procedure that<br />

requires a large number of cells. Diverse methods have<br />

been proposed for the purification of SCs: antimitotic<br />

agent and antibody-mediated cytolysis methods (Brockes<br />

*Corresponding author. E-mail: fhtg001@sina.com.<br />

et al., 1979); repeated explantation methods (Oda et al.,<br />

1989); the cold jet technique (Jirsová et al., 1997);<br />

immunoselective methods (Calderon-Martinez et al.,<br />

2002); in vivo or in vitro predegeneration methods<br />

(Komiyama et al., 2003; Mauritz et al., 2004); differential<br />

adhesion methods (Pannuzio et al., 2005); differential<br />

detachment methods (Jin et al., 2008); and a combination<br />

of the in vitro predegeneration and cold jet technique<br />

(Haastert et al., 2007; 2009). These methods can yield<br />

highly purified SCs with various efficiencies, but all<br />

require special equipments, complicated procedures<br />

and/or high costs.<br />

Preparation of SCs for tissue engineering requires a<br />

safe, rapid, easy and high yield methodology for<br />

purification. We aimed to develop an economical method<br />

besides those requirements from 7 days old Sprague<br />

Dawley (SD) rats by use of low concentration FBS.<br />

MATERIALS AND METHODS<br />

Animals<br />

Postnatal SD rats (7 days old, 11.3±0.5 g) were obtained from the<br />

Dalian Medical University Animal Center (Dalian, China). All animal


2368 Afr. J. Microbiol. Res.<br />

protocols were approved by the Animal Experiment and Care<br />

Committee of Dalian Medical University.<br />

Primary culture<br />

Rats were sacrificed by decapitation, and sciatic nerve segments<br />

(10 to 15 mm in length) were harvested aseptically under a<br />

dissecting microscope and were temporarily maintained in 60 mm<br />

culture plates (Corning, USA). Then, 2 ml of ice-cold D-Hanks<br />

solution (Sigma USA) was added, and the sciatic nerve segments<br />

were removed from the epineurium and cut into pieces (1 mm 3 )<br />

under a dissecting microscope. Finally, nerve fragments were<br />

placed in 15 mL conical centrifuge tubes (Corning, USA) and<br />

centrifuged (140 g × 5 mins). The supernatant was discarded. 1 mL<br />

each of 0.25% trypsogen (Gibco, USA) and collagen 0.2% type- II<br />

(Gibco, USA) were added, then incubated in a cell incubator<br />

(Thermo, USA) with 5% carbon dioxide at 37°C for 30 min and heat<br />

shocked once every 5 mins. Addition of 2 mL of Dulbecco’s<br />

Minimum Eagle’s Medium (DMEM) (Hyclone, USA) that contained<br />

10% FBS (Gibco, USA). The mixture was centrifuged again (140 g<br />

× 5 min), and the supernatant was again discarded. Removed<br />

nerve fragments to 60 mm culture plates. Then, 1.5 mL<br />

DME/10%FBS containing 50 U/ml penicillin and 50 µg/ml<br />

streptomycin (Gibco, USA) was added, and all plates were<br />

maintained in a cell incubator within a humidified atmosphere with<br />

5% CO2 at 37°C. The growth medium was changed every 48 h.<br />

Purification of SCs<br />

Cells isolated from nerve segments after the first 6 days were<br />

divided into three groups: Group A (control) were fed every 2 days<br />

using DMEM with 10% FBS; Group B were fed every 2 days using<br />

DMEM with 2% FBS and cells were collected on day 6; Group C<br />

were fed every 2 days using serum-free DMEM, and cells were<br />

collected on day 6.<br />

Immunocytochemical characterization of SCs and SC purity<br />

Immunohistochemical staining was accomplished in the culture dish<br />

after 12 days. Cells were marked with the Strept Avidin-Biotin<br />

Complex (SABC) kit and rabbit anti-S100 protein antibody (Boster,<br />

China) according to the manufacturer’s instructions. Then, cells<br />

were washed two times with PBS (pH 7.2) (Gibco, USA), fixed for<br />

90 mins in 4% paraformaldehyde, and then immersed in a mixture<br />

of 30% H2O2 and pure methanol (1:50) for 30 mins. After that, 5%<br />

BSA (confining liquid) was added drop-wise at room temperature in<br />

20 mins. BSA was shaken out of the dish. Then anti-S100 protein<br />

antibody was added drop-wise at 37°C for 1 h, washed 3 times with<br />

PBS, incubated with secondary antibodies at 37°C for 20 mins,<br />

washed 2 times with PBS. Then, SABC was added drop-wise at<br />

37°C for 20 mins, washed 4 times with PBS. Finally, cells were<br />

stained by DBA (Hsu and Ree, 1980; Klosen, 1989) using a kit<br />

(Boster, China), at room temperature for 8 min, then post-stained<br />

with hematoxylin and examined under a microscope. Cells with a<br />

bipolar or tripolar shapes were identified as SCs, whereas flat or<br />

polygonal cells were identified as fibroblasts (Pannuzio et al.,<br />

2005).<br />

SC purity was expressed as the percentage of SCs (SC purity =<br />

[number of SCs]/[number of SCs + number of fibroblasts]) from<br />

samples of ten visual fields at 200 × magnification.<br />

Flow cytometry<br />

Cells from Groups A and B were digested with 0.25% trypsogen and<br />

0.02% EDTA, treated into the single cell suspension, centrifuged<br />

(314 g × 5 min) and washed with PBS. Then, centrifuged again, and<br />

the precipitate were resuspended and fixed in 2 mL of cold 80%<br />

reagent grade ethanol and left overnight at 4°C. Then, centrifuged<br />

and used 0.01% RNAase (Sigma, USA) to resuspend the<br />

precipitate and heat shocked at 37°C for 10 min. The cells were<br />

centrifuged and0.5%PI (propidium iodide) (Sigma, USA) was added<br />

to the precipitate and cell concentration was adjusted to 1 × 10 6 per<br />

mL.<br />

Finally, the cells were stained in darkness for 30 min and<br />

detected by flow cytometry.<br />

Reverse transcriptase polymerase chain reaction (RT-PCR)<br />

Total RNA was extracted separately from Groups A and B after 12<br />

days and cDNA was synthesized from the total RNA using a RNA<br />

PCR Kit (AMV) Ver.3.0 (Takara, China) according to the<br />

instructions. The sequences of the primers for NGFb (Yan et al.,<br />

2007), BDNF (Yan et al., 2007), β-actin are depicted in Table 1. The<br />

PCR conditions were 94°C for 30s (denaturation), 56°C for 30 s<br />

(annealing) and 72°C for 1 min (extension) for a total of 30 cycles<br />

(NGFb, BDNF), or 55°C for 30 s (annealing) for β-actin. And a<br />

DL2000 DNA-ladder (TaKaRa, Japan) was run in parallel to the<br />

samples. Amplification products were separated by 2.0% agarose<br />

gel electrophoresis. After the gels were scanned, the relative<br />

intensity of NGFb and BDNF bands was determined by using Gelpro<br />

Analyzer software (Media Cybernetics, USA).<br />

Statistics<br />

SC purity and cell yields are presented as means ± SDs. Values<br />

were subjected to a Student’s t-test, with a P value less than 0.05<br />

considered to be statistically significant.<br />

RESULTS<br />

Primary culture and purification of SCs<br />

During the first 48 h of the primary culture, two distinct<br />

types of cells split off from the nerve fragments (Figure<br />

1a). Phase-contrast microscopy (× 200) indicated that<br />

most of the cells attached to the bottom of the culture<br />

dishes could be classified as SCs (spindle-shaped,<br />

bipolar, and sometimes tripolar, with a small cytoplasmto-nucleus<br />

ratio) or as fibroblasts (flat polygonal shaped,<br />

with a prominent ovoid nucleus and abundant cytoplasm).<br />

Most of the fibroblasts appeared to be scattered among<br />

the SCs, but some were beneath the SCs. During the first<br />

48 h, SCs proliferated faster than fibroblasts. However,<br />

96 h later, fibroblasts gradually became the dominant cell<br />

type, and SCs stopped growing and migrated onto<br />

fibroblasts. SCs and fibroblasts were both growing rapidly<br />

after 4 days.<br />

After 6 days of primary culture of Group A cells (DMEM<br />

with 10% FBS), most of the fibroblasts growing among<br />

the SCs were easily detached from the culture plates,<br />

whereas those growing under or interacting with SCs<br />

could not be removed (Figure 1b). For group B cells<br />

(DMEM with 2% FBS), the fibroblasts started to die after<br />

48 h, quite a number of cell were gone after 96 h of


Table 1. The sequences of the primers for NGFb, BDNF, β-actin.<br />

Feng et al. 2369<br />

Forward primer Reverse primer Ta (°C) Amplicon length (bp)<br />

NGFb 5'-GGCCACTCTGAGGTGCATAG-3' 5'-CATGGGCCTGGAAGTCTAAA-3' 56 349<br />

BDNF 5'-AAACCATAAGGACGCGGACT-3’ 5'-GATTGGGTAGTTCGGCATTG-3' 56 393<br />

β-actin 5’-TCTACGAGGGCTATGCTCTCC-3’ 5-GGATGCCACAGGATTCCATAC-3’ 55 320<br />

Figure 1. Observe the results of the growing cells with inverted microscope. A as SC, B as fibroblast, C as nerve<br />

segment and D as dead cells. During the 2nd day of the primary culture, two distinct types of cells emerged from<br />

the nerve fragments (a). And, at the 6th day of the primary culture, SCs and fibroblasts were growing fast (b).<br />

However, used DMEM with 2% FBS, numerous fibroblasts died within 96 h; (c) and almost all of the fibroblasts<br />

were cleared away by the 6th day and SCs morphology was not changed with 2% FBS (d). But after 6 days<br />

barely any cells survived at the culture plate in Group C. scale bar: 100 um).<br />

culturing (Figure 1c), and almost all fibroblasts were gone<br />

after 6 days of culturing. The morphology of SCs did not<br />

change during this time (Figure 1d). For group C cells<br />

(serum-free DMEM), a few cells died after 48 h, but after<br />

96 h, both types of cells had died in significant numbers.<br />

The death rate continued to increase, and after 6 days<br />

of culturing, almost all cells had died (Figure 1e).<br />

Immunocytochemical characterization of SC and SC<br />

purity<br />

S-100 is a well-established sensitive and specific marker<br />

for SCs (Raff et al., 1979). We observed S-100 staining in<br />

the SCs body and along the processes (Figure 2). Our<br />

purified SCs had typical bi- or tri-polar morphology and<br />

oval nuclei. In contrast, the contaminating fibroblasts<br />

were negative for S-100 staining. Because our<br />

immunological staining was performed in culture dishes,<br />

we had acquired very high yield and high-quality cells.<br />

Since so many of the Group C cells died, we could not<br />

collect enough cells for immunocytochemical staining.<br />

Table 2 shows the percentage of cells positive for S-100.<br />

SC purity in Group B was significantly better than in<br />

Group A (P < 0.01). However, the total numbers of SCs<br />

were too little to discriminate Group A from B (p > 0.05).<br />

Flow cytometry<br />

To further provide qualitative evidences of the characters<br />

of our SCs, we compare the SC cycle of Groups A and B<br />

cells (Figure 3). As shown in Table 3, there was no<br />

significant difference in the cells cycles between Groups


2370 Afr. J. Microbiol. Res.<br />

A and B cells (p > 0.05).<br />

Figure 2. Immunocytochemical characterization of SCs with<br />

S100 showed that the purified SCs had typical bi- or tri-polar<br />

morphology and oval nuclei. (a) In contrast, the contaminating<br />

fibroblasts were negative for S-100 staining (b). Used DMEM with<br />

10% FBS, numerous fibroblasts are shown at (A). Used DMEM<br />

with 2% FBS (B), by contrast, cells were very purified (scale bar:<br />

100 um).<br />

Table 2. Summary of results in the present method for SCs preparation.<br />

View Number of S100 protein positive cells/total number of counted cells<br />

Group A Group B<br />

1 646*/753 (0.857902)** 779*/824 (0.945388)**<br />

2 988*/1173 (0.842285)** 1045*/1094(0.955210)**<br />

3 1111*(/12340.900324)** 1368*/1415(0.966784)**<br />

4 845*/978 (0.864008)** 722*/746 (0.967828)**<br />

5 750*/849 (0.883392)** 901*/936 (0.962607)**<br />

6 731*/862 (0.848028)** 1711*/1752(0.976598)**<br />

7 1130*/1270(0.889764)** 749*/762 (0.982940)**<br />

8 1083*/1210(0.895041)** 939*/956 (0.982218)**<br />

9 1092*/1206(0.905473)** 839*/860 (0.975581)**<br />

10 592*/738 (0.802168)** 836*/856 (0.976636)**<br />

* There was an insignificant difference between SCs total at Groups A and B (p>0.05); **There was a<br />

significant difference between SC purity at Groups A and B (p


DISCUSSION<br />

Main findings<br />

Feng et al. 2371<br />

Figure 3. The cell cycle of SCs detected by flow cytometer showed that used DMEM with 10%FBS(A). DIP<br />

G2:0.53%, S:47.37%, and used DMEM with 2%FBS(B) DIP G2:0.10%, S:44.92%.<br />

Table 3. Summary of results in the present method for SCs cycle.<br />

Cell cycle<br />

Group A<br />

Group B<br />

G2 a (%) S b (%) G2 a (%) S b (%)<br />

0.53 47.37 0.10 44.92<br />

0.37 45.44 0.68 48.23<br />

0.13 44.80 0.42 46.04<br />

a There was a insignificant difference between G2 percentage at Groups A and B (p>0.05); b There was a<br />

insignificant difference between S percentage at Groups A and B (p>0.05).<br />

Our method of culturing the sciatic nerves of newborn SD<br />

rats in DMEM with 10% FBS allowed SCs and fibroblasts<br />

to split off from intact nerve fragments. Once SCs have<br />

migrated in high densities, they proliferate actively due to<br />

the production of autocrine growth factors, IGF-2, NT-3<br />

and PDGF-BB (Jessen and Mirsky, 1999), regardless of<br />

being cultured in a medium with low FBS concentration.<br />

However, cells in serum-free DMEM (Group C) did not<br />

have adequate nutrition for growth and proliferation.<br />

Thus, although there were few fibroblasts in Group C, the<br />

growth of SCs was also greatly affected. There was no<br />

significant difference in the number of SCs in Groups A<br />

and B, but the SC purity was significantly greater in<br />

Group B. Our results indicate that culturing of nerve cells<br />

in DMEM with 2% FBS can gradually reduce the<br />

percentage of fibroblasts, while having no effect on SC<br />

differentiation, morphology or character.<br />

The immuno staining results indicated that staining was<br />

evident in both SC cell body and processes during the<br />

culture period. The flow cytometry results indicated that<br />

the number of cells in S period and G2 period were similar<br />

in Groups A and B. NGFb and BDNF are the main growth<br />

factors secreted by SCs, which are known to have<br />

neurotrophic effects on nerve regeneration (Schicho et<br />

al., 1999; Serpe et al., 2005).<br />

Semi-quantitative analysis of NGF mRNA and BDNF<br />

mRNA by RT–PCR showed that SCs of Group B had<br />

similar productive function of secretion the normal<br />

cultured SCs. Thus, we conclude that our culturing<br />

protocol is a simple and efficient method to purify SCs<br />

that also preserves the function and morphology of SCs.<br />

Comparison with the existing literature<br />

Gross dissection of nerve cells may lead to contamination<br />

of the desired cells (Jessen and Mirsky, 1999) and<br />

peripheral nerve regeneration with tissue-engineered<br />

cells requires highly purified SCs for the construction of<br />

artificial nerve grafts. Many previous studies have


2372 Afr. J. Microbiol. Res.<br />

500bp<br />

250bp<br />

500bp<br />

250bp<br />

M<br />

5<br />

6<br />

M 1 2<br />

3 4<br />

Figure 4. Total RNA was analyzed for the expression of NGFb and BDNF by RT–PCR at 12 days.1: BDNF<br />

(Group A); 2 BDNF (Group B); 3: NGFb (Group A); 4::NGFb (Group B); 5: β-actin(Group A);6: β-actin(Group B);<br />

M: Marker.<br />

attempted to reduce the fibroblast contamination and<br />

increase SC purity by use of: (i) antimitotic treatment<br />

(Schicho et al., 1999); (ii) a combination of antimitotic<br />

treatment and antibody-mediated cytolysis with complements<br />

(Brockes et al., 1979); (iii) repeated explantation<br />

methods 6 (Serpe et al., 2005; Smith and Greenfield,<br />

2003); (iv) differential adhesion methods (Pannuzio et al.,<br />

2005; Wood, 1976); and (v) immunoselective methods<br />

(Manent et al., 2003; Askanas et al., 1980; Wrathall et<br />

al., 1981). Those previously proposed methods have<br />

been shown to enrich SCs with various efficiencies, but<br />

all have certain limitations. In particular, antimitotic agents<br />

are harmful to SC function (Serpe et al., 2005; Assouline<br />

et al., 1983) and can reduce SC yield because of their<br />

non-specific antimitotic effects. Antibodies and<br />

complements are expensive for large-scale SC<br />

preparations, so do not provide an economical approach<br />

to SC purification. Repeated explantation and<br />

differential adhesion can be comparatively complicated<br />

and time-consuming procedures, and may lead to a loss<br />

of SCs and delay of therapy. Immunoselection is a good<br />

method for achieving high purity, but requires expensive<br />

antibodies and special facilities.<br />

In a previous study, Needham et al. (1987), described<br />

the use of serum-free medium (S4) that allowed for<br />

optimal SC proliferation (up to 90% purity after 10 days),<br />

and suppressed fibroblast overgrowth in primary cultures<br />

of neonatal rat sciatic nerves (Assouline et al., 1983). But<br />

this serum-free media, though suitable for selective SC<br />

migration, was not as conducive for SC proliferation.<br />

However, use of a growth medium with 10% FBS<br />

supported not SC proliferation but an explosive fibroblast<br />

overgrowth. Komiyama et al. (2003) described the use of<br />

low FBS concentration for the purification of SCs, and<br />

obtained an average purity of 98% after 8 days by using<br />

DMEM for the first 6 days and changing to DMEM with<br />

2.5% FBS on day 6. However, this method yielded low<br />

cell counts, and the resulting SC purity was only 88.0%.


Another method described by Komiyama et al. (2003)<br />

cultured cells in DMEM with 10% FBS for the first 3 days,<br />

then in serum-free DMEM for 3 days, then, on day 6 in<br />

DMEM with 2.5% FBS. The SC purity was 93.6% at 14<br />

days, but was reduced to 89.5% at 21 days. Thus,<br />

regardless of method, the total cell counts were<br />

increasing, but the percentage of SCs decreased over<br />

time.<br />

This phenomenon was also noted in our study.<br />

IMPLICATIONS FOR FUTURE RESEARCH<br />

The protocol described here is a simple, economical,<br />

rapid and efficient method for the purification of SCs. Our<br />

method has a high yields and results in highly pure cells.<br />

We suggest that our method might be useful for the<br />

development of tissue-engineered nerves and for other<br />

studies related to peripheral nerve injuries.<br />

ACKNOWLEDGEMENT<br />

This research was supported by National Natural Science<br />

Foundation of China (30973066).<br />

REFERENCES<br />

Askanas V, EngelWK, Dalakas MC, Lawrence JV, Carter LS (1980).<br />

Human Schwann cells in tissue culture: histochemical and<br />

ultrastructural studies. Arch Neurol., 6: 329–337.<br />

Assouline JG, Bosch EP, Lim R (1983). Purification of rat Schwann cells<br />

from cultures of peripheral nerve: an immunoselective method using<br />

surfaces coated with anti-immunoglobulin antibodies. Brain Res., 2:<br />

389–9.<br />

Brockes JP, Fields KL, Raff MC (1979). Studies on cultured rat Schwann<br />

cells. I. Establishment of purified populations from cultures of<br />

peripheral nerve. Brain Res., 1: 105–118.<br />

Bunge RP (1993). Expanding roles for the Schwann cell: ensheathment,<br />

myelination, trophism and regeneration. Curr. Opin. Neurobiol., 3:<br />

805–809.<br />

Calderon-Martinez D, Garavito Z, Spinel C, Hurtado H (2002). Schwann<br />

cell-enriched cultures from adult human peripheral nerve: a technique<br />

combining short enzymatic dissociation and treatment with cytosine<br />

arabinoside (Ara-C). J. Neurosci. Methods, 114: 1–8.<br />

Fansa H, Keilhoff G (2004). Comparison of different biogenic matrices<br />

seeded with cultured Schwann cells for bridging peripheral nerve<br />

defects. Neurol. Res., 26(2): 167-173.<br />

Haastert K, Mauritz C, Chaturvedi S, Grothe C (2007). Human and rat<br />

adult Schwann cell cultures: fast and efficient enrichment and highly<br />

effective non-viral transfection protocol. Nat. Protoc., 2: 99–104<br />

Haastert K, Seef P, Stein VM, Tipold A, Grothe C (2009). A new cell<br />

culture protocol for enrichment and genetic modification of adult<br />

canine Schwann cells suitable for peripheral nerve tissue<br />

engineering. Res. Vet. Sci., 87: 140–142.<br />

Feng et al. 2373<br />

Hsu S, Ree H (1980). Self-sandwich method: an improved<br />

immunoperoxidase technique for the detection of small amounts of<br />

antigens. Am. J. Clin. Pathol., 74: 32–40.<br />

Jessen KR, Mirsky R (1999). Why do Schwann cells survive in the<br />

absence of axons. Ann New York Acad. Sci., 883: 109-115.<br />

Jin YQ, Liu W, Hong TH, Cao YL (2008). Efficient Schwann cell<br />

purification by differential cell detachment using multiplex collagenase<br />

treatment. J. Neurosci. Methods, 170: 140–148.<br />

Jirsová K, Sodaar P, Mandys V, Bär PR (1997). Cold jet: a method to<br />

obtain pure Schwann cell cultures without the need for cytotoxic,<br />

apoptosis-inducing drug treatment. J. Neurosci. Methods, 78: 133–<br />

137.<br />

Klosen P (1989). Techniques Immunocytochimiques. Louvain:<br />

Universite ´Catholique de Louvain, pp. 24–44.<br />

Komiyama T, Nakao Y, Toyam Y, Asou H, Vacanti CA, Vacanti MP<br />

(2003). Anovel technique to isolate adult Schwann cells for an<br />

artificial nerve conduit. J. Neurosci. Methods, 122: 195-200.<br />

Mackinnon SE, Dellon AL (1990). Clinical nerve reconstruction with a<br />

bioabsorbable polyglycolic acid tube. Plast Reconstr. Surg., 85(3):<br />

419-24.<br />

Manent J, Oguievetskaia K, Bayer J, Ratner N, Giovannini M (2003).<br />

Magnetic cell sorting for enriching Schwann cells from adult mouse<br />

peripheral nerves. J. Neurosci. Methods, 2: 167–173.<br />

Mauritz C, Grothe C, Haaster K (2004). Comparative study of cell<br />

culture and purification methods to obtain highly enriched cultures of<br />

proliferating adult rat Schwann cells. J. Neurosci. Res., 77: 453–461<br />

Needham LK, Tennekoon GI, McKhann GM (1987). Selective growth of<br />

rat Schwann cells in neuron- and serum-free primary culture. J.<br />

Neurosci., 7(1): 1-9.Oda Y, Okada Y, Katsuda S, Ikeda K, Nakanishi I<br />

(1989). A simple method for the Schwann cell preparation from<br />

newborn rat sciatic nerves. J. Neurosci. Methods, 3: 163–169.<br />

Pannuzio ME, Jou IM, Long A, Wind TC, Beck G, Balian G (2005). A<br />

new method of selecting Schwann cells from adult mouse sciatic<br />

nerve. J. Neurosci. Method, 1: 74–81.<br />

Raff MC, Fields KL, Hakamori S, Mirski R, Pruss RM, Winter J (1979).<br />

Cell-type-specific markers for distinguishing and studying neurons<br />

and the major classes of glial cells in culture. Brain Res., 174: 283–<br />

308.<br />

Schicho R, Skofitsch G, Donnerer J (1999). Regenerative effect of<br />

human recombinant NGF on capsaicin-lesioned sensory neurons in<br />

the adult rat. Brain Res., 815: 60–69.<br />

Serpe CJ, Byram SC, Sanders VM, Jones KJ (2005). Brain-derived<br />

neurotrophic factor supports facial motoneuron survival after facial<br />

nerve transection in immunodeficient mice. Brain Behav. Immun., 19:<br />

173–180.<br />

Smith L, Greenfield A (2003). DNA microarrays and development. Hum<br />

Mol. Genet ., 12: R1–R8.<br />

Vacanti CA, Bonassar LJ, Vacanti MP, Burger JS (2001) . Replacement<br />

of an avulsed phalanx with tissue-engineered bone. New Engl. J.<br />

Med., 344(20): 1511-1514.<br />

Wood PM (1976). Separation of functional Schwann cells and neurons<br />

from normal peripheral nerve tissue. Brain Res., 3: 361–75.<br />

Wrathall JR, Rigamonti DD, Braford MR, Kao CC (1981). Non-neuronal<br />

cell cultures from dorsal root ganglia of the adult cat: production of<br />

Schwann-like cell lines. Brain Res., 1: 163–81.<br />

Yan S, BingFang Z, ChangQing Z, Kai-Gang Z, XueTao X (2007). Study<br />

of biocompatibility of small intestinal submucosa (SIS) with Schwann<br />

cells in vitro. Brain Res., 1145: 41-47.


African Journal of Microbiology Research Vol. 5(16), pp. 2374-2379, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR09.455<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

First report on Enterobacter sakazakii from Sudanese<br />

patients<br />

Humodi A. Saeed¹* and Rania M. Musallam²<br />

Department of Microbiology, College of Medical laboratory Science, Sudan University of Science and Technology,<br />

Arbab El Agaid Street, P. O. Box 407 Khartoum, Sudan.<br />

Accepted 12 March, 2010<br />

Enterobacter sakazakii (E. sakazakii) has been identified as emerging opportunistic pathogens that can<br />

cause enterocolitis, bacteraemia, meningitis, brain abscess, and urinary tract infection. They have been<br />

particularly associated with meningitis in neonates where infant milk formulae have been<br />

epidemiologically linked to the disease. This study was carried out during the period of November 2008<br />

to March 2009; to determine the occurrence of E. sakazakii in clinical specimens and its resistance to<br />

traditionally used antimicrobial agents in the Sudan. A total of 389 (311 urine specimens, 11 wound<br />

specimens, and 67 stool specimens) were collected from outpatients of three leading hospitals in<br />

Khartoum State. The urine specimens and wound swabs were cultured on blood and MacConkey's<br />

agars for primary isolation of pathogen, while stool specimens were cultured on selenite F broth and<br />

incubated overnight then subcultured on xylose lysine deoxycholate agar. Identification of the E.<br />

sakazakii was done by colonial morphology, Grams stain and biochemical tests using API 20E. Modified<br />

Kirby-Bauer disc diffusion method was adopted to determine the resistance rate of E. sakazakii to<br />

fifteen antimicrobial agents. Minimum inhibitory concentration (MIC) of antimicrobial agents was<br />

determined by E-test. The result showed that out of 389 specimens examined, 6 (1.5%) E. sakazakii were<br />

recovered, 4 (1.03%) from urine, 1(0.3%) from wound and 1(0.3%) from stool. The results more over<br />

revealed that the antimicrobial resistance of E. sakazakii was as follows; ceftazidime, amoxicillin,<br />

amoxyclav (100% each), co-trimoxazole, ticarcyline (83.3% each), chloramphenicol, tetracycline,<br />

ceftriaxone, nitrofuratoin, cephotaxime, tobramycin (66.7% each), ciprofloxacin, amikacin and nalidixic<br />

acid (16.7% each). None of the isolates were found to be resistant to gentamicin. In addition to that, the<br />

result indicated that the MIC, MIC50 and MIC90 of different antimicrobial agents range from 0.001 to > 240,<br />

0.1 to > 240 and 0.5 to > 240 µg/ml respectively. The results indicated for the first time the presence of E.<br />

sakazakii in the examined clinical specimens in Sudan. The occurrence was high and the antimicrobial<br />

resistance of the isolated E. sakazakii was also high.<br />

Key words: Enterobacter sakazakii, API 20E, E-Test, Khartoum, Sudan.<br />

INTRODUCTION<br />

Enterobacter sakazakii (E. sakazakii) is a Gram-negative<br />

rod belonging to the family Enterobacteriaceae (Fiore et<br />

al., 2008). The organisms have been identified as<br />

emerging opportunistic pathogens that can cause<br />

enterocolitis, bacteraemia, meningitis, brain abscess and<br />

urinary tract infection. They have been particularly


associated with meningitis in neonates (Ongrádi, 2002;<br />

MacLean et al., 2009). Neonatal pathologies also include<br />

wound exudates, appendicitis, and conjunctivitis (Conte<br />

and Passantino, 2008). The organism can be found in<br />

broad range of foods including powdered infant formula,<br />

cheese, meat, vegetables, grain, herbs, spices, tomato,<br />

and in water, in a variety of areas, including hospitals and<br />

houses (Fiore et al., 2008; Kim et al., 2008; Beuchat et<br />

al., 2009).<br />

Although most documented cases involve infant<br />

reports, infections in adult have been also described<br />

(Bhat et al., 2009). Adults with E. sakazakii infection<br />

usually have serious underlying disease or malignancies.<br />

The wound infection with E. sakazakii wound infection in<br />

an adult patient has been reported. The organism was<br />

resistant to multiple antibiotics and required prolonged<br />

treatment with broad spectrum antibiotics (Hamilton et al.,<br />

2003). It is thus necessary to introduce various control<br />

measures to reduce the risk of contamination at various<br />

levels (Fiore et al., 2008).<br />

Antimicrobial agents from natural sources (muscadine<br />

seed extracts, rich sources of phenolic compounds and<br />

organic acids) demonstrated a strong antimicrobial effect<br />

against E. sakazakii (Kim et al., 2009). Control can be<br />

done using accurate antimicrobial agents. The later have<br />

been recovered from natural sources like, muscadine<br />

seed extracts, rich sources of phenolic compounds and<br />

organic acids to demonstrate a strong antimicrobial effect<br />

against E. sakazakii (Kim et al., 2009).<br />

Cited publications have demonstrated that E. sakazakii<br />

can be isolated in hospitals from clinical specimens taken<br />

from patients. Revising literature, we found that this<br />

organism has never been isolated in the Sudan. This<br />

study was designed to determine possible occurrence<br />

and antimicrobial resistance of E. sakazakii in clinical<br />

specimens.<br />

MATERIALS AND METHODS<br />

Study design<br />

This work is a descriptive and cross-sectional study that included<br />

389 patients, complaining from symptoms of UTI, enterocolitis, and<br />

wound infection in Khartoum state. Urine, stool, and wound swab<br />

*Corresponding author. E-mail: biotechsust@hotmail.com. Tel:<br />

+249 83773074. Fax: +249 83 771512.<br />

Humodi and Rania 2375<br />

collection and patient’s data were done in Kartoum Teaching<br />

Hospital (KTH), Gaffer Iben Auff Specialized Hospital for Children<br />

(GIASH) and Omdurman Teaching Hospital. Laboratory investigation<br />

was done in Research Laboratory, College of Medical Laboratories<br />

Science, Sudan University of Science and Technology. Data were<br />

collected in accordance with structured interview questionnaire.<br />

Collection of specimens<br />

Urine<br />

Mid-stream urine (MSU) was collected. Patients were asked to<br />

clean the pre-urethra area with soap and water. Adult females were<br />

asked to make swabbing of the urethra with sterile swab avoiding<br />

any antiseptic then were asked to pass the first drops of urine and<br />

collect 10-20 ml of mid stream urine in sterile, wide mouth<br />

containers.<br />

Wound swabs<br />

Sterile cotton wool swab was used to collect specimens of<br />

discharge from the infected wound. Where there was no discharge,<br />

swab was moistened with sterile normal saline, prior to collection of<br />

specimen and subsequently inserted in sterile tube. In case of<br />

deeply ulcerated wound, a syringe was employed to aspirate the<br />

sample from the side wall of the ulcer. Specimens were delivered to<br />

the laboratory as soon as possible.<br />

Stool<br />

Freshly passed stool specimens were collected in sterile wide<br />

mouth containers and cultures were performed immediately after<br />

collection. In case of delay, specimens were stored at 4°C.<br />

Cultivation of specimens<br />

Under aseptic conditions, urine and wound specimens were<br />

streaked on MacConkey's agar and blood agar (Oxoid Ltd., UK).<br />

Stool specimens were cultured in selenite F broth (Oxoid Ltd., UK),<br />

incubated overnight at 37°C then subcultured on xylose lysine<br />

deoxycholate agar (XLD), (Oxoid Ltd., UK) and incubated<br />

aerobically overnight at 37°C.<br />

Examination of growth<br />

Abundant growth was examined for lactose fermentation on<br />

MacConkey's agar, XLD agar and haemolysis on blood agar. The<br />

various morphological characters of the targeted organism were<br />

also observed and recorded. The isolates were then streaked on<br />

nutrient agar and incubated overnight at 37°C. The growth was<br />

checked for purity and stored in Bijou Bottles for further<br />

investigations.


2376 Afr. J. Microbiol. Res.<br />

Criteria of isolation of E. sakazakii<br />

Gram-negative rods were considered significant (Guptta et al.,<br />

2003) and were investigated.<br />

Identification of E. sakazakii<br />

Bacterial colonies were examined for round, irregular, crenated or<br />

branching appearance, transparency or opacity, smoothness or<br />

roughness, dullness or shiny appearance. The color of the colonies<br />

was also examined (Murray et al., 2007).<br />

Gram's stain<br />

The method was carried out according to Murray et al. (2007).<br />

Oxidase test<br />

The test was carried out according to Barrow and Filtham (2003).<br />

Briefly, oxidase reagent discs were placed on sterile Petri dish and<br />

rubbed with colony of the test organism removed by using a<br />

wooden stick. Formation of a red-purple color within 20 s indicated<br />

positive oxidase test.<br />

Analytical profile index (API 20E)<br />

The API 20E (20 biochemical tests) was performed according to<br />

manufacturer instructions. These were ONPG, ADH, LDC, ODC,<br />

CIT, H2S, URE, TDA, IND, VP, GEL, GLU, MAN, INO, SOR, RHA,<br />

SAC, MEL, AMY and ARA. Identification was obtained with the<br />

numerical profile and performed using the database with the<br />

analytical profile index by looking up to the numerical profile in the<br />

list of profile.<br />

Antimicrobial sensitivity test<br />

Modified Kirby-Bauer disc diffusion method was performed (WHO,<br />

1996; NCCLS. 1997; Murray et al., 2007). Briefly, plates of Mueller-<br />

Hinton agar (Oxoid Ltd, UK) were prepared according to the<br />

manufacturer instructions; the sterilized molten medium was cooled<br />

to 45 to 50°C and poured in sterile, dry Petri plates on a leveled<br />

surface, to a depth of 4 mm. The inoculum was prepared and<br />

turbidity was adjusted to 0.5 McFarland standard. A sterile non-toxic<br />

cotton swab on a wooden applicator was dipped into the<br />

standardized inoculum and the soaked swab was rotated firmly<br />

against the upper inside wall of the tube to express excess fluid.<br />

The entire agar surface of the plate was streaked with the swab<br />

three times with turning the plate at an angle of 60 degrees<br />

between each streaking; the inoculum was allowed to dry for 5-15<br />

min with lid in place. Using sterile forceps the antimicrobial discs<br />

were placed and evenly distributed on the inoculated plate. The<br />

plate was then inverted and incubated aerobically at 35-37°C for<br />

18-24 h. After overnight incubation, the control and test plates were<br />

examined to ensure the growth is confluent or near confluent. Using<br />

a ruler on the under side of the plate, the diameter of each inhibition<br />

zone was measured in (mm).<br />

Interpretation of the zone size<br />

Using the interpretive chart, the inhibition zone around each<br />

antibiotic disc was measured and reported as sensitive, resistant or<br />

intermediate according to WHO (1996), NCCLS (1997) and Murray<br />

et al. (2007).<br />

Minimum inhibitory concentration (MIC) test<br />

The MIC test was performed to each isolate by the E-test technique<br />

as recommended by the manufacturer (HiMedia Laboratories Pvt.<br />

Limited). Briefly, each isolate was tested against the fifteen<br />

antimicrobial agents. A 200 mm Petri plate containing 90 ml of<br />

Mueller-Hinton agar (M173) was used. The inoculum was prepared<br />

as suspension in 0.85% NaCl. The suspension was adjusted to a<br />

0.5 McFarland standard. Plates were seeded with a cotton swab as<br />

described by manufacturer. The E-test strips were applied after the<br />

excess moisture had been absorbed into the agar. The plates were<br />

then incubated at 35-37�C and examined after 18-24 h.<br />

RESULTS<br />

During the course of this study (November 2008 to March<br />

2009) a total of 389 patients were recruited. 311 of these,<br />

presented with UTI, 11 with wound infection and 67 with<br />

enterocolitis. Out of the total number of urine samples<br />

collected, 163 (52.4%) showed significant bacterial<br />

growth (127 lactose fermeters (LF) and 36 non-lactose<br />

fermeters (NLF)). 10 (90.9%) of wound swabs and 61<br />

(91%) of stool specimens revealed significant growth, 4<br />

LF; 6 NLF and 61 LF 6 NLF respectively.<br />

Identification of E. sakazakii<br />

Colonial morphology<br />

Colonial morphology was observed on blood and<br />

MacConkey’s agars after overnight incubation for urine<br />

and wound specimens, and on XLD for stool. Suggestive<br />

E. sakazakii isolates produced large (3-4 mm), dull-gray,<br />

and dry or mucoid colonies on blood agar, large and pink<br />

or colorless, mucoid colonies on MacConkey ’ s agar and


Table 1. Criteria for identification E. sakazakii on API 20 E.<br />

Biochemical tests Reaction Result<br />

ONPG Yellow-pale yellow Positive<br />

ADH Red / orange Positive<br />

LDC Yellow color Negative<br />

ODC Red / orange Positive<br />

CIT Blue-green blue Positive<br />

H2S Colorless Negative<br />

URE Yellow color Negative<br />

TDA Yellow color Negative<br />

IND Colorless Negative<br />

VP Pink/ red Positive<br />

GEL Diffusion of black pigment Positive<br />

GLU Yellow color Positive<br />

MAN Yellow color Positive<br />

INO Blue/ blue-green Negative<br />

SOR Blue/ blue-green Negative<br />

RHA Yellow color Positive<br />

SAC Yellow color Positive<br />

MEL Yellow color Positive<br />

AMY Blue/ blue-green Negative<br />

ARA Yellow color Positive<br />

Table 2. Antimicrobial resistance of E.<br />

sakazakii isolates (n=6).<br />

Antibiotic Resistance (%)<br />

Chloramphenicol 66.7<br />

Ciprofloxacin 16.7<br />

Tetracycline 66.7<br />

Gentamicin 00.0<br />

Ceftazidime 100.0<br />

Amikacin 16.7<br />

Ceftriaxone 66.7<br />

Ticarcyline 83.3<br />

Nitrofuratoin 66.7<br />

Co-trimoxazole 83.3<br />

Amoxicillin 100.0<br />

Amoxyclav 100.0<br />

Nalidixic acid 16.7<br />

Cefotaxime 66.7<br />

Tobramycin 66.7<br />

pink color on XLD.<br />

API 20 E<br />

Humodi and Rania 2377<br />

The results of API 20E revealed that ONPG, ADH, ODC<br />

and CIT were positive, H2S, LDC, URE, TDA and IND<br />

were negative. VP and GEL were also positive and all<br />

sugars GLU, MAN, RHA, SAC MEL, AMY and ARA were<br />

positive except INO, SOR, AMY which were negative<br />

(Table 1).<br />

The total number of E. sakazakii isolates were 6 (1.5%)<br />

including 4 (1.03%) from urine, 1 (0.3%) from wound and<br />

1(0.3%) from stool. The results revealed that the<br />

resistance rate of E. sakazakii was (100%) to amoxicillin<br />

and amoxyclav, ceftazidime, (0%) to gentamicin, (83.3 %)<br />

to co-trimoxazole and ticarcyline, (16.7%) to<br />

ciprofloxacin, amikacin, and nalidixic acid, (66.7%) to<br />

chloramphenicol, tetracycline, nitrofuratoin, cefotaxime,<br />

ceftriaxone, and tobramycin (Table 2). The Minimum<br />

Inhibitory Concentration (MIC), MIC50 and MIC90 values<br />

respectively were (3->240, 60 and >240 µg/ml) for<br />

nitrofurantoin, (0.5->240, 4 and 4 µg/ml ) for nalidixic<br />

acid, (0.1->240, >240 and >240 µg/ml) for cotrimoxazole,<br />

(0.01->240, 4 and 4 µg/ml) for<br />

chloramphenicol, (0.1-64, 0.1 and 0.5 µg/ml) for<br />

amikacin, (0.1-30, 5 and 30 µg/ml) for gentamicin, (1-16,<br />

4 and 8 µg/ml) for tobramicin, (0.001-0.5, 0.25 and 0.5<br />

µg/ml) for ciprofloxacin, (0.1-60, 30 and 30 µg/ml) to<br />

ceftriaxone, (0.1->240, 15 and 15 µg/ml) for ceftazidime,<br />

(0.01-3, 2 and 3 µg/ml) for tetracycline, (10->240, >240<br />

and >240 µg/ml) for ticarcycline, (0.1->240, >240 and<br />

>240 µg/ml) for amoxicillin, (0.1->240, 60 and 60 µg/ml )<br />

for cefotaxime, (2->240, >240 and >240 µg/ml) for<br />

amoxyclav (Table 3).<br />

DISCUSSION<br />

This study was conducted to determine the frequency<br />

and antimicrobial resistance of E. Sakazakii. Three<br />

hundred and eleven urine specimens, eleven wound<br />

specimens, and sixty seven stool specimens were<br />

investigated. Of these, 163 (52.4 %) from urine<br />

specimens, 10 (90.9%) from wound specimens, and<br />

61(91%) from stool specimens showed significant various<br />

bacterial growth.<br />

E. sakazakii was isolated from 6(1.5%) out of 389


2378 Afr. J. Microbiol. Res.<br />

Table 3. MIC range, MIC50 and MIC90 of antimicrobial agents to E. sakazakii.<br />

Antibiotics MIC µg/ml No tested<br />

MIC range<br />

MIC µg/ml<br />

MIC50 MIC90<br />

Nitrofuratoin 6 3 to >240 60 >240<br />

Nalidixic Acid 6 0.5 to> 240 4 4<br />

Co-trimoxazole 6 0.1 to > 240 >240 >240<br />

Chloramphenicol 6 0.01 to > 240 4 4<br />

Amikacin 6 0.1 to 64 0.1 0.5<br />

Gentamicin 6 0.1 to 30 5 30<br />

Tobramicin 6 1 to >16 4 8<br />

Ciprofloxacin 6 0.001 to >0.5 0.25 0. 5<br />

Ceftriaxone 6 0.1 to > 60 30 30<br />

Ceftazidime 6 0.1 to > 240 15 15<br />

Tetracycline 6 0.01 to 3 2 3<br />

Ticarcyline 6 10 to > 240 >240 >240<br />

Amoxycillin 6 0.1 to > 240 >240 >240<br />

Cefotazime 6 0.1 to > 240 60 60<br />

Amoxyclav 6 2 to > 240 >240 >240<br />

MIC; Minimum inhibitory concentration.<br />

patients (urine, wound and stool). The isolates showed<br />

large, dull-gray, and dry or mucoid colonies on blood agar<br />

and large, pink, mucoid colonies on MacConkey's agar.<br />

These results were comparable to those described by<br />

Erickson and Kornacki (2002) and Engelkirk and Duben-<br />

Engelkirk (2007). The frequency 6 (1.5%) of E. sakazakii<br />

in three types of infections (UTI, Enterocolitis, Wound<br />

infection) is similar to the results obtained by Paterso et<br />

al. (2005), who reported that E. sakazakii was responsible<br />

for (0.4%) of the three types of infection in five<br />

international regions including Asia, Europe, Latin<br />

America, North America, and Middle East.<br />

In vitro activities of antimicrobial agents indicated that<br />

the resistance rate of E. sakazakii to amoxicillin and<br />

ceftazidime were (100%). This result is consistent with<br />

Farajnia et al. (2009) who found that the resistance rate<br />

of E. sakazakii to amoxicillin was (100%) in Iran but<br />

different from those of Sader et al. (2005) who found the<br />

resistance rate to ceftazidime was (17.5%) in United<br />

States. The resistance rate of E. sakazakii to nitrofuratoin<br />

(66.7%), however is widely different from those of<br />

Farajnia et al. (2009) who reported a 37.5% resistance<br />

rate to nitrofuratoin in Iran.<br />

The efficacy of cefotaxime, tobramycin, gentamicin and<br />

ciprofloxacin were evaluated and the result showed the<br />

resistance rates of E. Sakazakii to these antibiotics was<br />

(66.7%), (66.7%), (0%) and (16.7%) respectively. This<br />

differs from results obtained by Pfaller and Jones (2002),<br />

who reported that the resistance rate to cefotaxime,<br />

tobramycin and gentamicin is (23%), (16%) and (11%)<br />

respectively but similar to ciprofloxacin (16%) in<br />

European countries. However, the results indicated that<br />

resistance to nalidixic acid (16.7%) was different<br />

compared to that of Farajnia et al. (2009) who reported a<br />

resistance rate of 0.0 % to this antibiotic. Similarly the<br />

resistance rate to amikacin in this investigation is quite<br />

different from (0.0%) resistance rate reported by Farajnia<br />

et al. (2009).<br />

On the other hand, this study showed that the MIC50<br />

and MIC90 of amikacin, ciprofloxacin, tobramycin, and<br />

gentamicin were in agreement with Pfaller and Jones<br />

(2002).<br />

It is concluded that resistance rate of E. sakazakii is<br />

high to commonly used antimicrobial agents. It is also<br />

concluded that gentamicin is the first choice for treatment<br />

of E. sakazakii infections. Ciprofloxacin, amikacin, and<br />

nalidixic acid are the second choice of antimicrobial<br />

therapy with resistance rate of (16.7%). Nitrofurantoin,


tetracycline, ceftriaxone, cephotaxime, chloramphenicol,<br />

and tobramycin on the other hand are less effective<br />

antimicrobial agents due to their high resistance rate<br />

(66.7 %). Amoxicillin, amoxyclav, and ceftazidime are not<br />

effective, having a resistance rate of 100 %. However;<br />

further future studies including larger sample sizes are<br />

recommended, and to our knowledge this report<br />

represents the first record of E. sakazakii in the Sudan.<br />

ACKNOWLEDGEMENT<br />

The authors are grateful to Prof. A. Rahim M. El Husien<br />

for manuscript review.<br />

REFERENCES<br />

Barrow GI, Feltham RKA (2003). Cowan and Steel's Manual for the<br />

Identification of Medical Bacteria: 3 rd edition; Cambridge University<br />

Press, p. 352. ISBN: 0521543282.<br />

Beuchat LR, Kim H, Gurtler JB, Lin L, Ryu J, Richards GM (2009).<br />

Cronobacter sakazakii in foods and factors affecting its survival,<br />

growth, and inactivation, Int. J. Food. Microbiol., 2: 570-749.<br />

Bhat G, Anandhi RS, Dhanya VC, Shenot MS (2009). Urinary tract<br />

infection due to Enterobacter sakazakii. India J. Pathol. Microbiol.<br />

52(3): 430-431. DOI: 10.4103/0377-4929.55017.<br />

Conte F, Passantino A (2008). Isolation of E. sakazakii from ass’ milk in<br />

Sicily: Case report, safety and legal issues, 2 nd Int Conference. J.<br />

Travel Med. Infect. Dis., 6(4): 250-252.<br />

Engelkirk PG, Duben-Engelkirk J (2007). Laboratory Diagnosis of<br />

Infectious Disease: Essentials of Diagnostic Microbiology, Edition<br />

illustrated, Lippincott Williams and Wilkins, Philadelphia, USA. Ch,<br />

12: 307-308.<br />

Erickson MC, Kornacki JL (2002). Enterobacter sakazakii: an emerging<br />

food pathogen, J. Center. Food Safety, pp. 2-8.<br />

Farajnia S, Alikhani MY, Ghotaslou R, Naghili B, Nakhband A (2009).<br />

Causitive agents and antimicrobial susceptibilities of urinary tract<br />

infections in the northwest of Iran, Int. J. Infect. Dis., 13(2): 140-144.<br />

Fiore A, Casale M, Aureli P (2008). Enterobacter sakazakii:<br />

epidemiology, clinical presentation, prevention and control. Ann Ist<br />

Super Sanità, 44(3): 275-280.<br />

Humodi and Rania 2379<br />

Guptta V, Yadav A, Joshi RM (2003). Antibiotics Resistance Pattern in<br />

uropathogen Indian. J. Med. Microbial., 20: 96-98.<br />

Hamilton JV, Lehane J, Braig HR (2003). Isolation of Enterobacter<br />

sakazakii from midgut of stomoxys calcitrans, J. Emerg. Infect. Dis.,<br />

9(10): 1355-1356.<br />

Kim JB, Cho Seund-Hak JC, Park YB, lee JB, lec Kim JC, Lee BK<br />

(2008). Surveillance of stool samples for the presence of E. sakazakii<br />

among Korean people, Yonsei Med. J., 49(6): 1017-1022.<br />

Kim TJ, Silva JL, Weng WL, Chen WW, Corbitt M, Jung YS, Chen YS<br />

(2009). Inactivation of E. sakazakii by water-soluble muscadine seed<br />

extracts, Int. J. Food. Microbiol., 129(3): 295-299.<br />

MacLean LL, Pagotto F, Farber JM, Perry MB (2009). The structure of<br />

the O-antigen in the endotoxin of the emerging food pathogen<br />

Cronobacter (Enterobacter) muytjensii strain 3270, J. Carbohydrate<br />

Res., 344(5): 667-671.<br />

Murray PR, Baron EJ, Jorgensen JH, Landry ML, Pfaller MA (2007).<br />

Manual of Clinical Microbiology. 9th ed. Washington DC: ASM Press,<br />

2007.<br />

NCCLS (1997). Performance standards for Antimicrobial disk<br />

Susceptibility tests. Six edition. ISBN 1 56238 306 6. Available from<br />

NCCLS. 940 West Valley Road. Suit 1400. Wayne. PA 19087-1898,<br />

USA.<br />

Ongrádi J (2002). Vaginal infection by Enterobacter sakazakii. Sex<br />

Transm Infect., 78: 467.<br />

Paterso DL, Rsossi F, Baquero F, Hsueh P, Woods GL, Satishchandran<br />

V, Snyder TA, Harvey CM, Teppler H, Dinubile MJ, Chow JW (2005).<br />

In vitro susceptibilities of aerobic and facultative Gram-negative bacilli<br />

isolate from patients with intra-abdominal infection world wide with<br />

2003 Study for Monitoring Antimicrobial Resistance Trends (SMART),<br />

J. Antimicrob. Chemother., 33(6): 965-973.<br />

Pfaller MA, Jones RN (2002). MYSTIC Study Group (Europe)<br />

Antimicrobial susceptibility of inducible Ampc B-lactamase-producing<br />

Enterobacteriaceae from the Meropenem Yearly Susceptibility Test<br />

Information Collection (MYSTIC) Programme Europe 1997-2000, Int .<br />

J. Antimicrob. Agents, 19(5): 383-388.<br />

Sader HS, Fritsche TR, Jones RN (2005). Potency and spectrum trends<br />

for cefepime tested against 65746 clinical bacterial Isolates Collected<br />

in North American Medical Centers: Results from the SENTRY<br />

Antimicrobial Surveillance Program (1998-2003), J. Diagnostic<br />

Microbiol. Infect. Dis., 52(3): 265-273.<br />

WHO (1996). Guideline for antimicrobial resistance Surveillance, ISBN<br />

92 9021 2136. Available from WHO Regional Office for the Eastern<br />

Mediterranean. PO Box 151. Alexandria 21511. Egypt.


African Journal of Microbiology Research Vol. 5(16), pp. 2380-2384, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR09.462<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Full Length Research Paper<br />

In search of enteroviruses in water media in Marrakech<br />

Amina Hssaine 1 , Jawhar Gharbi 2 , Rafik Harrath 2 , Rajae Harrak 3 , Abderrahman Chait 4 ,<br />

Mahjoub Aouni 2 and Jamal Hafid 1 *<br />

1 Equipe Immuno-Parasitologie et Physiologie, Laboratoire Aliments, Environnement et Santé,<br />

Faculté des Sciences et Techniques, Gueliz, Marrakech, Morocco.<br />

2 Laboratoire des maladies transmissibles et substances biologiquement actives (MDT-01), Faculté de Pharmacie,<br />

5000 Monastir, Tunisie.<br />

3 Laboratoire régional du diagnostic épidémiologique et de l’hygiène du Milieu, Marrakech Morocco.<br />

4 Laboratoire d’Ecophysiologie, Faculté des Sciences Semlalia, Marrakech, Morocco.<br />

Accepted 7 April, 2010<br />

Enteroviruses are among the most common viruses infecting the human intestine; they are very<br />

widespread in nature and resistant to external agents. They are eliminated in the faeces and<br />

contaminate water and food. These viruses cause various clinical syndromes and constitute a big<br />

public health problem. The aim of our study was to search for enteroviruses in the water samples. For<br />

this, a total of 225 L of tap water coming from the national network office for drinking water (ONEP) and<br />

18 samples of waste water originating from Marrakech city were studied. These samples were<br />

concentrated, treated with polyethylene glycol 6000 and then analyzed by RT-PCR. Only two samples<br />

were found to be positive for enteroviruses by RT-PCR among the 18 waste water samples analyzed,<br />

which gave a rate of 11.11%. On the other hand, no positive samples were found in the tap water. This<br />

study made it possible on the one hand to apply for the first time RT-PCR for the detection of<br />

enteroviruses in water samples originating from Marrakech city, and on the other hand to show that tap<br />

water of this city does not present any risk of contamination by this type of germ.<br />

Key words: Drinking water, waste water, enterovirus, RT-PCR, Marrakech.<br />

INTRODUCTION<br />

Water is essential to life, its availability in quantity and<br />

quality sufficient to meet basic needs is essential both for<br />

improving health and for sustainable development.<br />

Combating and reducing diseases linked to water is one<br />

of the important and urgent tasks which can be achieved<br />

only through enhanced cooperation at all levels.<br />

Enteric viruses can be present naturally in aquatic<br />

environments. These viruses can be transported into the<br />

environment through groundwater, estuaries, sea water,<br />

rivers, aerosols, sewage, inadequately treated water,<br />

drinking water, wells etc. (Fong and Lipp, 2005). The risk<br />

of infection by enteroviruses in water is 10 to 10,000<br />

times greater than bacterial infections (Bosch, 1998).<br />

Enteroviruses were isolated from environmental<br />

samples for more than 60 years (Paul and Trask, 1941)<br />

and have subsequently been widely studied and qualified<br />

*Corresponding author. E-mail: hfjamal@yahoo.fr.<br />

as a real threat to the quality of drinking water. The<br />

contamination rate allowed was set by the World Health<br />

Organization to be 3.2 x 10 -5 particles per litre (WHO,<br />

2006).<br />

Viruses excreted in faeces are found in sewage, the<br />

viral concentration of waste water is variable depending<br />

on many factors, geographic, socio-economic, seasonal<br />

and especially health. Thus, the higher the proportion of<br />

children in a community, the lower the level of hygiene<br />

and the higher the quantity of virus found in waste water.<br />

Enteroviruses are transmitted by the faecal-oral route,<br />

their effects may exceed gastroenteritis, because they<br />

pass from the intestinal tract to other organs. The enteric<br />

virus infections are primarily associated with diarrhoea in<br />

humans (Kocwa-Haluch, 2001). Thus, in the United<br />

States, cases of acute gastroenteritis recorded exceeded<br />

50% of all waterborne diseases from 1946 until 1980<br />

(Lippy and Waltrip, 1984) and, viruses were identified as<br />

the cause of 12% of the cases registered during this<br />

period.


In Morocco, few studies have been devoted to sources of<br />

contamination of people by enteroviruses, which is why in<br />

the present work we focused on assessing the virological<br />

quality of water intended for public consumption as well<br />

as surface waste in the region of Marrakech. The aim of<br />

this study was to search for enteroviruses, by RT-PCR, in<br />

225 L of tap water coming from the national network<br />

office for drinking water (ONEP) and in 18 samples of<br />

waste water originating from Marrakech city.<br />

MATERIALS AND METHODS<br />

Samples<br />

Water faucet<br />

Samples were taken directly from the tap (Régie autonome de<br />

distribution d'eau et d'électricité de Marrakech: RADEEMA). 15<br />

samples were taken during the months of July, September and<br />

August 2007 and October 2008 making a total volume of 225 L.<br />

Wastewater<br />

Samples were taken from the waste water treatment plant of<br />

Marrakech. Eighteen samples of 250 ml were taken, in June, July<br />

and October 2008, in clean glass containers. Eleven were taken<br />

from the raw water at the entrance to the station and seven from<br />

the treated waste water as it exited. These samples were taken to<br />

the Environment, Food and Health Laboratory of Marrakech for<br />

extraction and concentration of the virus. The last pellets after<br />

concentration were kept at -20°C for analysis by RT-PCR in the<br />

Diseases and Biologically Active Substances Laboratory of<br />

Monastir, Tunisia.<br />

Water wells<br />

During the months of June and July 2007, eight samples of one litre<br />

each were collected in eight wells located within a 2 km radius<br />

around the Oued Tensift located 6.3 km north of the city of<br />

Marrakech at 410 m altitude. The wells were located in clusters of<br />

habitats near Oued Tensift. This water is used by the people both<br />

for drinking and for the preparation of food without any treatment.<br />

These villages are not served by the national drinking water<br />

system.<br />

Sample processing<br />

Concentration of enteroviruses<br />

The concentration of virus in samples of tap water and wells has<br />

been done by filtration through membranes of sterile cellulose<br />

nitrate with porosity of 0.22 microns. The filtration device was<br />

connected to a vacuum pump. Each filter is then placed in a moist<br />

chamber and stored at 4°C (Wallis et al., 1972).<br />

Elution<br />

Each filter was cut into very fine pieces separately and put in 20 ml<br />

of a solution of meat extract pH 9.5 and centrifuged at 10, 000<br />

rounds/min for 35 min. The float was recovered and its pH adjusted<br />

to 7.2 with 5M HCl. One milliliter of the mixture of antibiotics and<br />

Hssaine et al. 2381<br />

antifungal (penicillin, streptomycin and fungizone) was added and<br />

the final mixture was placed at 4°C overnight and stored at -20°C.<br />

Extraction and concentration from waste water<br />

We adapted the protocol described by Gerba et al. (1979), based<br />

on the adsorption of virus to acidic pH and elution at basic pH.<br />

Thus, 100 ml of sample were homogenized by magnetic stirring. A<br />

solution of AlCl3 was added until a final concentration of 0.0005 M<br />

and the pH was adjusted to 3.5 with 5N HCl. The mixture was<br />

stirred vigorously for 30 min then centrifuged at 2500 g for 15 min at<br />

4°C and the pellet was taken in 100 ml of 10% beef extract of at pH<br />

9. The mixture was stirred for 30 min and centrifuged at 10 000 g<br />

for 30 min at 4°C and the supernatant was collected in a sterile<br />

container. PEG 6000 was added until a final concentration of 10%<br />

and the mixture was homogenized and incubated overnight at 4°C.<br />

The next day, after a centrifugation step at 10 000 g for 45 min at<br />

4°C, the pellet was resuspended in 10 ml phosphate buffer pH 7.2<br />

and kept at -20°C.<br />

Extraction of viral RNA<br />

A volume of 120 µl of each sample was put in 500 l of Tri-reagent<br />

(Sigma) and vortexed for 15 s and left in contact 5 min at room<br />

temperature. Then, 200 l of chloroform were added to the mixture<br />

to separate the nucleic acids of proteins to which they are<br />

associated. The mixture was vortexed, left to react at room<br />

temperature for 15 min and then centrifuged at 13 500 rpm for 15<br />

min at 4°C. The aqueous phase was transferred to a new tube, and<br />

mixed with 500 l of isopropanol. The mixture was allowed to react<br />

for 15 min at -20°C and then centrifuged at 13500 rounds/min for 10<br />

min at 4°C to remove the cell debris. The pellet underwent two<br />

successive washes with 75% ethanol followed by centrifugation at<br />

7500 rounds/min at 4°C for 15 min for the first wash and 5 min for<br />

the second. The pellet was left to dry at room temperature for 15<br />

min. To standardize the RNA in the solution, the pellet was put in<br />

30 l of Diethylpyrocarbonate treated water.<br />

Detection of the viral genome<br />

We used RT-PCR, a method considered sensitive in the detection<br />

of enteroviruses in both environmental and clinical samples (Zoll et<br />

al., 1992; Rutjeset al., 2005; Soule et al., 2000) using reverse<br />

transcriptase (Moloney-Murine Leukemia Virus) and two primers<br />

from the conserved non-coding region of the viral genome. The<br />

product of amplification gives a 154 bp band.<br />

Choice of primers: In the case of enteroviruses, a sequence which<br />

is stable and common to all serotypes must be identified. Thus,<br />

several sequences have been described by Zoll et al. (1992). The<br />

primers used in this work are positioned at 445 - 464 and 599 - 580<br />

on the genome of Coxackievirus B3 and their nucleotide sequences<br />

are 5 'TCCTCCGGCCCCTGAATGCG 3' and 3<br />

'ATTGTCACCATAAGCAGCCA 5' (Invitrogen), respectively.<br />

Preparation of complementary DNA (cDNA): For the preparation<br />

of cDNA, we mixed 2.5 l of of reverse transcriptase buffer<br />

(Promega), 1 l of dithiothreitol (Promega), 0.5 l of NTPS<br />

(Boehring Mannheim), 0.5 l of antisense primer (25 pmole) and<br />

0.5 l of the enzyme reverse transcriptase (Promega). To this<br />

mixture, we added 5 l of extracted RNA and incubated for 30 min<br />

at 42°C and then amplified.


2382 Afr. J. Microbiol. Res.<br />

Table 1. Presence of enteroviruses in waste water<br />

samples of Marrakech.<br />

Samples Date Results<br />

RW1<br />

-<br />

02/06/2008<br />

TW1 -<br />

RW 2<br />

-<br />

18/06/2008<br />

TW2 -<br />

RW 3<br />

-<br />

20/06/2008<br />

TW3 -<br />

RW 4<br />

-<br />

25/06/2008<br />

TW4 -<br />

RW 5<br />

-<br />

27/06/2008<br />

TW5 -<br />

RW 6<br />

01/07/2008<br />

RW 7<br />

-<br />

02/07/2008<br />

TW7 -<br />

RW 8<br />

07/07/2008<br />

RW 9<br />

+<br />

08/07/2008<br />

TW9 -<br />

RW 10<br />

RW 11<br />

25/10/2008<br />

26/10/2008<br />

RW: Raw water; TW: Treated water.<br />

Amplification: The amplification mixture consisted of 2.5 l Taq<br />

polymerase buffer, 0.5 l of NTPs: dATP, dCTP, dGTP and dTTP,<br />

0.5 l antisense primer (25pmole), 0.5 l of sense primer 25 pmole:<br />

oligo (dt) 12 - 18 (pharmacia Biotech), 0.15 l of Taq polymerase<br />

1.25 l (Invitrogen), 0.75 l of MgCl2 and 18 l of sterile doubledistilled<br />

water.<br />

The positive control (RNA Coxackievirus Nancy B) and negative<br />

control (sterile distilled water) were treated in the same conditions<br />

as the samples studied. The amplification was performed in an<br />

Eppendorf thermal cycler with a denaturing RNA-cDNA for 5 min at<br />

94°C and a series of thirty cycles of the following: 30 s at 94°C,<br />

hybridization of primers for a minute at 42°C, elongation 2 min at<br />

72°C and an extension of 10 min at 72°C, the final stretch.<br />

Revelation: After amplification, electrophoretic migration of the<br />

PCR product was performed on 2% agarose gel. Ten milliliters of<br />

each sample of PCR product mixed with 5 l of migration blue were<br />

deposited on the gel. Migration was done in TBE buffer (1x) at 90 V<br />

and lasted for approximately 45 min. The gel was then soaked in<br />

a solution of ethidium bromide (Sigma) at 10 mg/ml. Visualization of<br />

the specific band was conducted by transillumination under<br />

ultraviolet light (UV) against a size marker of 100 bp (Pharmacia<br />

Biotech).<br />

-<br />

+<br />

-<br />

-<br />

RESULTS AND DISCUSSION<br />

During the study, a total of 41 samples were collected<br />

including 18 samples of waste water during the months of<br />

May, June, July and October 2008, 15 samples of tap<br />

water during the months of July, September and August<br />

2007 and October 2008 and 8 samples of water wells<br />

during the months of June and July 2007.<br />

Using RT-PCR enabled the detection of enterovirus<br />

RNA in 2 samples of waste water; this gives a rate of<br />

11.1% (2/18). These two positive samples were collected<br />

in July, known as the driest month of the year 2008 in the<br />

city of Marrakech. However, no positive result was<br />

obtained in the treated waste water, tap water or water<br />

wells (Table 1 and Figure 1).<br />

We have little data on the detection of enteroviruses in<br />

the environment in Morocco because there are few<br />

studies that have been done. This allows us to emphasize<br />

that our present work is original. Our results are<br />

compared to those recorded by researchers in other<br />

countries.<br />

Several studies have shown that the increase in<br />

enteroviruses is seasonal Summer-Fall (Troudi et al.,<br />

1997), while other studies claim that the rainy season is<br />

the perfect time when the frequency of enterovirus<br />

reaches its peak (Bini et al., 2006). A more prolonged<br />

follow-up of our study throughout the year can confirm<br />

either study. We also noted that no positive sample of<br />

treated waste water coming out of the treatment plant has<br />

been detected. This can be explained by the fact that<br />

much of the suspended matter is removed during the<br />

treatment; where the viruses are adsorbed on this<br />

material under the influence of physical surface<br />

properties. Some authors argue that the setting of virus<br />

particles suspended in the water environment is linked<br />

not only to the species and serotype but also to the strain<br />

itself (Nestor and Brisou, 1986). For more sensitivity, it<br />

will be interesting to use nested-PCR for low levels of<br />

enteroviruses (Lee and Lee, 2008; Zhang et al., 2008).<br />

Indeed, RT-PCR is a sensitive technique but is influenced<br />

by the presence of inhibitors which are rich environmental<br />

samples (Sdiri et al., 2006), a factor that limits its<br />

effectiveness. However, many precautions must be taken<br />

both in the implementation of the RT-PCR (including<br />

choosing the method of virus concentration and<br />

extraction technique of RNA). Thus the large amount of<br />

genetic material from environmental samples makes<br />

possible a non-specific amplification of sequences unrelated<br />

to the virus searched.<br />

Regarding the tap water, 225 L of water were analyzed<br />

without any positive result. This could be explained bythe<br />

efficiency of the water decontaminating treatment<br />

process by the treatment plant. Our results are consistent<br />

with those found in Korea (Kyung and Jeong, 2004) where<br />

no enterovirus was detected in treated water for<br />

consumption. However they are different from the study<br />

of Coin (1966) which was the first to have isolated viruses<br />

in water intended for consumption in Paris, and those of


(-) PM 1 2 3 4 (+) PM<br />

Hssaine et al. 2383<br />

154bp<br />

Figure 1. Results of electrophoresis of amplification products by RT-PCR of gene 5'NC<br />

enteroviruses from samples of waste water.<br />

(-): negative control; (+): positive control (Coxackievirus B Nancy); 1, 4: positive samples; 2,<br />

3: negative samples; PM: Size Marker 100 bp.<br />

other studies that were conducted in Romagne (France)<br />

(Nestor and Costin, 1976) and Russia (Rabshko, 1974).<br />

The authors had explained the presence of the virus by<br />

inadequate treatment which did not allow effective<br />

disinfection. In addition, other studies have shown high<br />

contamination of drinking water by enteroviruses in Korea<br />

(Lee and Kim, 2002), South Africa (Ehlers et al., 2005)<br />

and Egypt (Ali et al., 2004) with positivity rates of 47.8,<br />

18.7% and 17.5 PFU/L respectively. These high rates<br />

have been explained by the insufficiency of the treatment<br />

used and the possible contamination of water pipes.<br />

As the methods used in environmental virology are<br />

limited by the low concentration of virus in drinking water,<br />

the techniques of adsorption/elution using electropositive<br />

filters or electronegative methods are most recommended<br />

by APHA (1985) and Karim et al. (2009). Nevertheless,<br />

our study has demonstrated the effectiveness of<br />

the treatment technologies for water consumption, the<br />

technique used and the volume of samples analyzed to<br />

meet international quality standards (EPA, 1999b).<br />

Regarding groundwater, we have made samples in 8<br />

wells and none was found to be positive. These wells<br />

which are not deep (between 6 and 10 m) are located<br />

near agricultural fields irrigated with waste water. Study<br />

should be conducted using wider areas and larger<br />

volumes of water. In other studies, conducted between<br />

1971 and 1982, groundwater accounted for 51% of<br />

waterborne diseases (Craun, 1986). Hepatitis viruses and<br />

rotaviruses are considered the main causes of these<br />

diseases (Gerba et al., 1985). These infections are<br />

mainly due to infiltration of waste water through the pits<br />

as well as due to the spread of waste water and sewage<br />

sludge, domestic discharge, oxidation ponds etc<br />

(Borchardt et al., 2007). With their small size, viruses are<br />

able to travel hundreds of meters through soil to reach<br />

groundwater (Gerba and Bitton, 1984) this movement<br />

depends mainly on the type of virus, the type of soil and<br />

climate.<br />

The present work allowed us to detect for the first time,<br />

enteroviruses in waste water of the region of Marrakech.<br />

This encourages us to continue the search for these<br />

viruses in a larger number of water and human biological<br />

samples.<br />

REFERENCES<br />

Ali MA, Al-Herrawy AZ, El-Hawaary SE (2004). Detection of enteric<br />

viruses, Giardia and Cryptosporidium in two different types of drinking<br />

water treatment facilities. Water Res., 38: 3931-3939.<br />

American Public Health Association (APHA) (1985). Standard Methods<br />

for the examination of water and wastewater, 16 th ed. Washington,<br />

D.C.<br />

Bini JC, Ekaza E, Gnagne T, Borget AMY, Veh KA, Akran AV, Coulibaly<br />

ND, Faye-Kette H, Sess ED, Dosso M (2006). Apport de la RT-PCR<br />

pour la détection des entérovirus dans les eaux usées à Abidjan<br />

(Côte d’ivoire). Cah. Santé Publique, 5: 55-65.<br />

Borchardt MA, Bradbury KR, Gotkowitz MB, Cherry JA, Parker BL<br />

(2007). Human enteric viruses in groundwater from a confined<br />

bedrock aquifer. Environ. Sci. Technol., 15: 6606-6612.<br />

Bosch A (1998). Human enteric viruses in the water environment: a<br />

minireview. Microbiol., 1: 191-196.<br />

Coin L (1966). Modern microbioligical and virological aspects of water<br />

pollution. In Jaag, O (eds) Advances in water Pollution Research,<br />

Pergamon Pres, London, pp. 1-10.<br />

Craun GF (1986). ed. Waterborne diseases in the United States. Boca<br />

Raton: CRC Press, p.13.<br />

Ehlers MM, Grabow WOK, Pavlov DN (2005). Detection of<br />

enteroviruses in untreated and treated drinking water supplies in<br />

South Africa. Water Res., 39: 2253-2258.<br />

EPA (1999b). Drinking water criteria document for enteroviruses and<br />

hepatitis a: an addendum, p. 173.<br />

Fong TT, Lipp EK (2005). Enteric Viruses of Humans and Animals in<br />

Aquatic Environments: Health Risks, Detection, and Potential Water<br />

Quality Assessment Tools. Microbiol. Mol. Biol. R, 69: 357-371.<br />

Gerba CP, Bitton G (1984). Microbial pollutants: their survival and


2384 Afr. J. Microbiol. Res.<br />

transport pattern to groundwater. In Bitton G, Gerba CP (eds)<br />

Groundwater pollution microbiology. Wiley, New York, pp. 39-54.<br />

Gerba CP, Goyal SM, Labelle RL, Cech I, Bodgan GF (1979). Failure of<br />

indicatorbacteria to reflect the occurrence of enteroviruses in marine<br />

water. A. J. Pub. Health, 69: 1116-1119.<br />

Gerba CP, Sing SN, Rose JB (1985). Waterborne viral gastroenteritis<br />

and hepatitis. CRC Crit. Rev. Environ. Contr., 15: 213-216.<br />

Karim M, Rhodes E, Brinkman N, Wymer L, Fout GA (2009). New<br />

Electropositive Filter for Concentrating Enterovirus and Norovirus<br />

from Large Volumes of Water. Appl. Environ. Microbiol., 75: 2393-<br />

2399.<br />

Kocwa-Haluch R (2001). Waterborne Enteroviruses as a Hazard for<br />

Human. Pol. J. Environ. Stud., 10: 485-487.<br />

Kyung Lee H, Jeong YS (2004). Comparison of Total Culturable Virus<br />

Assay and Multiplex Integrated Cell Culture-PCR for Reliability of<br />

Waterborne Virus Detection. Appl. Environ. Microbiol., 70: 3632-<br />

3636.<br />

Lee G, Lee C (2008). Molecular detection and characterization of<br />

human enteroviruses in Korean surface water. J. Microbiol., 46: 319-<br />

324.<br />

Lee S, Kim S (2002). Detection of infectious enteroviruses and<br />

adenoviruses in tap water in urban areas in Korea. Water Res., 36:<br />

248-256.<br />

Lippy EC, Waltrip SC (1984). Waterborne disease outbreaks-1946-<br />

1980: a thirty-five –year perspective. J. Am. Water Works Assn, 76:<br />

60-67.<br />

Nestor I, Costin L (1976). Presence of certain enteroviruses (coxsakie )<br />

in sewage effluents and river waters of Romania. J. Hyg. Epidemiol.<br />

Microbiol. Immunol., 21: 137-149.<br />

Nestor J, Brisou J (1986). Incidence sanitaires de l’adsorption sur les<br />

sédiments marins et fluviaux. Rev. Epidemiol. Sante Publique, 34:<br />

181-190.<br />

Paul JR, Trask JD (1941). The virus of poliomyelitis in stools and<br />

sewage. J. Am. Med. Ass., 116: 493-498.<br />

Rabshko EV (1974). Certain problems of circulation of enteroviruses in<br />

the environemental objects. Gig. Sanit. 39: 105-106.<br />

Rutjes SA, Italiaander R, van den Berg HHJL, Lodder WJ, de Roda<br />

Husman AM (2005). Isolation and Detection of Enterovirus RNA from<br />

Large-Volume Water Samples by Using the NucliSens miniMAG<br />

System and Real-Time Nucleic Acid Sequence-Based Amplification.<br />

Appl. Environ. Microbiol., 71: 3734-3740.<br />

Sdiri K, Khelifi H, Belghith K, Aouni M (2006). Comparaison de la<br />

culture cellulaire et de la RT–PCR pour la détection des entérovirus<br />

dans les eaux usées et les coquillages en Tunisie. Pathol. Biol.,<br />

54: 280-284.<br />

Soule H, Genoulaz O, Gratacap-Cavallier B, Chevallier P, Liu J,<br />

Seigneurin J (2000). Ultrafiltration and reverse transcriptionpolymerase<br />

chain reaction: an efficient process for poliovirus,<br />

rotavirus and hepatitis A virus detection in water. Water. Res., 34:<br />

1063-1067.<br />

Troudi M, Aouni M, Poozzetto B, Dhidah M (1997). Surveillance de la<br />

circulation des entérovirus dans la région du Sahel. Tunis Med., 75:<br />

23-26.<br />

Wallis C, Henderson M, Melnick J (1972). Enterovirus concentration on<br />

cellulose membranes. Appl. Environ. Microbiol., 19: 476-480.<br />

World Health Organization (2006). Guidelines for Drinking-water<br />

Quality. First Addendum To Third Edition Recommendations. – 3rd<br />

ed. 1: 121-144c.<br />

Zhang CM, Wang XC, Liu YJ, Xue XP (2008). Characteristics of<br />

bacterial and viral contamination of urban waters: a case study in<br />

Xi'an, China. Water Sci. Technol., 58: 653-560.<br />

Zoll GJ, Melchers WJG, Kopecka H, Jambroes G, van der Poel HJ,<br />

Galama JM (1992). General primer-mediated polymerase chain<br />

reaction for detection of enteroviruses: application for diagnostic<br />

routine and persistant infections. J. Clin. Microbiol., 30: 160-165.


African Journal of Microbiology Research Vol. 5(16), pp. 2385-2387, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR09.389<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Short Communication<br />

Laboratory analysis of a fatal meningococcal case due<br />

to serogroup B Neisseria meningitidis belonging to ST-<br />

4821 complex<br />

Zengguo Wang*, Tiejun Hou, Zhijun Chen, Jinsong Li, Shouzhi Wu, Xiaoguang Wei,<br />

Yahui Sun and Quanli Du<br />

Xi’an Center for Disease Control and Prevention, 599 Xi Ying Road, Xi’an, 710054, PR China.<br />

Accepted 12 November, 2009<br />

We describle a suspicious Meningococcal death case, confirmating by the laboratory PCR methods.<br />

The infection developed rapidly within only 24 h until the patient died. Analyzing the close contact strain<br />

and the DNA template extracted from the serum of this patient, we characterized a serogroup B<br />

Neisseria meningitidis as the pathogen of this case, which, in terms of sequence typing, belonged to<br />

ST-4821 complex which first characterized in serogroup C Neisseria meningitidis as a new unique<br />

hypervirulent meningococcal lineages.<br />

Key words: Neisseria meningitidis, multilocus sequence typing, serogroup.<br />

INTRODUCTION<br />

As a major bacterial meningititis pathogen, Neisseria<br />

meningitidis could cause a rapidly progressive and often<br />

fatal illness mainly in children and young adults all over<br />

the world. 13 serogroups were known of this organism<br />

based on the chemical and serological properties of the<br />

capsular polysaccharide. Serogroup A meningococci<br />

were the main cause of meningitis and responsibled for<br />

several nationwide epidemics during the last century in<br />

China (Hu, 1991). After the mass vaccination campaign<br />

with mainly the MenA vaccine initiated in 1982, the<br />

incidence of meningococcal disease rapidly declined and<br />

only some sporadic case reported. However, several<br />

outbreaks caused by serogroup C, ST -4821 meningococci<br />

occurred in Anhui province in 2003 and then spread<br />

to other provinces (Shao et al., 2006). To control this<br />

disease, a mass vaccination using polysaccharide<br />

vaccine for serogroup A plus C were undertaken in<br />

several provinces and the A plus C vaccine was used<br />

more prevalent than before in China.<br />

An etiologic diagnosis of N. meningitidis is confirmed by<br />

the isolation of strain from cerebrospinal fluid, blood or<br />

other body fluids. However, this diagnosis was hindered<br />

*Corresponding author. E-mail: upwilly@gmail.com. Tel: 86-10-<br />

8551-2367.<br />

by the failure to isolate bacteria because of the early<br />

treatment of the patient (Cartwright et al., 1992). So the<br />

PCR-based methods have been extensively used for the<br />

diagnosis and also the serotying based on several genes<br />

special for Neisseria meningitidis or the serotype. In this<br />

study, we confirmed a suspicious death case of meningococci<br />

infection with laboratory methods such as PCR<br />

(multiplex-PCR) (Jordens et al., 2002), MLST (Maiden et<br />

al., 1998). We also found the etiologic serogroup B<br />

meningococci belonging to the ST-4821 complex which<br />

was a newly identified lineage that most prevalent in<br />

serogroup C meningococci in China (Zhang et al., 2007).<br />

CASE REPORT<br />

A 16 years old female student was characterized by a<br />

high fever and headache without other symptoms like<br />

cough, diarrhea, vomit, which began on March 11, 2009.<br />

The body temperature was 39.4°C. The symptom was<br />

exacerbated when the patient was checked in LanTian<br />

Hosiptal next day at 7:30 am. Several petechiate rashes<br />

emerged on the whole body with the blood pressure was<br />

118/68 mmHg and pulse rate of 132 per minute. The<br />

numbers of white cell in the blood were 1.6 × 1010/L. The<br />

patient receiving azithromycin, and within drug administration,<br />

the patient had nausea several times. After the


2386 Afr. J. Microbiol. Res.<br />

drug administration, the body temperature was normal<br />

but also with the headache. Then the patient was<br />

transferred to Tang Du Hospital at 10:50 am, the body<br />

temperature was 36°C and the blood pressure was 84/65<br />

mm Hg with pulse rate of 116 per minute. Neck stiffness<br />

and purpura fulminans were noted during clinical examination.<br />

The blood and CSF samples were obtained and<br />

submitted for laboratory examination. Laboratory investigations<br />

revealed thrombocytopenia, the TP, ALB and<br />

GLO were all decreased, the blood culture was negative<br />

for any bacterial. The patient was initially sta-bilized with<br />

volume resuscitation, dopamine infusion, until 13:30, the<br />

patient was shortness of breath and dysphoria again, and<br />

died of acidemia at 14:35. In accordance with the<br />

observed clinical symptoms, this case was diagnosed as<br />

suspicious meningococcal meningitis. The serum of the<br />

patient was transferred to Xi’an CDC laboratory for<br />

detection via PCR and MLST.<br />

The close contact in this case was defined as 5<br />

roomates with the patient. Within the 5 roommates’<br />

oropharyngeal swabs, the ctrA gene was positive in two<br />

samples and 1 strain was isolated. The ctrA gene was<br />

also positive in the patient’s serum. To check the serotype<br />

we use the molecular serogroup methods to predict the<br />

possible serotype described elsewhere (Taha, 2000),<br />

(Bennett et al., 2004). A 450 bp fragment predicted<br />

serogroup B Neisseria meningitidis was positive in the<br />

two ctrA gene positive oropharyngeal swabs sample and<br />

also the patient’s serum. MLST analyses were verified<br />

here with the close contact strain. The sequence type of<br />

this close contact strain was ST -7623 which was a new<br />

ST type found in our study belonging to ST-4821 complex.<br />

MLST was also conducted using the DNA template<br />

extracted from the serum of this patient. Only three<br />

fragments (adk, aroE and gdh) were obtained and<br />

sequenced. To search the type in the PubMLST net, the<br />

allelic profile was identical to 12 sequence types that all<br />

belonging to ST-4821 complexs. Also, the sequence of<br />

these three fragments identical to the close contact strain.<br />

According to the above-listed, we conclude that the<br />

pathogen of this fatal case was serogroup B N.<br />

meningitidis which belonging to ST-4821 complexs.<br />

DISCCUSSION<br />

Currently, rapid laboratory diagnosis of meningococcal<br />

disease has been widely used as routine methods<br />

(Bryant et al., 2004; Hoang et al., 2005). Especially, when<br />

the bacterial could not been isolated, PCR based<br />

methods will be very important in confirming the<br />

meningococci infection. With the only sample like serum<br />

of patient, MLST could be used in epidemiology research<br />

(Zhang et al., 2006). In this case, PCR and MLST method<br />

was used in laboratory confirmation and epidemiology<br />

research as only the serum sample was obtained. To our<br />

knowledge, this is the first death case causing by<br />

serogroup B meningococci belonging to ST- 4821<br />

complexs in China.<br />

Serogroup C meningococci have been associated with<br />

the outbreaks in 2003. Then a unique ST-4821 clone has<br />

been characterized (Shao et al., 2006). ST-4821 complex<br />

isolates were observed as early as 1980 in China and<br />

existed in several serogroups like C, B, H, I and K (Yang<br />

et al., 2007; 2008; Zhang et al., 2007). In addition to ST-<br />

4821 complex has been seen in serogrop B<br />

meningococcal isolated from healthy carriers on the<br />

Chinese mainland, it also have been seen from invasive<br />

cases in Taiwan (Chiou et al., 2006). Here we also found<br />

the serogroup B meningococcal belonging to ST- 4821<br />

complex in a fatal case. Because the pathogen strain of<br />

the patient was not able to isolate and we can only get 3<br />

gene fragments from the patient’s serum. Moreover we<br />

isolated two N. meningitidis strains from the oropharyngeal<br />

swabs of patient’s other 10 classmates. This two<br />

strains also belongs to B serogroup and the sequence<br />

type was ST-4821 and ST-5586 each. The 3 gene<br />

fragments from the patient’s serum also indentical to the<br />

ST-4821 strain. We also got ctrA gene positive result from<br />

2 of 5 close contacts, one close contact strain with the<br />

exact sequence type is unsatisfactory. So we presumed<br />

there was not sufficient evidence to confirm the exact<br />

sequence type of this etiological strain. After all it seems<br />

belonging to ST-4821 complex.<br />

Multivalence vaccines A plus C have used widely in<br />

China after the serogroup C meningococci outbreaks.<br />

Thus, the B serogroup has been observed in healthy<br />

carrier is relative higher than before and even maybe the<br />

capsule switch between B and C (Beddek et al., 2009).<br />

The case presented here indicate the need for better<br />

surveillance of the epidemiology of meningococcal meningitis<br />

in China, especially the new unique hypervirulent<br />

meningococcal lineage ST- 4821 complexs.<br />

ACKNOWLEDGEMENTS<br />

We thank all the researchers in hospitals and Wang Qian<br />

in the Baqiao district CDC. We also thank Dr Julia<br />

Bennett at the University of Oxford for the registration of<br />

the close contact strain as new ST in the MLST database.<br />

REFERENCES<br />

Beddek AJ, Li MS, Kroll JS, Jordan TW, Martin DR (2009). Evidence for<br />

capsule switching between carried and disease-causing Neisseria<br />

meningitidis strains. Infect. Immun., 77: 2989-2994.<br />

Bennett DE, Mulhall RM, Cafferkey MT (2004). PCR-based assay for<br />

detection of Neisseria meningitidis capsular serogroups 29E, X, and<br />

Z. J. Clin. Microbiol., 42: 1764-1765.<br />

Bryant PA, Li HY, Zaia A, Griffith J, Hogg G, Curtis N, Carapetis JR<br />

(2004). Prospective study of a real-time PCR that is highly sensitive,<br />

specific, and clinically useful for diagnosis of meningococcal disease<br />

in children. J. Clin. Microbiol., 42: 2919-2925.<br />

Cartwright K, Reilly S, White D, Stuart J (1992). Early treatment with<br />

parenteral penicillin in meningococcal disease. Bmj, 305: 143-147.<br />

Chiou CS, Liao JC, Liao TL, Li CC, Chou CY, Chang HL, Yao SM, Lee<br />

YS (2006). Molecular epidemiology and emergence of worldwide


epidemic clones of Neisseria meningitidis in Taiwan. BMC Infect. Dis.,<br />

6: 25.<br />

Hoang LM, Thomas E, Tyler S (2005). Rapid and fatal meningococcal<br />

disease due to a strain of Neisseria meningitidis containing the<br />

capsule null locus. Clin. Infect. Dis., 40: 38-42.<br />

Hu X (1991). Study on periodically prevalent feature for epidemic<br />

cerebrospinal meningitis in China. J. Chin. Epidemiol., 12: 136-139.<br />

Jordens JZ, Williams JN, Jones GR, Heckels JE (2002). Detection of<br />

meningococcal carriage by culture and PCR of throat swabs and<br />

mouth gargles. J. Clin. Microbiol., 40: 75-79.<br />

Maiden MC, Bygraves JA, Feil E (1998). Multilocus sequence typing: a<br />

portable approach to the identification of clones within populations of<br />

pathogenic microorganisms. Proc. Natl. Acad. Sci. USA., 95: 3140-<br />

3145.<br />

Shao Z, Li W, Ren J (2006). Identification of a new Neisseria<br />

meningitidis serogroup C clones from Anhui province, China. Lancet,<br />

367: 419-423.<br />

Wang et al. 2387<br />

Taha MK (2000). Simultaneous approach for non-culture PCR-based<br />

identification and serogroup prediction of Neisseria meningitidis. J.<br />

Clin. Microbiol., 38: 855-857.<br />

Yang J, Zhang X, Xu X (2007). Genotypic analysis of serogroups other<br />

than A, B or C of Neisseria meningitidis in China. Scand. J. Infect.<br />

Dis., 39: 819-821.<br />

Yang L, Shao Z, Zhang X, Xu L, Peng J, Xu X, Liang X, Qi Y, Jin Q<br />

(2008). Genotypic characterization of Neisseria meningitidis<br />

serogroup B strains circulating in China. J. Infect., 56: 211-218.<br />

Zhang TG, He X, Chen LJ, He JG, Luo M, Yang J, Shao ZJ, Sun MP<br />

(2006). Laboratory confirmation of a suspicious meningococcal<br />

meningitis death case. J. Microbiol., 44: 457-460.<br />

Zhang X, Shao Z, Yang E (2007). Molecular characterization of serogroup<br />

C Neisseria meningitidis isolated in China. J. Med. Microbiol.,<br />

56: 1224-1229.


African Journal of Microbiology Research Vol. 5(16), pp. 2388-2390, 18 August, 2011<br />

Available online http://www.academicjournals.org/ajmr<br />

DOI: 10.5897/AJMR11.536<br />

ISSN 1996-0808 ©2011 <strong>Academic</strong> <strong>Journals</strong><br />

Short Communication<br />

Genotyping of Hepatitis C virus (HCV) in infected<br />

patients from Saudi Arabia<br />

Mohammed Ali M. Marie<br />

Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud University,<br />

P. O. Box 10219 Riyadh 11433, Kingdom of Saudi Arabia. E-mail: mmarie@ksu.edu.sa. Tel: +96614696022.<br />

Accepted 30 June, 2011<br />

A molecular study was carried to investigate the prevalence of Hepatitis C virus (HCV) genotypes in<br />

HCV infected population of Saudi Arabia. A total of HCV-positive clinical specimens (serum, EDTA<br />

plasma) with viral loads above 2,000 IU/ml were collected for genotyping. Genotyping of 358 samples<br />

revealed four different genotypes including 1 (1a and 1b), 2a, 3 (3a) and 4. The most prevalent genotype<br />

was 4 with rate of 74.2% followed by genotype 3 (11.7%) and 1a (3.9%). The predominance of HCV<br />

genotype 4 in our population confirms the predominance of HCV genotype 4 in Saudi Arabia and most<br />

of the Arab countries in the Middle East.<br />

Key words: Hepatitis C virus (HCV), genotypes, Saudi Arabia, Riyadh.<br />

INTRODUCTION<br />

Hepatitis C virus (HCV) infection is a global public health<br />

problem. World Health Organization (WHO) estimates up<br />

to 3% of the world's population to be infected with HCV<br />

and there are approximately 180 million individuals are<br />

thought to be infected. On average, 80% of acutely infected<br />

individuals develop a chronic infection (Lavanchy,<br />

2009; Ghany, 2009). HCV has a positive-sense singlestranded<br />

RNA genome of flavivirus and about 9.6 kb<br />

containing one long open reading frame (ORF) with<br />

untranslated regions at both ends (Choo et al., 1989). So<br />

far, six major genotypes (HCV-1 to HCV-6) have been<br />

described, each containing multiple subtypes (for<br />

example, 1a, 1b, etc.) (Tokita et al., 1995). The genotype<br />

of the HCV strain appears to be an important determinant<br />

of the severity and aggressiveness of liver infection, as<br />

well as patient response to antiviral therapy (Zein, 2000).<br />

HCV genotypes display significant differences in their<br />

global distribution and prevalence, making genotyping a<br />

useful method for determining the source of HCV transmission<br />

in an infected localized population (Hnatyszyn,<br />

2005).<br />

Abbreviations: HCV, Hepatitis c virus; WHO, world health<br />

organization; ORF, open reading frame; RNA, ribonucleic acid;<br />

PCR, polymerase chain reaction; EDTA,<br />

ethylenediaminetetraacetic acid; HCC, hepatocellular<br />

carcinoma.<br />

HCV genotype 1, 2 and 3 are distributed worldwide and<br />

their relative prevalence varies from one geographic area<br />

to another, whereas genotype 4 is predominantly prevalent<br />

in the Middle East and Africa, genotype 5 in South<br />

Africa and genotype 6 in Southeast Asia (Zein, 2000;<br />

Gish and Lua, 1997). According to the recent studies,<br />

genotype 4 is predominant in Egypt, 4 and 1 in Kuwait<br />

and Syria, and genotype 1 in Lebanon, Iraq and Iran<br />

(Osaba, 2002; Pacsa et al., 2001; Zali et al., 2000). Saudi<br />

Arabia shows an intermediate endemicity for HCV.<br />

Seroprevalence rates ranging from 0.9 to 5% have been<br />

reported among children and adults, respectively<br />

(Shobokshi et al., 1999). Some studies have indicated<br />

the predominance of genotype 4 in this region (Mellor et<br />

al., 1995; Al-Faleh et al., 1995). Hence HCV subtype<br />

distribution is needed to provide clues for studying<br />

epidemiology, the mode of transmission, and response to<br />

treatment. The objective of this study was to determine<br />

the distribution of HCV genotypes among the patients<br />

attending different hospitals and polyclinics in Riyadh,<br />

Saudi Arabia.<br />

MATERIALS AND METHODS<br />

This was a descriptive study conducted from January 2008 to<br />

December 2010 from different hospitals as well as polyclinics at<br />

Advanced Cell laboratory, Riyadh, Saudi Arabia. The latter is a<br />

reference laboratory serving a number of hospitals and polyclinics<br />

in Riyadh. Patients positive for anti-HCV antibodies were referred


Table 1. Genotype distribution of 358 HCV positive patients.<br />

Nationality Total<br />

HCV genotype<br />

Mohammed 2389<br />

1 1a 1b 2a 3 3a 4<br />

Saudi 208 - 6 7 5 8 - 182<br />

Egyption 98 13 - - - 8 - 79<br />

Pakistan 33 - - - - 26 7 -<br />

Others 17 - 8 5 - - - 4<br />

Total (%) 358 (100) 13 (3.6) 14 (3.9) 12 (3.3) 5 (1.4) 42 (11.7) 7 (1.9) 265 (74.2)<br />

Table 2. HCV Genotype among male and female.<br />

Gender<br />

1 1a 1b<br />

HCV genotype<br />

2a 3 3a 4<br />

Total (%)<br />

Male 13 3 2 1 36 7 178 240 (67)<br />

Female 0 11 10 4 6 0 87 118 (33)<br />

to the reference laboratory for detection of HCV RNA level and for<br />

genotyping. The study was designed to include patient's demographics<br />

(age, sex and nationality) as well. Detection of HCV<br />

ribonucleic acid (RNA) was carried out on a 500 ul sample of each<br />

serum sample positive for anti-HCV antibodies, using a commercial<br />

polymerase chain reaction (PCR) -based test (Taqman amplicor,<br />

Roche, USA) and following manufacturer’s instructions. Internal<br />

control supplied by the manufacturer was added to each specimen,<br />

as an extraction and amplification control. HCV positive clinical<br />

specimens (serum, ethylenediaminetetraacetic acid) (EDTA) plasma<br />

with viral loads >2,000 IU/ml were selected for genotyping using<br />

Versant HCV genotype assay (LiPA) 2.0 (Innogenetics, Siemens<br />

Healthcare Diagnostics, USA). This kit allows an improved and<br />

more accurate distinction between HCV genotype 1 and subtypes c<br />

to l of genotype 6 as well as between subtypes a and b of genotype<br />

1. Data was analyzed using SPSS-14 version. A p Value of < 0.5<br />

was considered significant for statistical analysis.<br />

RESULTS<br />

Genotype distribution in 358 HCV-positive patients is<br />

shown in Table 1. Overall, HCV genotype 4 was the most<br />

predominant genotype (74.2%) followed by genotype 3<br />

(11.7%) and 1a (3.9%). Differences in genotype distribution<br />

were statistically significant (p


2390 Afr. J. Microbiol. Res.<br />

Saudi Arabian patients can be attributed to many factors.<br />

These include the expatriates from different nationalities<br />

resided in Saudi Arabia for quite some time and participated<br />

in blood donation.<br />

According to the World Health Organization, 180 million<br />

individuals in the world are infected with HCV and this is<br />

a growing global problem. The development of an<br />

effective vaccine remains the ideal way to combat HCV<br />

infection. In addition to the implications for clinical outcome<br />

of infection, and for treatment, genotyping of HCV<br />

also has major implications for HCV vaccine development.<br />

Recent data suggest that for a vaccine to be fully<br />

protective it should contain a range of deferent envelope<br />

proteins corresponding to the common genotypes in<br />

particular geographic regions. Vaccines for use in the<br />

Middle East should, therefore, not be based only on<br />

genotype 4 sequences; other genotypes such as 1a and<br />

1b are also equally important. Finally, genotyping of HCV<br />

may be a useful epidemiological marker particularly in<br />

establishing suspected unconventional routes of HCV<br />

transmission such as vertical, intraspousal or interfamilial<br />

transmission (Alfaresi, 2011).<br />

ACKNOWLEDGMENT<br />

The author would like to thank all the staff at Research<br />

center, college of applied medical sciences, King Saud<br />

University for their valuable support.<br />

REFERENCES<br />

Al-Faleh FZ, Ramia S, Arif M, Ayoola EA, Al-Rashed RS, Al-Jeffry M,<br />

Hossain A, El-Hazmi M (1995). Profile of hepatitis C virus (HCV) and<br />

the possible modes of transmission of the virus in Gizan, area of<br />

Saudi Arabia: a community-based study. Ann. Trop. Med. Parasitol.,<br />

89(4): 431-437.<br />

Alfaresi MS (2011). Prevalence of hepatitis C virus (HCV) genotypes<br />

among positive UAE patients. Mol. Biol. Rep., 38(4): 2719-2722.<br />

Bdour S (2002). Hepatitis C virus infection in Jordanian haemodialysis<br />

units, serological diagnosis and genotyping. J. Med. Microbiol., 51(8):<br />

700-704.<br />

Choo QL, Kuo G, Weiner AJ, Overby LR, Bradley DW, Houghton M<br />

(1989). Isolation of a cDNA clone derived from a bloodborne non-A,<br />

non-B hepatitis genome. Science, 244(4902): 359-362.<br />

Ghany MG (2009). Diagnosis, management, and treatment of hepatitis<br />

C: An update. Hepatology, 49(4): 1335-1374.<br />

Gish RG, Lua JYN (1997). Hepatitis C virus: eight years old. Viral Hep.<br />

Rev., 3: 17-37.<br />

Hnatyszyn HJ (2005). Chronic hepatitis C and genotyping: the clinical<br />

significance of determining HCV genotypes. Antivir. Ther., 10(1): 1-<br />

11.<br />

Idrees M, Riazuddin S (2008). Frequency distribution of hepatitis C virus<br />

genotypes in different geographical regions of Pakistan and their<br />

possible routes of transmission. BMC Infect. Dis., 23(8): 69-78.<br />

Lavanchy D (2009). The global burden of hepatitis C. Liver Int., 29(1):<br />

74-81.<br />

McHutchison JG, Gordon SC, Schiff ER, Shiffman ML, Lee WM, Rustgi<br />

VK (1998). Interferon alfa-2b alone or in combination with ribavirin as<br />

initial treatment for chronic hepatitis C. Hepatitis Interventional<br />

Therapy Group. N. Eng. J. Med., 339(21): 1485-1492.<br />

Mellor J, Holmes EC, Jarvis LM, Yap PL, Simmonds P (1995).<br />

Investigation of the pattern of hepatitis C virus sequence diversity in<br />

different geographical regions: implications for virus classification.<br />

The international HCV collaborative study group. J. Gen. Virol., 76(Pt<br />

10): 2493-2507<br />

Mujeeb SA (2002). HCV 3 in Pakistan: does it offer more hope for cure<br />

or control. J. Pak. Med. Assoc., 52: 191-192.<br />

Osaba AO (2002). Hepatitis C virus genotypes in Saudi Arabia. Saudi<br />

Med. J., 23(1): 7-12.<br />

Pacsa AS, Al-Mufti S, Chugh TD, Said-Adi G (2001). Genotypes of<br />

Hepatitis C virus in Kuwait. Med. Principles Pract., 10: 55-57.<br />

Ray SC, Arthur RR, Carella A, Bukh J, Thomas DL (2000). Genetic<br />

Epidemiology of hepatitis C throughout Egypt. J. Infect. Dis., 182(3):<br />

239-244.<br />

Shobokshi OA, Serebour FE, Skakni L, Al-Saffy YH, Ahdal MN (1999).<br />

Hepatitis C genotypes and subtypes in Saudi Arabia. J. Med. Virol.,<br />

58(1): 44-48.<br />

Tokita H, Okamoto H, Luengrojanakul P, Vareesangthip K, Chainuvati<br />

T, Iizuka H, Tsuda F, Miyakawa Y, Mayumi M (1995). Hepatitis C<br />

virus variants from Thailand classifiable into five novel genotypes in<br />

the sixth (6b), seventh (7c, 7d) and ninth (9b, 9c) major genetic<br />

groups. J. Gen. Virol., 76(Pt 9): 2329-2335.<br />

Zali MR, Mayumi M, Rouf M, Noowroozi A (2000). Hepatitis C virus<br />

genotypes in the Islamic Republic of Iran: a preliminary study.<br />

Eastern Mediterr. Health J., 6: 372-377.<br />

Zein NN (2000). Clinical significance of hepatitis C virus genotypes.<br />

Clin. Microbiol. Rev., 13(2): 223-235.<br />

Zekri AR, Bahnassy AA, Shaarawy SM, Mansour OA, Maduar MA,<br />

Khaled HM, El-Ahmadi O (2000). Hepatitis C virus genotyping in<br />

relation to neu-oncoprotein overexpression and the development of<br />

hepatocellular carcinoma. J. Med. Microbiol., 49(1): 89-95.


UPCOMING CONFERENCES<br />

7th Conference of the World Mycotoxin Forum and<br />

XIIIth IUPAC International Symposium on Mycotoxins and Phycotoxins,<br />

Rotterdam, Netherlands, 5 Nov 2012<br />

13th International Congress on Yeasts, Madison, USA,<br />

26-30 Aug 2012


Conferences and Advert<br />

August 2012<br />

13th International Congress on Yeasts, Madison, USA, 26 Aug 2012<br />

September 2012<br />

6th International Congress on Biocatalysis, Hamburg, Germany, 2 Sep 2012<br />

9th International Congress on Extremophiles, Sevilla, Spain, 10 Sep 2012<br />

October 2012<br />

Actinobacteria within Soils: Capacities for Mutualsim, Symbiosis and Pathogenesis,<br />

Muenster, Germany, 25 Oct 2012<br />

November 2012<br />

7th Conference of the World Mycotoxin Forum and XIIIth IUPAC International<br />

Symposium on Mycotoxins and Phycotoxins, Rotterdam, Netherlands, 5 Nov 2012<br />

December 2012<br />

Marine Microbiology and Biotechnology: Biodiscovery, Biodiversity and<br />

Bioremediation, Cork, Ireland, 19 Dec 2012<br />

International Conference on Pathology and Microbiology, Bangkok, Thailand, 26 Dec<br />

2012


African Journal of<br />

Microbiology Research<br />

Related <strong>Journals</strong> Published by <strong>Academic</strong> <strong>Journals</strong><br />

■ Journal of Cell and Animal Biology<br />

■ African Journal of Biotechnology<br />

■ Biotechnology and Molecular Biology Reviews<br />

■ African Journal of Microbiology Research<br />

■ African Journal of Biochemistry Research<br />

■ Journal of Bacteriology Research

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!