27.02.2013 Views

project honey as antimicrobial agent final full.pdf - University of ...

project honey as antimicrobial agent final full.pdf - University of ...

project honey as antimicrobial agent final full.pdf - University of ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

Honey <strong>as</strong> an <strong>antimicrobial</strong> <strong>agent</strong> against<br />

multi-drug resistant Gram negative<br />

bacterial rods<br />

By<br />

Rahma Ali Saleh Al-Maaini<br />

This dissertation is submitted in partial fulfilment <strong>of</strong> the<br />

degree <strong>of</strong> MPhil in Biomedical Sciences<br />

(Microbiology)<br />

School <strong>of</strong> Health Sciences<br />

<strong>University</strong> <strong>of</strong> Wales Institute Cardiff, UK<br />

May 2011<br />

1


Contents<br />

Title I<br />

Contents II<br />

Declaration XI<br />

Dedication XII<br />

Acknowledgments XIII<br />

Poster Presentation XIV<br />

Index <strong>of</strong> Tables XV<br />

Index <strong>of</strong> Figures XIX<br />

Abbreviations XXVI<br />

Abstract XXX<br />

Chapter 1: Introduction 1<br />

1.1 Antimicrobial Resistance: 2<br />

1.2 Extended Spectrum Beta-Lactam<strong>as</strong>es (ESBLs): 3<br />

1.2.1 Cl<strong>as</strong>sification <strong>of</strong> ESBL: 4<br />

1.2.1.1 Functional Cl<strong>as</strong>sification: 4<br />

1.2.1.2 Molecular Cl<strong>as</strong>sification: 4<br />

1.2.2 Types <strong>of</strong> ESBL: 5<br />

1.2.2.1 TEM-type ESBLs (cl<strong>as</strong>s A) 5<br />

1.2.2.2 SHV-type ESBLs (cl<strong>as</strong>s A)<br />

1.2.2.3 CTX-M type ESBLs (cl<strong>as</strong>s A): 6<br />

1.2.2.4 OXA-type ESBLs (cl<strong>as</strong>s D) 7<br />

1.2.2. 5 AmpC-type ESBLs (cl<strong>as</strong>s C) 7<br />

1.2.2.6 Carbapenem<strong>as</strong>es (cl<strong>as</strong>s A, B, D) 8<br />

2<br />

6


1.2.3 Emergence <strong>of</strong> ESBL: 9<br />

1.2.4 Risk factor for ESBL: 9<br />

1.2.5 Treatment <strong>of</strong> ESBLs: 10<br />

1.3 Acinetobacter: 10<br />

1.3.1 Taxonomy and Historical Features <strong>of</strong> Acinetobacter: 11<br />

1.3.2 Laboratory Diagnosis: 12<br />

1.3.3 Clinical features <strong>of</strong> Acinetobacter infections: 12<br />

1.3.4 Pathogenesis <strong>of</strong> Acinetobacter infection: 14<br />

1.3.4.1 The production <strong>of</strong> exopolysaccharide: 15<br />

1.3.4.2 Quorum-sensing: 15<br />

1.3.4.3 The property <strong>of</strong> adhesion to human epithelial cells via<br />

the capsule or fimbriae.<br />

1.3.4.4 Surface and mitochondrial porins: 16<br />

1.3.5 Emergence <strong>of</strong> Resistance: 17<br />

1.3.6 Carbapenem action on Acinetobacter: 18<br />

1.3.7 Mechanisms <strong>of</strong> Carbapenem Resistance: 18<br />

1.3.8 Acinetobatcer Treatment with Honey: 20<br />

1.4 Alternative Antimicrobial Therapies: 21<br />

1.4.1 Ancient Use <strong>of</strong> Honey <strong>as</strong> a Medicine: 22<br />

1.4.2 Honey <strong>as</strong> a Modern Medicine: 22<br />

1.4.3 Antimicrobial Activity <strong>of</strong> Honey: 24<br />

1.4.4 Chemical composition <strong>of</strong> <strong>honey</strong>: 26<br />

3<br />

15


1.4.5 Factors Contributing Antibacterial Properties <strong>of</strong> Honey: 27<br />

1.4.5.1 Osmotic effect: 28<br />

1.4.5.2 Acidity 28<br />

1.4.5.3 Hydrogen peroxide production 29<br />

1.4.5.4 Non-peroxide Components 30<br />

1.4.5.5 Antioxidant activity: 31<br />

1.4.5 Manuka Honey: 34<br />

1.4.6 Omani Honey: 36<br />

1.5 Aims and Objectives: 39<br />

Chapter 2: Materials & Methods: 40<br />

2.1 Characterization <strong>of</strong> test organisms: 49<br />

2.1.1 Confirmation <strong>of</strong> the identity <strong>of</strong> test organisms: 49<br />

2.1.2 Antibiotics sensitivities: Extended Spectrum Beta- lactam<strong>as</strong>es<br />

test (ESBLs):<br />

2.1.2.1 Inoculum preparation: 49<br />

2.1.2.2 Disc application: 49<br />

2.1.2.3 Screening test for ESBL: 50<br />

2.1.2.4 Phenotypic confirmatory test for ESBL: 50<br />

2.1.2.5 ESBL/AmpC * confirmation test: 50<br />

2.2 Characterization <strong>of</strong> <strong>honey</strong> samples: 51<br />

4<br />

49


2.2.1 Honey sample collection: 51<br />

2.2.2 Bio<strong>as</strong>say <strong>of</strong> antibacterial activity: 51<br />

2.2.2.1 Phenol standards preparation for bio<strong>as</strong>say: 52<br />

2.2.2.3 Inoculum preparation: 52<br />

2.2.2.4 Plate preparation: 52<br />

2.2.2.5 Sample preparation: 53<br />

2.2.2.6 Samples and standards application: 53<br />

2.2.2.7 Zone me<strong>as</strong>urement: 54<br />

2.2.2.8 Calculation <strong>of</strong> antibacterial activity <strong>of</strong> <strong>honey</strong>: 54<br />

2.2.3 Determination <strong>of</strong> pH: 55<br />

2.2.4 Sugar and water content: 55<br />

2.2.5 Hydroxymethylfurfural (HMF) concentration: 56<br />

2.2.6 Protein content: 56<br />

2.2.7 Colour: 58<br />

2.2.8 Pollen analysis: 58<br />

2.2.9 Total phenolic content: 59<br />

2.2.9.1 Re<strong>agent</strong>/standard preparation: 59<br />

2.2.9.2 Assay method: 60<br />

2.2.10 Free radical activity <strong>of</strong> <strong>honey</strong>: 61<br />

2.2.10.1 Re<strong>agent</strong> / Standard Preparations 61<br />

5


2.2.10.2 Assay procedure: 62<br />

2.3 Determination <strong>of</strong> antibacterial activity <strong>of</strong> <strong>honey</strong> samples against test<br />

cultures:<br />

2.3.1 Minimum Inhibitory Concentration (MIC) method: 62<br />

2.3.1.1 Agar incorporation method: 62<br />

2.3.1.1.1 Honey selection: 62<br />

2.3.1.1.2 Inoculum preparation: 63<br />

2.3.1.1.3 Plates preparation: 63<br />

2.3.1.1.4 Honey preparation: 63<br />

2.3.1.1.5 Plates reading (MIC determination): 64<br />

2.3.1.2 Broth dilution method:<br />

2.3.1.2.1 Honey selection: 65<br />

2.3.1.2.2 Honey dilution: 65<br />

2.3.2.1.3 Microtitre plate inoculation: 65<br />

2.3.2.1.4 Visual inspection <strong>of</strong> MIC: 66<br />

2.3.2.1.5 Spectrophotometric determination <strong>of</strong> MIC: 66<br />

2.3.2 Minimum Bactericidal concentration (MBC) method: 66<br />

2.4 Time Kill Curve Assay: 67<br />

2.4.1 Strain selection: 67<br />

2.4.2 Time-kill curve: 67<br />

6<br />

62


2.5 Effect <strong>of</strong> <strong>honey</strong> on bacterial structure: 68<br />

2.5.1 Scanning Electron Microscopy (SEM): 68<br />

2.5.1.1 Bacterial Selection for SEM 68<br />

2.5.1.2 Preparation <strong>of</strong> cells in the exponential ph<strong>as</strong>e <strong>of</strong> growth: 69<br />

2.5.1.3 Preparation <strong>of</strong> cells for scanning electron microscopy: 69<br />

2.5.2 Transmission Electron Microscopy (TEM): 70<br />

2.5.2.1 Bacterial Selection for TEM 70<br />

2.5.2.2 Preparation <strong>of</strong> cells for transmission electron<br />

microscopy:<br />

2.5.2.3 Pallet trimming and sectioning: 71<br />

2.5.2.4 Staining <strong>of</strong> thin sections 71<br />

2.6 Effect <strong>of</strong> <strong>honey</strong> on bacterial proteins: 72<br />

2.6.1 Two Dimensional Gel Electrophoresis: 72<br />

2.6.1.1 Buffers preparation: 73<br />

2.6.1.2 Cell preparation: 74<br />

2.6.1.3 Protein determination <strong>of</strong> Acinetobacter extracts for each<br />

extract:<br />

2.6.1.4 Rehydration and sample application: 76<br />

2.6.1.5 Rehydration in the PROTEAN ® IEF Focusing Tray: 76<br />

2.6.1.6 Equilibration and SDS-PAGE: 77<br />

7<br />

71<br />

74


2.6.1.7 Staining and gel visualising: 77<br />

2.7 Statistical analysis <strong>of</strong> the data: 78<br />

Chapter 3: Results 79<br />

3.1 Confirmations <strong>of</strong> the identity and antibiotics sensitivities <strong>of</strong> test<br />

organisms:<br />

3.2 Characterization <strong>of</strong> <strong>honey</strong> samples: 88<br />

3.2.1 Determination <strong>of</strong> antibacterial activity: 88<br />

3.2.2 Chemical & physical analysis <strong>of</strong> <strong>honey</strong> samples 91<br />

3.2.2.1 Pollen analysis 94<br />

3.2.2.2 Antioxidant activity <strong>as</strong>say: 96<br />

3.3 Determination <strong>of</strong> antibacterial activity <strong>of</strong> <strong>honey</strong> samples against test<br />

cultures:<br />

3.3.1 Minimum Inhibitory Concentration (MIC) and Minimum<br />

Bactericidal Concentration (MBC) <strong>of</strong> manuka <strong>honey</strong>:<br />

3.3.1.1 Agar incorporation method: 97<br />

3.3.1.2 Broth dilution method: 99<br />

3.3.2 Sensitivity <strong>of</strong> MDR and ESBLs to Omani <strong>honey</strong>: 107<br />

3.4 Time Kill Curves 115<br />

3.4.1 Inhibition <strong>of</strong> test organisms by manuka <strong>honey</strong> monitored by<br />

optical density:<br />

3.4.2 Inhibition <strong>of</strong> test organisms by manuka <strong>honey</strong> monitored by total 119<br />

8<br />

80<br />

97<br />

97<br />

115


viable count<br />

3.4.2.1 Acinetobacter spp 120<br />

3 3.4.2.2 E.coli 122<br />

3.4.2.3 Klebsiella 124<br />

3.4.2.4 Citrobacter 126<br />

3.4.2.5 Enterobacter 128<br />

3.4.2.6 Serratia 130<br />

3.5 Effect <strong>of</strong> <strong>honey</strong> on bacterial structure: 134<br />

3.5.1 Growth Curves: 134<br />

3.5.2 Scanning Electron Microscopy (SEM): 138<br />

3.5.2.1 SEM <strong>of</strong> Acinetobacter 138<br />

3.5.2.2 SEM <strong>of</strong> E.coli 148<br />

3.5.2.3 SEM <strong>of</strong> Klebsiella 151<br />

3.5.2.4 SEM <strong>of</strong> Enterobacter 160<br />

3.5.2.5 SEM <strong>of</strong> Citrobacter 165<br />

3.5.2.6 SEM <strong>of</strong> Serratia 170<br />

3.5.2 Transmission Electron Microscopy (TEM): 174<br />

3.5.2.1 TEM for Acinetobacter: 174<br />

3.5.2.2. TEM for E.coli: 179<br />

9


3.6 Effect <strong>of</strong> <strong>honey</strong> on bacterial proteins: 184<br />

3.6.1 Two Dimensional Gel Electrophoresis: 184<br />

Chapter 4: Discussion 186<br />

4.1 Antibacterial activity <strong>of</strong> <strong>honey</strong> samples: 187<br />

4.2. Chemical & physical analysis <strong>of</strong> <strong>honey</strong> samples 189<br />

4.3 Effect <strong>of</strong> <strong>honey</strong> samples against test cultures: 193<br />

4.4 Inhibition <strong>of</strong> test organisms by manuka <strong>honey</strong> using time kill curve <strong>as</strong>say 201<br />

4.5 Effect <strong>of</strong> <strong>honey</strong> on bacterial structure: 202<br />

4.6 Further investigations: 213<br />

Chapter 5: References 215<br />

10


Declaration<br />

This work h<strong>as</strong> not previously been accepted in substance for any degree and is<br />

not being concurrently submitted in candidature for any degree.<br />

Signed ...................................................................... (candidate)<br />

Date ..........................................................................<br />

STATEMENT 1<br />

This thesis is the result <strong>of</strong> my own investigations, except where otherwise<br />

stated. Where correction services have been used, the extent and nature <strong>of</strong> the<br />

correction is clearly marked in a footnote(s).<br />

Other sources are acknowledged by footnotes giving explicit references. A<br />

bibliography is appended<br />

Signed ..................................................................... (candidate)<br />

Date .........................................................................<br />

STATEMENT 2<br />

I hereby give consent for my thesis, if accepted, to be available for<br />

photocopying and for inter-library loan, and for the title and summary to be<br />

made available to outside organisations.<br />

Signed ..................................................................... (candidate)<br />

Date .........................................................................<br />

I hereby give consent for my thesis, if accepted, to be available for<br />

photocopying and for inter-library loans after expiry <strong>of</strong> a bar on access<br />

approved by UWIC.<br />

Signed ..................................................................... (candidate)<br />

Date .............................<br />

11


Dedication<br />

This <strong>project</strong> is lovingly dedicated to my dear parents whose<br />

legacy. I will tre<strong>as</strong>ure throughout my life. They thought me to<br />

strive and do my best in all things that I undertake. They are<br />

indeed my inspiration in doing my best in this endeavour. I<br />

thank them for bringing me up the best way they could. I <strong>full</strong>y<br />

dedicate this humble accomplishment <strong>of</strong> mine to them.<br />

12


Acknowledgments<br />

I wish to express my special and sincere thanks to my supervisor Pr<strong>of</strong>. Rose Cooper<br />

for all her help, supervision, guidance and valuable suggestions during the execution<br />

<strong>of</strong> this <strong>project</strong>.<br />

I owe my deepest gratitude to Mr. John Philips (dean <strong>of</strong> international <strong>of</strong>fice- UWIC)<br />

to <strong>of</strong>fer me a research scholarship.<br />

I wish also to record my grateful thanks to Mr. Leighton Jenkins for his support and<br />

guidance throughout the practical <strong>as</strong>pects <strong>of</strong> this dissertation. Special thanks to Dr.<br />

Hann (Cardiff <strong>University</strong>- Cardiff) and Mr. Issa Al-Amri (Sultan Qaboos <strong>University</strong>-<br />

Oman) for their <strong>as</strong>sistant in using the facility <strong>of</strong> electron microscopy at their<br />

Universities.<br />

Great thanks to Dr. Charles Bakheit (Associate Pr<strong>of</strong>. in Statistics- SQU) for his help<br />

in statistical analysis.<br />

I would like to express my appreciation to my family who support and encourage me<br />

to do the best in writing this dissertation. Special thanks to my friends Shafiqa,<br />

Suhaila, Kulthom and Rahima.<br />

13


Presented posters<br />

Al-Maaini, R. A, Cooper, R. and Burton, N. „Honey <strong>as</strong> an <strong>antimicrobial</strong> <strong>agent</strong> against<br />

multi-drug resistant Gram negative bacterial rods‟, poster presented in the Society for<br />

Applied Microbiology (SfAM) 2010 Summer Meeting, Brighton, 5-8 July 2010.<br />

14


Index <strong>of</strong> Tables<br />

Tables Title Page<br />

Table 1.1 Honey composition 26<br />

Table 1.2 Comparisons between peroxide and non-peroxide <strong>honey</strong> 33<br />

Table 2.1 Media, Chemicals and Re<strong>agent</strong>s used for general experiment<br />

in the <strong>project</strong><br />

Table 2.2 Chemicals and Re<strong>agent</strong>s used for electron microscopy<br />

experiment in the <strong>project</strong><br />

Table 2.3 Chemicals and Re<strong>agent</strong>s used for 2- dimensional<br />

electrophoresis experiment in the <strong>project</strong><br />

Table 2.4 Equipment used in general experiments in the <strong>project</strong> 44<br />

Table 2.5 Equipment used in electron microscopy and 2-dimentional<br />

Table 2.6<br />

electrophoresis experiments in the <strong>project</strong><br />

Thirty isolates <strong>of</strong> MDR Acinetobacter and their resistance<br />

pattern<br />

Table 2.7a Clinical isolates <strong>of</strong> E.coli & Klebsiella provided for this<br />

study<br />

Table 2.7b Clinical isolates <strong>of</strong> Citrobacter, Enterobacter & Serratia<br />

provided for this study<br />

Table 2.8 List <strong>of</strong> <strong>honey</strong>s used in this <strong>project</strong> 51<br />

Table 2.9 Preparation <strong>of</strong> phenol standards 52<br />

Table 2.10 Preparation <strong>of</strong> gallic acid standard solutions 60<br />

Table 2.11 Preparation <strong>of</strong> varying concentration <strong>of</strong> <strong>honey</strong> solution from<br />

40% (w/v) stock <strong>honey</strong> for MIC method<br />

Table 2.12 Preparation <strong>of</strong> tubes for MICs 65<br />

Table 2.13: Cultures and <strong>honey</strong> concentrations used in the time-kill<br />

curves<br />

15<br />

41<br />

42<br />

43<br />

45<br />

46<br />

47<br />

48<br />

64<br />

67


Index <strong>of</strong> Tables (Continued)<br />

Tables Title Page<br />

Table 2.14 Re<strong>agent</strong>s and buffers used in 2-D Electrophoresis 73<br />

Table 2.15: Quantity <strong>of</strong> proteins in Acinetobacter cells with and without<br />

Table 3.1 a<br />

& b:<br />

20% <strong>honey</strong><br />

Identification and antibiotics sensitivities <strong>of</strong> 30 MDR<br />

Acinetobacter isolates<br />

Table 3.2: Confirmation <strong>of</strong> identity and antibiotics sensitivity including<br />

ESBL tests for 10 E.coli isolates.<br />

Table 3.3: Confirmation <strong>of</strong> identity and antibiotics sensitivity including<br />

ESBL tests for 12 Klebsiella isolates<br />

Table 3.4: Confirmation <strong>of</strong> identity and antibiotics sensitivity including<br />

Table 3.5:<br />

ESBL tests for 12 Citrobacter isolates.<br />

Confirmation <strong>of</strong> identity and antibiotics sensitivity including<br />

ESBL tests for15 Enterobacter isolates.<br />

Table 3.6: Confirmation <strong>of</strong> identity and antibiotics sensitivity including<br />

ESBL tests for 8 Serratia isolates.<br />

Table 3.7: Antibacterial activity <strong>of</strong> <strong>honey</strong> samples 91<br />

Table 3.8: Chemical & physical analysis <strong>of</strong> different types <strong>of</strong> Omani<br />

<strong>honey</strong> compared to manuka <strong>honey</strong><br />

Table 3.9: Represent the identification <strong>of</strong> flora sources by pollen<br />

analysis<br />

Table 3.10: Amount <strong>of</strong> free radical and phenolic content in each <strong>honey</strong><br />

samples<br />

16<br />

75<br />

81-<br />

82<br />

83<br />

84<br />

85<br />

86<br />

87<br />

93<br />

94<br />

96


Index <strong>of</strong> Tables (Continued)<br />

Tables Title Page<br />

Table 3.11: Susceptibility <strong>of</strong> Acinetobacter isolates against manuka<br />

<strong>honey</strong><br />

Table 3.12: Sensitivity <strong>of</strong> 30 Acinetobacter isolates treated with manuka<br />

<strong>honey</strong> by broth dilution method<br />

Table 3.13: Sensitivity <strong>of</strong> 10 Klebsiella isolates to manuka <strong>honey</strong> tested<br />

by the broth dilution method<br />

Table 3.14: Sensitivity <strong>of</strong> 8 Serratia and 8 E.coli isolates to manuka<br />

<strong>honey</strong> using a broth dilution method<br />

Table 3.15: Susceptibility <strong>of</strong> 15 Enterobacter isolates to with manuka<br />

<strong>honey</strong> determined by broth dilution method<br />

Table 3.16: Susceptibility <strong>of</strong> 12 Citrobacter isolates to manuka <strong>honey</strong> 106<br />

Table 3.17: Susceptibility <strong>of</strong> 30 MDR Acinetobacter isolates against 4<br />

types <strong>of</strong> Omani <strong>honey</strong> using broth dilution method<br />

Table 3.18: Susceptibility <strong>of</strong> 12 Klebsiella isolates against 4 types <strong>of</strong><br />

Omani <strong>honey</strong> using broth dilution method<br />

Table 3.19: Susceptibility <strong>of</strong> 10 E.coli isolates against 4 types <strong>of</strong> Omani<br />

<strong>honey</strong> using broth dilution method<br />

Table 3.20: Susceptibility <strong>of</strong> 15 Enterobacter isolates against 4 types <strong>of</strong><br />

Omani <strong>honey</strong> using broth dilution method<br />

Table 3.21: Susceptibility <strong>of</strong> 12 Citrobacter isolates against 4 types <strong>of</strong><br />

Omani <strong>honey</strong> using broth dilution method<br />

Table 3.22: Susceptibility <strong>of</strong> 8 Serratia isolates against 4 types <strong>of</strong> Omani<br />

<strong>honey</strong> using broth dilution method<br />

Table 3.23: Cultures and <strong>honey</strong> concentrations used in the time-kill<br />

curves <strong>as</strong>say<br />

17<br />

98<br />

100<br />

102<br />

103<br />

105<br />

107-<br />

108<br />

109<br />

110<br />

111<br />

112<br />

113<br />

115


Index <strong>of</strong> Tables (Continued)<br />

Tables Title Page<br />

Table 3.24: Decimal reduction dose (DRD) for each isolate after 5 h<br />

exposure to 2x respective MICs <strong>of</strong> manuka <strong>honey</strong><br />

Table 3.25 Comparison <strong>of</strong> the mean viable cell count between non<br />

<strong>honey</strong> and <strong>honey</strong> treated cells using paired sample test (T-<br />

test).<br />

Table 3.26: Comparison <strong>of</strong> changes in cell sizes <strong>of</strong> isolates observed in<br />

scanning electron microscopy between untreated and <strong>honey</strong><br />

treated cells (P value) (Mann-Whitney Test)<br />

Table 4.1: The physicochemical analysis <strong>of</strong> selected <strong>honey</strong>s tested 190<br />

Table 4.2: Comparison between previous studies and current study on<br />

MIC <strong>of</strong> different <strong>honey</strong>s including manuka <strong>honey</strong> against six<br />

bacteria species.<br />

Table 4.3: Summary <strong>of</strong> the growth inhibition, killing rate and ultra-<br />

structure changes in EM for six species selected after<br />

exposure to 2x MIC (%w/v) <strong>of</strong> manuka <strong>honey</strong>:<br />

18<br />

132<br />

133<br />

173<br />

196-<br />

197<br />

208


Index <strong>of</strong> Figures<br />

Figure Title Page<br />

Figure 2.1: Calibration curve for protein determination in <strong>honey</strong><br />

samples<br />

Figure 2.2: Standard curve for total phenolic content in <strong>honey</strong> samples 60<br />

Figure 2.3: Calibration curve <strong>of</strong> protein concentration in Acinetobacter<br />

with and without 20% manuka <strong>honey</strong>.<br />

Figure 3.1: A typical <strong>honey</strong> bio<strong>as</strong>say plate. 88<br />

Figure 3.2: A typical calibration curve <strong>of</strong> the bio<strong>as</strong>say 90<br />

Figure 3.3: Image <strong>of</strong> pollen present in Omani <strong>honey</strong> samples at 100x<br />

magnification.<br />

Figure 3.4: The effect <strong>of</strong> manuka <strong>honey</strong> on the growth <strong>of</strong> Acinetobacter 116<br />

Figure 3.5: The effect <strong>of</strong> manuka <strong>honey</strong> on the growth <strong>of</strong> E.coli 116<br />

Figure 3.6: The effect <strong>of</strong> manuka <strong>honey</strong> on the growth <strong>of</strong> Klebsiella 117<br />

Figure 3.7: The effect <strong>of</strong> <strong>honey</strong> on the growth <strong>of</strong> Citrobacter 117<br />

Figure 3.8: The effect <strong>of</strong> <strong>honey</strong> on the growth <strong>of</strong> Enterobacter 118<br />

Figure 3.9: The effect <strong>of</strong> <strong>honey</strong> on the growth <strong>of</strong> Serratia 118<br />

Figure 3.10: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong><br />

Acinetobacter.<br />

Figure 3.11: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> E.coli 123<br />

Figure 3.12: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> Klebsiella 125<br />

Figure 3.13: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> Citrobacter 127<br />

Figure 3.14: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> Enterobacter 129<br />

19<br />

58<br />

75<br />

95<br />

121


Index <strong>of</strong> Figures (Continued)<br />

Figure Title Page<br />

Figure 3.15: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> Serratia<br />

Figure 3.16: Growth curve <strong>of</strong> Acinetobacter in ISB 135<br />

Figure 3.17: Growth curve <strong>of</strong> E.coli in ISB 135<br />

Figure 3.18: Growth curve <strong>of</strong> Klebsiella in ISB 136<br />

Figure 3.19: Growth curve <strong>of</strong> Enterobacter in ISB 136<br />

Figure 3.20: Growth curve <strong>of</strong> Citrobacter in ISB 137<br />

Figure 3.21: Growth curve <strong>of</strong> Serratia in ISB 137<br />

Figure 3.22: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after 0<br />

minutes at x5,000 magnification<br />

Figure 3.23: SEM micrograph <strong>of</strong> Acinetobacter cells exposed to 20%<br />

(w/v) manuka <strong>honey</strong> <strong>of</strong> 0 minutes at x5,000 magnification<br />

Figure 3. 24: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after<br />

60 minutes at x5,000 magnification<br />

Figure 3.25: SEM micrograph <strong>of</strong> Acinetobacter cells exposed to 20%<br />

(w/v) manuka <strong>honey</strong> after 60 minutes at x5,000<br />

magnification<br />

Figure 3.26: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after<br />

90 minutes at x5,000 magnification<br />

Figure 3.27: SEM micrograph <strong>of</strong> Acinetobacter cells exposed to 20%<br />

(w/v) manuka <strong>honey</strong> after 90 minutes at x5,000<br />

magnification<br />

Figure 3.28: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after<br />

150 minutes at x5,000 A & x20,000 B magnification<br />

respectively<br />

20<br />

131<br />

140<br />

140<br />

141<br />

141<br />

142<br />

142<br />

143


Index <strong>of</strong> Figures (Continued)<br />

Figure Title Page<br />

Figure 3.29: SEM micrographs <strong>of</strong> Acinetobacter cells exposed to 20%<br />

(w/v) manuka <strong>honey</strong> after 150 minutes at x5,000A,<br />

x10,000B, x20,000C & x25,000D magnification<br />

respectively<br />

Figure 3.30: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after<br />

180 minutes at x5,000 A & x10,000 B magnification<br />

respectively<br />

Figure 3.31: SEM micrographs <strong>of</strong> Acinetobacter cells exposed to 20%<br />

(w/v) manuka <strong>honey</strong> after 180 minutes at x 5,000 (A) &<br />

(B) magnification respectively<br />

Figure 3.32: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> E.coli after 30<br />

minutes at x5,000 magnification<br />

Figure 3.33: SEM micrograph <strong>of</strong> E.coli cells exposed to 30% (w/v)<br />

manuka <strong>honey</strong> after 30 minutes at x5,000 magnification<br />

Figure 3.34 : SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> E.coli after 180<br />

minutes at x5,000 magnification<br />

Figure 3.35 : SEM micrograph <strong>of</strong> E.coli cells exposed to 30% (w/v)<br />

manuka <strong>honey</strong> after 180 minutes at x5,000 magnification<br />

Figure 3.36: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Klebsiella after 30<br />

minutes at x5,000 (A) & 15,000 (B) magnification<br />

respectively<br />

Figure 3.37: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Klebsiella after 180<br />

minutes at x5,000 (A) & 15,000 (B) magnification<br />

respectively<br />

21<br />

144<br />

146<br />

147<br />

149<br />

149<br />

150<br />

150<br />

152<br />

153


Index <strong>of</strong> Figures (Continued)<br />

Figure Title Page<br />

Figure 3.38 : SEM micrographs <strong>of</strong> Klebsiella cells exposed to 30%<br />

manuka <strong>honey</strong> after 30 minutes at x5,000 (A) & 15,000 (B)<br />

magnification respectively<br />

Figure 3.39: SEM micrographs <strong>of</strong> Klebsiella cells exposed to 30%<br />

(w/v) manuka <strong>honey</strong> after 180 minutes at x5,000A,<br />

x15,000B,C & x20,000D magnification<br />

Figure 3.40: SEM micrographs <strong>of</strong> Klebsiella cells exposed to 40%<br />

(w/v) Omani <strong>honey</strong> after 30 minutes at x5,000 A &<br />

x15,000 B magnification respectively<br />

Figure 3.41 : SEM micrographs <strong>of</strong> Klebsiella cells exposed to 40%<br />

(w/v) Omani <strong>honey</strong> after 180 minutes at x5,000 A & x<br />

20,000 B,C & D magnification respectively<br />

Figure 3.42: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Enterobacter after<br />

30 minutes at x5,000 A & x15,000 B magnification<br />

respectively<br />

Figure 3.43: SEM micrographs <strong>of</strong> Enterobacter cells exposed to 30%<br />

(w/v) manuka <strong>honey</strong> after 30 minutes at x5,000 A & B<br />

magnification respectively<br />

Figure 3.44: SEM micrographs <strong>of</strong> untreated Enterobacter cells after 180<br />

minutes at x5,000 A & x20,000 B magnification<br />

respectively<br />

Figure 3.45: SEM micrographs <strong>of</strong> Enterobacter cells exposed to 30%<br />

(w/v) manuka <strong>honey</strong> after 180 minutes at x5,000 (A & B)<br />

magnification respectively<br />

Figure 3.46: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Citrobacter after 30<br />

minutes at x5,000 A & x15,000 B magnification<br />

respectively<br />

22<br />

154<br />

155<br />

157<br />

158<br />

161<br />

162<br />

163<br />

164<br />

166


Index <strong>of</strong> Figures (Continued)<br />

Figure Title Page<br />

Figure 3.47: SEM micrographs <strong>of</strong> Citrobacter cells exposed to 20%<br />

(w/v) manuka <strong>honey</strong> after 30 minutes at x10,000 A &<br />

x15,000 B magnification respectively<br />

Figure 3.48: SEM micrographs <strong>of</strong> untreated Citrobacter cells after 180<br />

minutes at x5,000 (A & B) magnification respectively<br />

Figure 3.49: SEM micrographs <strong>of</strong> Citrobacter cells exposed to 20%<br />

(w/v) manuka <strong>honey</strong> after 180 minutes at x5,000 A &<br />

x15,000 B magnification respectively<br />

Figure 3.50: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Serratia after 30<br />

minutes at x5,000 magnification<br />

Figure 3.51: SEM micrograph <strong>of</strong> Serratia cells exposed to 30% (w/v)<br />

manuka <strong>honey</strong> for 30 minutes at x 5,000 magnification<br />

Figure 3.52: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Serratia after 180<br />

minutes at x5,000 magnification<br />

Figure 3.53: SEM micrograph <strong>of</strong> Serratia cells exposed to 30% (w/v)<br />

manuka <strong>honey</strong> after 180 minutes at x5,000 magnification<br />

Figure 3.54: Transmission micrographs <strong>of</strong> untreated cells <strong>of</strong><br />

Acinetobacter after 1 h incubation with isosensitest borth<br />

(ISB) at 16,000x magnification<br />

Figure 3.55: Transmission micrographs <strong>of</strong> Acinetobacter incubated with<br />

isosensitest borth (ISB) containing 20% (w/v) manuka<br />

<strong>honey</strong> for 1 h at 16,000x magnification.<br />

Figure 3.56: Transmission micrographs <strong>of</strong> untreated cells <strong>of</strong><br />

Acinetobacter incubated with isosensitest borth (ISB) after<br />

3 h at 16,000x magnification<br />

23<br />

167<br />

168<br />

169<br />

171<br />

171<br />

172<br />

172<br />

175<br />

176<br />

177


Index <strong>of</strong> Figures (Continued)<br />

Figure Title Page<br />

Figure 3.57: Transmission micrographs <strong>of</strong> Acinetobacter incubated with<br />

isosensitest borth (ISB) containing 20% (w/v) manuka<br />

<strong>honey</strong> after 3 h at 16,000x magnification<br />

Figure 3.58: Transmission micrographs <strong>of</strong> untreated cells <strong>of</strong> E.coli after<br />

1 h incubation with isosensitest borth (ISB) at 16,000x<br />

magnification<br />

Figure 3.59: Transmission micrographs <strong>of</strong> E.coli incubated with<br />

isosensitest borth (ISB) containing 20% (w/v) manuka<br />

<strong>honey</strong> after 1 h at 16,000x magnification<br />

Figure 3.60: Transmission micrographs <strong>of</strong> untreated cells <strong>of</strong> E.coli after<br />

3 h incubation with isosensitest borth (ISB) at 16,000x &<br />

30,000x magnification<br />

Figure 3.61: Transmission micrographs <strong>of</strong> E.coli incubated with<br />

isosensitest borth (ISB) containing 20% (w/v) manuka<br />

<strong>honey</strong> after 3 h at 16,000x magnification<br />

Figure 3.62: 2-D protein electrophoresis gel <strong>of</strong> Acinetobacter cells<br />

without <strong>honey</strong> treatment<br />

Figure 3.63: 2-D protein electrophoresis gel <strong>of</strong> Acinetobacter cells<br />

exposed to 20% (w/v) manuka <strong>honey</strong><br />

Figure 4.1: Mean MIC and MBC (%w/v) for 30 Acinetobacter strain<br />

against 5 types <strong>of</strong> Hone<br />

Figure 4.2: Mean MIC and MBC (%w/v) for 12 Klebsiella strain<br />

against 5 types <strong>of</strong> <strong>honey</strong><br />

Figure 4.3: Mean MIC and MBC (%w/v) for 10 E.coli strain against 5<br />

types <strong>of</strong> <strong>honey</strong><br />

24<br />

178<br />

180<br />

181<br />

182<br />

183<br />

185<br />

185<br />

198<br />

198<br />

199


Index <strong>of</strong> Figures (Continued)<br />

Figure Title Page<br />

Figure 4.4: Mean MIC & MBC (%w/v) for 12 Citrobacter strain<br />

against 5 types <strong>of</strong> <strong>honey</strong><br />

Figure 4.5 Mean MIC and MBC (%w/v) for 15 Enterobacter strain<br />

against 5 types <strong>of</strong> <strong>honey</strong><br />

Figure 4.6: Mean MIC and MBC (%w/v) for 8 Serratia strain against 5<br />

types <strong>of</strong> <strong>honey</strong><br />

25<br />

199<br />

200<br />

200


AHL-N-acylhomoserine-lactone<br />

Abbreviations list<br />

ATCC- American Type Culture Collection<br />

APF - Antibacterial phenolic fraction<br />

AIF- Apoptosis inducing factor<br />

AMP- Ampicillin<br />

AK- Amikacin<br />

AZT- Aztreonam<br />

BDMA- Benzyl dimethylamine<br />

BSA- Bovine serum albumin<br />

CDC- Centres for Dise<strong>as</strong>e Control<br />

CAZ- Ceftazidime<br />

CE – Cephradin<br />

CFU- Colony forming unit<br />

CHAPS--[3-cholamidopropyl)dimethylammonio]-1-propanesulfonate<br />

CLA- Clavulanic acid<br />

CLSI- Clinical and Laboratory Standards Institute<br />

CRO- Ceftriaxone<br />

CTX- Cefotaxime<br />

CPM- Cefepem<br />

DDSA- Dodecenyl Succinic Anhydride<br />

DDT- DL-Dithiothreitol<br />

DPPH- Di(4-tert-octylphenyl)-1-picryl-hydrazyl<br />

DRD- Decimal reduction dose<br />

26


ESBL- Extended spectrum beta lactam<strong>as</strong>e<br />

ERT- Ertapenem<br />

EDTA- Ethylenediaminetetraacetic acid<br />

EMB- Eosin Methylene Blue<br />

FOX- Cefoxitin<br />

GN- Gentamicin<br />

HPA- Health Protection Agency<br />

HMF- hydroxymetheyl furfural<br />

IMP- Imipenem<br />

IEF- Iso- electric focusing<br />

ISB- Isosensitest broth<br />

MALDI-TOF- MS- Matrix-<strong>as</strong>sisted l<strong>as</strong>er desorption ionisation-time-<strong>of</strong>-flight m<strong>as</strong>s<br />

spectroscopy<br />

MBC- Minimum bactericidal concentration<br />

MDR- Multi-drug resistant<br />

MEM- Meropenem<br />

MG or MGO- Methyglyoxal<br />

MH- Mueller-Hinton<br />

MIC- Minimum inhibitory concentration<br />

MOPS- Morpholino-Propansulfonsaure acid<br />

MRAB-C Imipenem resistant Acinetobacter baumannii- Carbapenem<strong>as</strong>es<br />

MRD- Maximum recovery diluent<br />

MRSA- Methicillin resistant Staph. aureus<br />

MSSA- Methicillin sensitive Staph. aureus<br />

27


MW- Molecular weight<br />

NB- Nutrient broth<br />

NDM-1- New delhi metalobetalactam<strong>as</strong>es<br />

NCTC- National Collection Type Culture<br />

NHB- National <strong>honey</strong> board<br />

NPARU- National Pollen And Research Unit<br />

OH- Omani <strong>honey</strong><br />

OMPs- Outer membrane proteins<br />

OSO4- Osmium tetroxide<br />

OXA- Oxacillin<strong>as</strong>es<br />

PBPs- Penicillin binding protein<br />

pI- Iso- electric point<br />

ROS- Reactive oxygen species<br />

RTI- Respiratory tract infection<br />

SEM- Scanning electron microscopy<br />

SD- Standard deviation<br />

SDS- Sodium dodecylsulphate<br />

SDS- PAGE – Sodium dodecyl sulphate polyacrylamide gel electrophoresis<br />

SHV- Sulphydryl variable<br />

SXT- Septrin<br />

TEM- Transmission electron microscopy<br />

TAZ- Piperacillin-tazobactam<br />

TSB- Tryptone Soya Broth<br />

TVC- Total viable count<br />

28


TEM- Temoneira<br />

UTI- Urinary tract infection<br />

UMF- Unique manuka factor<br />

VRE- Vancomycin resistant Enterococcus<br />

VSE- Vancomycin sensitive Enterococcus<br />

V/V- Volume per volume<br />

W/V- Weight per volume<br />

29


Abstract<br />

Honey h<strong>as</strong> been shown to have therapeutic properties, which include<br />

immunomodulatory and antibacterial activity in vitro and anti-inflammatory,<br />

antipyretic and wound healing properties in vivo. A complex mix <strong>of</strong> factors such <strong>as</strong><br />

acidity, osmolality and hydrogen peroxide content contribute to antibacterial<br />

activity. Unusually manuka <strong>honey</strong> h<strong>as</strong> been shown to contain methylglyoxal which<br />

is derived from nectar collected from the blossom <strong>of</strong> manuka trees and this confers<br />

high antibacterial activity. Manuka <strong>honey</strong> is used in licensed wound dressings in<br />

the UK. Its ability to inhibit staphylococci h<strong>as</strong> been reported, but its efficacy with<br />

Gram negative bacteria is less well documented. Since these bacteria are difficult to<br />

control and commonly infect military wounds and burns, there is a need to<br />

investigate their susceptibility to manuka <strong>honey</strong>. The main aim <strong>of</strong> this study is to<br />

<strong>as</strong>sess the <strong>antimicrobial</strong> potential <strong>of</strong> manuka <strong>honey</strong> against multi-drug resistant<br />

(MDR) Gram negative rods with the potential to infect wounds. Eighty five clinical<br />

isolates were tested in this study (30 MDR Acinetobacter and 55 extended spectrum<br />

beta-lactam<strong>as</strong>es [ESBL] producing members <strong>of</strong> the Enterobacteriaceae). The<br />

minimum inhibitory concentration (MIC) <strong>of</strong> manuka <strong>honey</strong> for each isolate w<strong>as</strong><br />

determined by agar incorporation and broth dilution methods, <strong>as</strong> well <strong>as</strong> the<br />

minimum bactericidal concentration (MBC). The kinetics <strong>of</strong> inhibition <strong>of</strong> selected<br />

isolates with high MIC values w<strong>as</strong> monitored by total viable counts. Also,<br />

ultr<strong>as</strong>tructural changes in cell morphology were studied before and after exposure<br />

to manuka <strong>honey</strong> using scanning (SEM) and transmission electron microscopy<br />

(TEM). Electron micrographs were examined for structural changes, such <strong>as</strong><br />

altered shape, surface abnormalities and evidence <strong>of</strong> cell division.<br />

Eight Omani <strong>honey</strong>s were <strong>as</strong>sayed for their antibacterial activity using bio<strong>as</strong>say, MIC<br />

and MBC methods. Omani <strong>honey</strong>s were also analysed for their chemical and<br />

physical properties such <strong>as</strong> pH, protein, water and sugar contents,<br />

hydroxymetheylfurfural (HMF), colour and antioxidant properties. Pollen analysis<br />

w<strong>as</strong> also used for identifying the flora origin <strong>of</strong> <strong>honey</strong>. All Omani <strong>honey</strong>s were found<br />

to possess peroxide activity nonetheless it exhibited a bactericidal mode <strong>of</strong> activity<br />

against all MDR and ESBLs tested. In addition <strong>honey</strong> analysis revealed<br />

unadulterated and natural <strong>honey</strong>. A study <strong>of</strong> anti-radical activity and phenolic<br />

contents demonstrated that Omani <strong>honey</strong> could be used to promote a rapid wound<br />

healing and aid its antibacterial activity. The proximity <strong>of</strong> MIC and MBC values<br />

indicates that manuka <strong>honey</strong> had a bactericidal mode <strong>of</strong> action against these isolates<br />

and this w<strong>as</strong> confirmed by the time to kill curves. The SEM and TEM <strong>of</strong> images <strong>of</strong><br />

representative isolates after treatment with manuka <strong>honey</strong> showed some physical<br />

membrane damage, septa formation and irregular shape; where<strong>as</strong> non <strong>honey</strong> treated<br />

cells (control) did not appear to be obvious damage. In conclusion manuka <strong>honey</strong><br />

possesses strong antibacterial activity against the antibiotic-resistant wound<br />

pathogens tested here and further investigation into cellular target sites is needed.<br />

Both manuka and selected Omani <strong>honey</strong>s have clinical potential to inhibit pathogens<br />

that commonly colonise wounds.<br />

30


Chapter 1<br />

Introduction<br />

31


1.1 Antimicrobial Resistance:<br />

An <strong>antimicrobial</strong> <strong>agent</strong> is a substance that inhibits or kills microbial growth;<br />

unfortunately, the introduction <strong>of</strong> a new <strong>antimicrobial</strong> <strong>agent</strong> into clinical practice is<br />

usually followed by the rapid emergence <strong>of</strong> resistance. Resistance in bacteria can be<br />

intrinsic, because not all bacterial species are naturally sensitive to all <strong>antimicrobial</strong>s,<br />

or it can be acquired, b<strong>as</strong>ed on genetic mutation or genetic transfer from other<br />

organisms (Forbes et al., 2007). The production <strong>of</strong> drug-inactivating enzymes,<br />

alteration <strong>of</strong> an active target site, acquirement <strong>of</strong> a target byp<strong>as</strong>s system, decre<strong>as</strong>e cell<br />

permeability and an efflux pump in cell membrane are the five mechanisms which<br />

admit microorganisms to acquire resistance through a biochemical b<strong>as</strong>is (Sefton<br />

2002). The mechanisms by which β lactam resistance is manifest in bacteria involve<br />

enzymatic inactivation, or by altered receptors <strong>of</strong> penicillin binding proteins (Winn et<br />

al., 2006).<br />

Resistance to <strong>antimicrobial</strong> <strong>agent</strong>s is not a new incident. Soon after penicillin h<strong>as</strong><br />

established <strong>as</strong> an antibiotic in the 1940s, the rate <strong>of</strong> penicillin resistance had risen to<br />

14%, and is over 90% for Staphylococcus aureus today (Aksoy 2007). In the p<strong>as</strong>t,<br />

resistant strains were believed to be a problem confined to hospitals, however today<br />

resistance h<strong>as</strong> incre<strong>as</strong>ed in the community (Aksoy 2007).<br />

Urinary tract infections (UTI), respiratory tract infections (RTI), and tuberculosis are<br />

considered to be problem dise<strong>as</strong>es both in the community and the hospital setting<br />

with regard to <strong>antimicrobial</strong> resistance. Misuse <strong>of</strong> antibiotics by healthcare<br />

pr<strong>of</strong>essionals, unpr<strong>of</strong>essional doctors, poor drug quality, poor hygienic conditions<br />

and inadequate surveillance me<strong>as</strong>ures account for the emergence <strong>of</strong> resistant bacteria.<br />

All <strong>of</strong> these factors contribute to the spread <strong>of</strong> multidrug resistant (MDR) organisms,<br />

32


such <strong>as</strong> methicillin resistant Staphylococcus aureus (MRSA), vancomycin resistant<br />

Enterococcus species (VRE), Extended spectrum β-lactam<strong>as</strong>e (ESBL) producing<br />

Enterobacteriaceae, Pseudomon<strong>as</strong> aeruginosa, and Acinetobacter baumannii<br />

(Madigan et al., 2009). According to the Centre for Dise<strong>as</strong>e Control and Prevention<br />

(CDC), more than 70% <strong>of</strong> bacteria that are concidered now <strong>as</strong> a sources <strong>of</strong> hospital<br />

acquired infections are resistant to at le<strong>as</strong>t one <strong>of</strong> the drugs that are most commonly<br />

used to treat them (Aksoy 2007).<br />

1.2 Extended Spectrum Beta-Lactam<strong>as</strong>es (ESBLs):<br />

ESBLs are enzymes that inactivate or hydrolyse β-lactam antibiotics by cleaving the<br />

C-N bond on the β-lactam ring. Most <strong>of</strong> these enzymes are pl<strong>as</strong>mid mediated; they<br />

hydrolyze penicillins, cephalosporins, and aztreonam and are inhibited by β-<br />

lactam<strong>as</strong>e inhibitors, such <strong>as</strong> clavulanate, sulbactam and tazobactam, (Moland et al.,<br />

2008).<br />

Beta lactam<strong>as</strong>es are commonly circulated in nature and are generally cl<strong>as</strong>sified<br />

according to the main compounds that they inactivate (e.g., <strong>as</strong> penicillin<strong>as</strong>es or<br />

cephalosporin<strong>as</strong>es) (Baron 1996). The enzymes can be produced in either a<br />

constitutive or an inducible manner. Infections with ESBLs can vary from urinary<br />

tract infections (UTIs) to more complicated deadly sepsis (Bhattacharya 2006).<br />

Many β-lactam<strong>as</strong>e resistance genes <strong>of</strong> Gram negative bacteria are present on the<br />

chromosome and some are carried on pl<strong>as</strong>mids which can be transferred to other<br />

organisms. A recently discovered transfer mechanism is the transposon which carries<br />

genes between the chromosomes and the pl<strong>as</strong>mids (Greenwood et al., 2007) and an<br />

incre<strong>as</strong>ing number <strong>of</strong> β-lactam<strong>as</strong>es have been <strong>as</strong>sociated with integrons. Today,<br />

ESBL producing bacteria have spread worldwide and have emerged <strong>as</strong> significant<br />

33


community-and hospital acquired pathogens (Murray et al., 2007). European<br />

countries recorded higher incidence <strong>of</strong> ESBL producing Enterobacteriaceae than the<br />

USA, especially for Klebsiella strains (Canton et al., 2008). More than 100 European<br />

intensive care units (ICUs) were involved in a study on the prevalence <strong>of</strong> ESBLs in<br />

Klebsiella. Sweden demonstrated the lowest prevalence <strong>of</strong> ESBL with 3%, where<strong>as</strong><br />

Portugal obtained the highest value with 34% (Paterson & Bonomo 2005)<br />

1.2.1 Cl<strong>as</strong>sification <strong>of</strong> ESBL:<br />

There are many different β -lactam<strong>as</strong>es, and they can be differentiated according to<br />

their substrate and inhibitor specificities, physical factors (pH, isoelectric point) and<br />

immunological differences. These factors makes cl<strong>as</strong>sification very difficult (Scholar<br />

& Pratt 2000), but two schemes are currently used to cl<strong>as</strong>sify β-lactam<strong>as</strong>es, which are<br />

the Bush-Medeiros-Jacopy system (functional cl<strong>as</strong>sification) and the Ambler system<br />

(molecular cl<strong>as</strong>sification) (Murray et al., 2007).<br />

1.2.1.1 Functional Cl<strong>as</strong>sification:<br />

A cl<strong>as</strong>sification scheme for β-lactam<strong>as</strong>es b<strong>as</strong>ed on functional characteristics w<strong>as</strong><br />

categorized on three major groups <strong>of</strong> enzymes. These enzymes have been defined by<br />

the effects <strong>of</strong> their substrate and inhibitor product: group one cephalosporin<strong>as</strong>es<br />

which are inhibited by clavulanic acid; group two penicillin<strong>as</strong>es, cephalosporin<strong>as</strong>es<br />

and broad spectrum β-lactam<strong>as</strong>es are inhibited by active site-occupied β-lactam<strong>as</strong>e<br />

inhibitors and the group three metallo-β-lactam<strong>as</strong>es which hydrolyze penicillins,<br />

cephalosporins, and carbapenems and weakly inhibited by nearly all β-lactam<strong>as</strong>es<br />

inhibitors (Bush, Jacopy & Medeiros 1995)<br />

1.2.1.2 Molecular Cl<strong>as</strong>sification:<br />

34


Beta-lactam<strong>as</strong>es in this cl<strong>as</strong>sification are depended on the nucleotide and amino acid<br />

sequences built in these enzymes under the Ambler system. Four cl<strong>as</strong>ses have been<br />

recognized (A-D), which <strong>as</strong>sociate with the functional cl<strong>as</strong>sification. Cl<strong>as</strong>ses A, C,<br />

and D have serine residues at the active site, similar to penicillin binding proteins<br />

(PBPs), where<strong>as</strong> cl<strong>as</strong>s B or metallo-β-lactam<strong>as</strong>es use a metallic ion, preferentially<br />

zinc for their action (Amabile-Cuev<strong>as</strong> 2007; Murray et al., 2007).<br />

1.2.2 Types <strong>of</strong> ESBL:<br />

1.2.2.1 TEM-type ESBLs (cl<strong>as</strong>s A)<br />

TEM-1 is the most common β-lactam<strong>as</strong>e found in Gram-negative bacteria; it w<strong>as</strong><br />

first deteced from a patient named Temoneira in 1965. At le<strong>as</strong>t 90% <strong>of</strong> ampicillin<br />

resistance in Escherichia coli w<strong>as</strong> found to be due to TEM-1 production, which h<strong>as</strong><br />

an iso-electric point (pI) <strong>of</strong> 5.4 (Amabile-Cuev<strong>as</strong> 2007). TEM-1 inactivate most<br />

penicillins and first generation cephalosporins such <strong>as</strong> cephalothin, cefaclor and<br />

cephaloridine but does not hydrolyse more stable cephalosporins, like cefotaxime,<br />

cefuroxime, cefixime, ceftriaxone, cefepime and ceftazidime and the monobactam,<br />

aztreonam (Moland et al., 2008).<br />

There are two TEM-ESBL families, one derived from TEM-1 and other derived from<br />

TEM-2. These enzymes are pl<strong>as</strong>mid-mediated β-lactam<strong>as</strong>es and can be produced by<br />

several members <strong>of</strong> the Enterobacteriaceae. TEM enzymes involve more than 160<br />

different ESBLs. TEM is commonly distributed because it seems to have emerged by<br />

point mutations, and it is also e<strong>as</strong>ily created in the laboratory (Amabile-Cuev<strong>as</strong>,<br />

2007). More than 100 derived <strong>of</strong> TEM β -lactam<strong>as</strong>es have been identified on a world<br />

wide b<strong>as</strong>is (Greenwood et al., 2007)<br />

35


1.2.2.2 SHV-type ESBLs (cl<strong>as</strong>s A)<br />

SHV is an abbreviation for sulphydryl variable (Paterson and Bonomo, 2005). The<br />

SHV-1 β-lactam<strong>as</strong>e h<strong>as</strong> been most commonly found in Klebsiella pneumoniae<br />

isolates and pl<strong>as</strong>mid-mediated ampicillin resistance h<strong>as</strong> accounted for more than 20%<br />

<strong>of</strong> this enzyme in this species. This enzyme can also be produced by Citrobacter<br />

diversus, Escherichia coli, and P. aeruginosa. SHV or a related gene is usually<br />

incorporated into the bacterial chromosome, but it h<strong>as</strong> also been found in pl<strong>as</strong>mids<br />

(Jacopy and Munoz-Price 2005). The majority <strong>of</strong> SHV family variant ESBLs<br />

producers are differentiated by point mutation with the replacement <strong>of</strong> a serine for<br />

glycine at position 238 (Bradford 2001). Recently outbreaks <strong>of</strong> SHV-producers have<br />

been reported in Acinetobacter spp, and P. aeruginosa (Amabile-Cuev<strong>as</strong> 2007).<br />

Currently, there are approximately 100 SHV- variants β-lactam<strong>as</strong>es (Moland et al.,<br />

2008).<br />

1.2.2.3 CTX-M type ESBLs (cl<strong>as</strong>s A):<br />

CTX-M ESBLs are not mutated forms <strong>of</strong> broad spectrum β -lactam<strong>as</strong>es but are<br />

derived from chromosomal enzymes found in rare bacteria known <strong>as</strong> Kluyvera<br />

species. The encoding gene <strong>of</strong> this enzyme is also commonly located on pl<strong>as</strong>mids<br />

with different sizes (7-160 kb). At present approximately half <strong>of</strong> the CTX-M<br />

enzymes have been discovered to be pl<strong>as</strong>mid encoded (Amabile-Cuev<strong>as</strong> 2007).<br />

CTX-M w<strong>as</strong> first detected in 1990 and it spread between Enterobacteriaceae such <strong>as</strong><br />

E.coli, K. pneumoniae, Salmonella, Shigella, Citrobacter freundii, Enterobacter and<br />

Serratia marcescens (Murray et al., 2007). More than 90 molecular variants <strong>of</strong> CTX-<br />

M have been described (Zong and Yu 2010). It is called CTX (a common<br />

abbreviation for cefotaxime) because many CTX-M enzymes are able to hydrolyse<br />

36


cefotaxime f<strong>as</strong>ter than ceftazidime; however, a few CTX-Ms significantly hydrolyse<br />

ceftazidime (e.g., CTX-M-15, -19, -25 and -32). CTX-M carried on specific bacterial<br />

genotypes w<strong>as</strong> related to different geographical regions (Hawkey & Jones 2009).<br />

CTX-M-14 and-15 seems to be the most pandemic genotypes in recent times<br />

(Hawkey 2008). Moreover, these enzymes were more readily inhibited by<br />

tazobactam compared to other β-lactam<strong>as</strong>e inhibitors such <strong>as</strong> sulbactam and<br />

clavulanate (Bradford 2001). This may explain the higher sensitivity <strong>of</strong> some CTX-<br />

M producers to piperacillin-tazobactam than to cefepime (Amabile-Cuev<strong>as</strong> 2007).<br />

1.2.2.4 OXA-type ESBLs (Cl<strong>as</strong>s D)<br />

OXA ESBLs are enzymes that hydrolyse oxacillin and cloxacillin. These ESBLs<br />

occur commonly in the Enterobacteriaceae, Acinetobacter and P. aeruginosa. They<br />

belong to molecular cl<strong>as</strong>s D which dishtinguishes them from the TEM and SHV<br />

enzymes (Murray et al., 2007). Although resistance to ceftazidime is a phenotypic<br />

indicator in the detection <strong>of</strong> this type <strong>of</strong> ESBL, their detection can be difficult<br />

because <strong>of</strong> the weak inhibition by clavulanate (Moland et al., 2008).<br />

1.2.2. 5 AmpC-type ESBLs (Cl<strong>as</strong>s C)<br />

AmpC β-lactam<strong>as</strong>es enzyme (also termed cl<strong>as</strong>s C or group 1) are commonly isolated<br />

from Gram-negative bacteria that are resistant to extended-spectrum cephalosporins.<br />

This enzyme is usually determined on the chromosome <strong>of</strong> many Enterobacteriaceae<br />

including Citrobacter, Serratia and Enterobacter species and usually h<strong>as</strong> inducible<br />

expression; where<strong>as</strong> in E. coli it is not usually inducible, even if it is hyper-<br />

expressed. Thus, AmpC β-lactam<strong>as</strong>e genes <strong>of</strong> Gram-negative bacteria have been<br />

transferred onto pl<strong>as</strong>mids and have spread worldwide by incre<strong>as</strong>ing their number and<br />

variety (Hawkey 2008).<br />

37


AmpC β-lactam<strong>as</strong>es, compared to ESBLs are able to hydrolyse third generation<br />

cephalosporins but are not inhibited by β-lactam<strong>as</strong>e inhibitors such <strong>as</strong> clavulanic<br />

acid, sulbactam and tazobactam (Murray et al., 2007). The detection <strong>of</strong> the ESBL<br />

gene in the Enterobacteriaceae can be difficult if AmpC pl<strong>as</strong>mids are present in the<br />

same isolate, <strong>as</strong> well <strong>as</strong> ESBL gene. To avoid this cefepime or cefpirome (AmpC-<br />

stable cephalosporins) can be used in combination with β-lactam<strong>as</strong>es inhibitors such<br />

<strong>as</strong> clavulanate or boronic acid, even though this detection is not 100% perfect when<br />

using these combinations (Hawkey 2008).<br />

1.2.2.6 Carbapenem<strong>as</strong>es (cl<strong>as</strong>s A, B, D)<br />

Carbapenems have been suggested <strong>as</strong> the most effective treatment for the most<br />

extended-spectrum-β-lactam<strong>as</strong>es. However, incre<strong>as</strong>ing reports <strong>of</strong> carbapenem<strong>as</strong>es<br />

enzymes that are able to hydrolyse oxyimino-cephalosporins, cephamycins and<br />

carbapenems makes treatment difficult (Murray et al., 2007). Carbapenem<strong>as</strong>es<br />

belong to three molecular cl<strong>as</strong>ses: A, B and D.<br />

Cl<strong>as</strong>s A carbapenem<strong>as</strong>es can hydrolyse imipenem but are inhibited by clavulanic<br />

acid. Most <strong>of</strong> this cl<strong>as</strong>s h<strong>as</strong> chromosomal genes, but some are pl<strong>as</strong>mid-mediated,<br />

such <strong>as</strong> KPC-1 which is found mainly in K. pneumoniae. Cl<strong>as</strong>s B such <strong>as</strong> IMP or<br />

VIM group are not inhibited by clavulanic acid (Murray et al., 2007). Recently<br />

Yong et al., (2009) w<strong>as</strong> first to report a new subgroup c<strong>as</strong>e <strong>of</strong> cl<strong>as</strong>s B <strong>of</strong> metallo β-<br />

lactam<strong>as</strong>es called NDM-1 isolated from New Delhi, India. This mobile gene w<strong>as</strong><br />

detected on a pl<strong>as</strong>mid and found mainly in K. pneumoniae. NDM-1 with 28 kDa can<br />

hydrolyse all β-lactams antibiotics except aztreonam and it represents a potential<br />

public health problem (Kumar<strong>as</strong>amy et al., 2010).<br />

Cl<strong>as</strong>s D carbapenam<strong>as</strong>es are mostly found in Acinetobacter baumannii. However,<br />

they can develop resistant to carbapenems if there is an alteration in a porin.<br />

38


1.2.3 Emergence <strong>of</strong> ESBL:<br />

The production <strong>of</strong> the TEM and SHV β-lactam<strong>as</strong>es in pl<strong>as</strong>mid mediated resistance<br />

strains initiated many clinical problems after the introduction <strong>of</strong> ampicillin in the<br />

1960s. In the early 1980s, third generation extended spectrum cephalosporins, such<br />

<strong>as</strong> cefotaxime and ceftazidime had been developed and <strong>of</strong>fered reliable treatment for<br />

patients infected with Enterobacteriaceae. However, in the mid 1980s the first ESBL<br />

producing organisms (resistant to third generation cephalosporins) were isolated.<br />

Very rapidly mutations in amino acids in both TEM and SHV genes occurred, which<br />

spread specifically between Klebsiella species and some strains <strong>of</strong> E. coli. As a<br />

result in the emergence <strong>of</strong> the ESBL mutant, derivatives <strong>of</strong> these widely spread β-<br />

lactam<strong>as</strong>es that were capable <strong>of</strong> hydrolysing third generation cephalosporins and<br />

monobactams appeared (Hawkey 2008). Both excessive use <strong>of</strong> antibiotics and<br />

environmental conditions were therefore consider to be main re<strong>as</strong>ons for the<br />

emergence and spread <strong>of</strong> resistance. ESBL have also been reported in Enterobacter,<br />

Salmonella, Proteus, Citrobacter, Morganella morganii, Serratia marcescens,<br />

Shigella dysenteriae, Pseudomon<strong>as</strong> aeruginosa, Burkholderria cepacia and<br />

Acinetobacter baumannii (Moland et al., 2008).<br />

1.2.4 Risk factors for ESBL:<br />

To determine the risk factors <strong>of</strong> ESBL producing organisms, several studies were<br />

conducted in different countries. It w<strong>as</strong> found that ESBL-producing strains were<br />

<strong>as</strong>sociated with low patient outcome and crowded hospitals and were also linked to<br />

improper first line treatment (Amabile-Cuev<strong>as</strong> 2007). The risk factors for most<br />

ESBLs are excessive previous use <strong>of</strong> multiple ranges <strong>of</strong> antibiotics, including third<br />

39


generation cephalosporins, cotrimoxazole and cipr<strong>of</strong>loxacin, severely ill patients<br />

with prolonged hospital stay, or patients with indwelling medical devices such <strong>as</strong> the<br />

presence <strong>of</strong> urinary catheters, placement <strong>of</strong> endotracheal tubes for more than 10 days<br />

and old age (Hawkey 2008; Paterson & Bonomo 2005).<br />

1.2.5 Treatment <strong>of</strong> ESBLs:<br />

Due to high rates <strong>of</strong> resistance <strong>of</strong> ESBL-producing bacteria to fluoroquinolones, the<br />

treatment can be complicated. If an ESBL-producing strain is detected in an isolate<br />

<strong>of</strong> K. pneumoniae, E.coli or P. mirabilis, the CLSI recommends the laboratory to<br />

report it <strong>as</strong> resistant to all penicillins, cephalosporins, and aztreonam, even if it<br />

showed sensitivity (Moland et al., 2008).<br />

Carbapenems have been recommended <strong>as</strong> the drug cl<strong>as</strong>s selected for serious<br />

infections caused by ESBL producing Enterobacteriaceae strains. However, in the<br />

c<strong>as</strong>e <strong>of</strong> carbapenem<strong>as</strong>es emergence, tigecycline revealed excellent potential activity<br />

against ESBL producing Enterobacteriaceae and Acinetobacter (Hawkey 2008).<br />

Recently, according to the survey <strong>of</strong> 104 isolates the presence <strong>of</strong> world wide a<br />

resistant strain to tigecycline w<strong>as</strong> not reported (C<strong>as</strong>tanheira 2008). If CTX-M ESBL<br />

is not prevalent, cefepime h<strong>as</strong> been suggested <strong>as</strong> another treatment choice (Amabile-<br />

Cuev<strong>as</strong> 2007).<br />

1.3 Acinetobacter:<br />

During the l<strong>as</strong>t two decades, bacteria <strong>of</strong> the genus Acinetobacter have been selected<br />

<strong>as</strong> one <strong>of</strong> the most important nosocomial pathogens, especially in ICU units. They<br />

have been also involved in many infections such <strong>as</strong> bacteraemia, urinary tract<br />

infection, pneumonia, skin and tissue infections and in secondary meningitis.<br />

40


Acinetobacter spp are widespread in water and soil <strong>as</strong> free-living saprophytes<br />

(Hawkey & Bergogne-Berezin 2006). Acinetobacter is now also involved in<br />

aggressive situations such <strong>as</strong> war district zones or earthquake are<strong>as</strong> (Dallo and<br />

Weitao 2010).<br />

1.3.1 Taxonomy and Historical Features <strong>of</strong> Acinetobacter:<br />

Acinetobacter w<strong>as</strong> first described under the group <strong>of</strong> "Micrococcus calcoaceticus" by<br />

Beijerinck in 1911. In 1956 these bacteria were then cl<strong>as</strong>sified under the name <strong>of</strong><br />

Moraxella in France, but at the same time a group <strong>of</strong> French researchers had<br />

recognised a genus named Acinetobacter. In 1968 a phenotypic study <strong>of</strong> 106 strains<br />

w<strong>as</strong> completed by Baumann which resulted in the recognition <strong>of</strong> only a single<br />

species named Acinetobacter baummanii (Towner 1997). In the early 1970's, most<br />

isolates <strong>of</strong> Acinetobacter were sensitive to many <strong>antimicrobial</strong> <strong>agent</strong>s (Greenwood et<br />

al., 2003). However during the same period many microbiologists in hospitals<br />

noticed that these organisms were pathogenic and implicated in various nosocomial<br />

infections. In 1986, Bovetand and Grimont were compeleted a b<strong>as</strong>ic subdivision <strong>of</strong><br />

the genus Acinetobacter and identified 12 genomic species by DNA-DNA<br />

hybridization. However, today there are at le<strong>as</strong>t 19 genomic species <strong>of</strong><br />

Acinetobacter (Murray et al., 2007).<br />

Although, Acinetobacter baumannii is known to be the most clinically important<br />

strains among species, there are other important nosocomial pathogens called the „A.<br />

baumannii A. calcoaceticus complex‟ (referred to the genomospecies 1, 2, 3 and 13<br />

<strong>of</strong> Tjernberg and Ursing) (Gerischer 2008). This complex is accountable for many<br />

epidemic infections throughout the world because <strong>of</strong> the multi-resistant gene that it<br />

contains. Other species are not very important because they are rarely involved in<br />

41


outbreaks <strong>of</strong> human dise<strong>as</strong>e (Brauers et al., 2005; Joly-Guillou, 2005; Murray et al.,<br />

2007).<br />

1.3.2 Laboratory Diagnosis:<br />

Morphologically, Acinetobacter are aerobic, non motile Gram-negative coccobacilli<br />

and are usually found in diploid shape or chains <strong>of</strong> different length. They are strictly<br />

aerobic and grow simply on all common media at temperatures from 20 to 30 o C for<br />

most strains, the optimum temperature for this bacterium at 33-35 o C (Winn et al.,<br />

2006). They are oxid<strong>as</strong>e-negative, catal<strong>as</strong>e-positive, indole-negative and nitrate-<br />

negative. Furthermore, the initial clue in recognising these bacteria is the appearance<br />

<strong>of</strong> tiny (1.0x 0.7 µm) diplococci with the Gram stain (Koneman et al., 1997).<br />

Colonies appear smooth, opaque, sometimes mucoid and slightly smaller than those<br />

<strong>of</strong> members <strong>of</strong> the family Enterobacteriaceae on blood agar. Most strains appear<br />

colourless, slightly pink or lavender in colour on MacConkey agar due to lactose<br />

oxidation (Engelkirk 2007). The genus <strong>of</strong> Acinetobacter can be therefore subdivided<br />

into two groups. Acinetobacter that are able to oxidise glucose are called<br />

saccharolytic, with those that are unable called <strong>as</strong>accharolytic (Engelkirk 2007).<br />

Most glucose-oxidizing non-haemolytic clinical strains are A. baumannii, most<br />

glucose-negative non-haemolytic ones are A. lw<strong>of</strong>fii, and most haemolytic ones are<br />

A. haemolytic (Murray et al., 2007).<br />

1.3.3 Clinical features <strong>of</strong> Acinetobacter infections:<br />

Acinetobacter species particularly Acinetobacter baumannii, can cause many clinical<br />

disorders, including pneumonia, secondary meningitis, bacteraemia, wound<br />

infections in burn patients and UTI. They are also isolated from skin, throat and<br />

42


many secretions <strong>of</strong> normal people and are part <strong>of</strong> the commensal flora (Hawkey &<br />

Bergogne-Berezin 2006). Other species such <strong>as</strong> A. lw<strong>of</strong>fii, A. johnsonii, and A.<br />

radioresistens, seem to be natural inhabitants <strong>of</strong> human skin and <strong>as</strong> commensals in<br />

the oropharynx and vagina (Winn et al., 2006). They are considered <strong>as</strong> less resistant<br />

to antibiotics and e<strong>as</strong>ier to eliminate. Two recent species have been described which<br />

are <strong>as</strong>sociated with infections; these are A. ursingii and A. schindleri (Bergogne-<br />

Berezin et al., 2008).<br />

Acinetobacter can only be obtained from soil, water, food and sewage (Towner<br />

1997) and are also able to live for long periods in lifeless environments (Greenwood<br />

et al., 2003). In the c<strong>as</strong>e <strong>of</strong> wound infections, bacteraemia within 3-5 days following<br />

infection can <strong>of</strong>ten develop. In several large c<strong>as</strong>e series, 4-27% <strong>of</strong> all Acinetobacter<br />

that caused bacteraemia occurred <strong>as</strong> a result <strong>of</strong> infected surgical or burn wounds<br />

(Gillespie 2004; Hawkey & Bergogne-Berezin 2006). Such infections are <strong>of</strong>ten<br />

difficult to treat because <strong>of</strong> the ability <strong>of</strong> Acinetobacter to become rapidly resistance<br />

to multiple antibiotics, including aminoglycosides, expanded-spectrum<br />

cephalosporins, carbapenems and fluoroquinolones (Gerischer 2008).<br />

Inappropriate or excessive use <strong>of</strong> antibiotics therapy (i.e third generation<br />

cephalosporin), surgery, use <strong>of</strong> medical machinery (e.g. ventilators), insertion <strong>of</strong><br />

intravenous or urinary catheters, and prolonged hospital stay are all identified <strong>as</strong> risk<br />

factors for colonization and infection with Acinetobacter (Hawkey & Bergogne-<br />

Berezin 2006). Soap and water hand w<strong>as</strong>hing and alcohol b<strong>as</strong>ed gels, could therefore<br />

reduce the spread <strong>of</strong> this strain (Joly-Guillou 2005).<br />

Acinetobacter h<strong>as</strong> been detected from a large selection <strong>of</strong> clinical samples, including<br />

blood, urine, faeces, cerebrospinal fluid and sputum (Gillespie 2004). It is an<br />

opportunistic pathogen and is commonly found in patient samples. However, serious<br />

infection caused by Acinetobacter depends upon the site <strong>of</strong> infection <strong>as</strong> well <strong>as</strong> the<br />

patient‟s immunity to infection (Murray et al., 2007). Furthermore, Chiang, et al.,<br />

43


(2008) added incre<strong>as</strong>ed serum creatinine level and malignancy <strong>as</strong> risk factors<br />

<strong>as</strong>sociated with incre<strong>as</strong>ed mortality in patients with bacteraemia caused by<br />

Acinetobacter. A European survey <strong>of</strong> the main cause <strong>of</strong> nosocomial pneumonia<br />

carried out in seven countries h<strong>as</strong> established an overall incidence <strong>of</strong> approximately<br />

10% for Acinetobacter (Hawkey & Bergogne-Berezin 2006). Also, during a study<br />

period from 2003 to 2006 for over 270 patients admitted every year in a burn clinic,<br />

an incre<strong>as</strong>ed trend <strong>of</strong> Acinetobacter strains w<strong>as</strong> confirmed (Babik et al., 2008).<br />

Recently, over 21,000 American army personnel who were injured during the Iraq<br />

war have suffered severe wound infections, mostly from resistant strains <strong>of</strong><br />

Acinetobacter baumanii (Murray et al., 2008). Scientists examined Iraq and Kuwait<br />

soil for the presence <strong>of</strong> this pathogen but it proved negative. The source <strong>of</strong> this<br />

outbreak could therefore be from European hospitals (Silberman 2007). However,<br />

this bacterium caused bacteraemia, osteomyelitis and respiratory infections for the<br />

soldiers. Also this pathogen is able to form a bi<strong>of</strong>ilm which will reduce its<br />

susceptibility to systemic antibiotics and make treatment more difficult (Dallo and<br />

Weitao 2010). In this situation significant Acinetobacter infections have incre<strong>as</strong>ed<br />

worldwide and the outbreaks <strong>of</strong> resistant strains <strong>of</strong> Acinetobacter have been<br />

described in the medical literature (Bergogne-Berezin et al., 2008).<br />

1.3.4 Pathogenesis <strong>of</strong> Acinetobacter infection:<br />

Bacteria produce many substances and molecules that allow them to survive and<br />

grow in a host. These molecules are proteins, enzymes, capsules, toxins and surface<br />

carbohydrates (Bergogne-Berezin et al., 2008). Acinetobacter spp. were thought to<br />

be relatively low-grade pathogens, but a number <strong>of</strong> virulence factors have been<br />

identified. These include:<br />

44


1.3.4.1 The production <strong>of</strong> exopolysaccharide:<br />

The presence <strong>of</strong> exopolysaccharide capsule helps in the protection <strong>of</strong> bacteria from<br />

host defences. A capsule is produced by approximately 30% <strong>of</strong> Acinetobacter strains<br />

and it consists <strong>of</strong> L-rhamnose, D-glucose, D-glucuronic acid, and D-mannose, which<br />

make the cell surface <strong>of</strong> strains more hydrophilic (Joly-Guillou 2005; Hawkey &<br />

Bergogne-Berezin 2006). Acinetobacter strains which produce exopolysaccharide are<br />

known to be more dangerous than those without. This is because this capsule can<br />

block the entry <strong>of</strong> complement to the bacterial cell wall and interrupt the alternative<br />

pathway <strong>of</strong> complement activation (Joly-Guillou 2005).<br />

1.3.4.2 Quorum-sensing:<br />

Quorum-sensing is defined <strong>as</strong> the ability <strong>of</strong> bacteria to initiate the transcription <strong>of</strong><br />

certain genes only when a certain population density is reached. It is known <strong>as</strong> a<br />

widely distributed regulatory mechanism in Gram-negative bacteria such <strong>as</strong><br />

Pseudomon<strong>as</strong> aeruginosa (Wilson et al., 2002). Acinetobacter isolates in the<br />

stationary growth ph<strong>as</strong>e have demonstrated four different signal molecules <strong>of</strong><br />

quorum sensing involved in activating N-acylhomoserine-lactone (AHL) biosensors.<br />

The AHLs system can therefore act <strong>as</strong> a main mechanism for auto-induction <strong>of</strong><br />

several virulence factors in an opportunistic pathogen such <strong>as</strong> Acinetobacter. This<br />

process need to be studied for its clinical implications (Joly-Guillou 2005).<br />

1.3.4.3 The property <strong>of</strong> adhesion to human epithelial cells via<br />

the capsule or fimbriae.<br />

The initial step in the infection process is the ability <strong>of</strong> bacteria to penetrate the host.<br />

This step depends on the adherence capacity and the survival time <strong>of</strong> microorganisms<br />

on mucosal surfaces <strong>of</strong> the host. Bacterial adherence involves the possession <strong>of</strong><br />

45


fimbriae, the production <strong>of</strong> capsular polysaccharides and cell wall components<br />

(Bergogne-Berezin et al., 2008). Recently, two different types <strong>of</strong> adherence were<br />

observed in A. baumannii to epithelial cells <strong>of</strong> human bronchial. The first one w<strong>as</strong><br />

diffusing adherence <strong>of</strong> bacteria to the cell surface and the other type w<strong>as</strong> a group <strong>of</strong><br />

clusters <strong>of</strong> bacteria that adhered to a localized area <strong>of</strong> the cell by producing small<br />

colonies (Bergogne-Berezin et al., 2008).<br />

1.3.4.4 Surface and mitochondrial porins:<br />

On the outer membrane <strong>of</strong> the surface <strong>of</strong> Gram negative bacteria there are special<br />

channels consisting <strong>of</strong> protein molecules called porins. Depending on bacterial<br />

species, porins play a role in the maintenance <strong>of</strong> the cell structure, bacteriophage and<br />

resistance mechanisms <strong>of</strong> <strong>antimicrobial</strong> <strong>agent</strong> (Bergogne-Berezin et al., 2008).<br />

Surface porins allow the p<strong>as</strong>sive diffusion <strong>of</strong> low molecular weight components to<br />

penetrate through this membrane. Large antibiotic molecules penetrate slowly, which<br />

may account for the high antibiotic resistance <strong>of</strong> A.baumannii. For example, the<br />

permeability <strong>of</strong> the outer membrane varies from one Gram negative species to<br />

another; in Pseudomon<strong>as</strong> aeruginosa (which is extremely resistance to antibiotics)<br />

the outer membrane is 100 times less permeable than in E.coli (Brooks et al., 2001).<br />

A study showed the permeability <strong>of</strong> the outer membrane <strong>of</strong> Acinetobacter for a<br />

cephalosporin w<strong>as</strong> 2-7 times lower than in P.aeruginosa (Vila 1998).<br />

Many considerations that involve host factors, the bacterial load, the virulence <strong>of</strong><br />

strains and the production <strong>of</strong> lip<strong>as</strong>e enzymes such <strong>as</strong> butyrate ester<strong>as</strong>e, caprylate<br />

ester<strong>as</strong>e and leucine arylamid<strong>as</strong>e which may damage tissue lipids may play important<br />

roles in initiating infection in colonised patients (Towner 2002).<br />

46


1.3.5 Emergence <strong>of</strong> Resistance:<br />

A.baumannii and related species have acquired resistance to multiple antibiotics<br />

rather than being inherently resistant. When these species were first introduced <strong>as</strong><br />

pathogens to human, most strains were sensitive to ampicillin and cephalosporins. In<br />

1975, less than 20% <strong>of</strong> these strains were resistant to ticarcillin. However, at the end<br />

<strong>of</strong> the 1970s and early 1980s, A.baumannii caused incre<strong>as</strong>e resistance to second<br />

generation cephalosporins which being used to control nosocomial infections. When<br />

the third generation cephalosporins were first introduced, A.baumannii developed<br />

resistance to cefotoxime and ceftazidime. This bacterium therefore h<strong>as</strong> an excessive<br />

acquired resistance to β-lactam drugs (Towner 1997). There are no specific treatment<br />

guidelines for Acinetobacter spp. due to the large variation in antibiotic resistance.<br />

To determine the best mode <strong>of</strong> treatment for a particular isolate, <strong>antimicrobial</strong><br />

susceptibility testing must be performed (Forbes 2007).<br />

Although, carbapenems have been the drug <strong>of</strong> preference in the treatment <strong>of</strong><br />

Acinetobacter infections numerous reports in the medical and scientific literature<br />

have documented resistant strains to carbapenems such <strong>as</strong> imipenem and meropenem<br />

(Costa et al., 2000; Levin 2002; Perez et al., 2007).<br />

According to the Health Protection Agency in the UK (HPA) a survey <strong>of</strong> 1,225 c<strong>as</strong>es<br />

<strong>of</strong> bacteraemia due to Acinetobacter spp were reported from England, Wales and<br />

Northern Ireland in 2007, with total incidence rate <strong>of</strong> 2.2 per 100,000 populations. In<br />

the same survey 12% <strong>of</strong> Acinetobacter spp were shown to be resistant to imipenem,<br />

where<strong>as</strong> the prevalence <strong>of</strong> cipr<strong>of</strong>loxacin and gentamicin resistance w<strong>as</strong> 16% and 12%<br />

respectively (HPA 2008)<br />

Currently, several surveys have studied the prevalence, mode <strong>of</strong> transmission and<br />

risk factors <strong>of</strong> multi-drug resistant A. baumannii in ICUs and burn clinics. They<br />

reported an incre<strong>as</strong>ed incidence <strong>of</strong> an outbreak clone <strong>of</strong> A. baumannii (Babik et al.,<br />

47


2008; Bacakoglu et al., 2009; Barchitta et al., 2009; Cootz and Marra 2008; Fontana<br />

et al., 2008).<br />

1.3.6 Carbapenem action on Acinetobacter:<br />

Carbapenem is the most effective broad spectrum antibiotic among all <strong>of</strong> the β-<br />

lactams and imipenem is one <strong>of</strong> the most important carbapenems (Bergogne-Berezin<br />

et al., 2008). Imipenem is an active <strong>agent</strong> against many organisms including Gram<br />

positive and Gram negative aerobes and anaerobes. It is a bactericidal <strong>agent</strong> that kills<br />

or destroys bacteria at 2-4 times the MIC for most species (Greenwood et al., 2003).<br />

The initial step <strong>of</strong> drug action in destroying bacteria is drug binding to the cell wall<br />

receptors known <strong>as</strong> penicillin binding proteins (PBPs). The transpeptidation process<br />

is stopped and peptidoglycan synthesis is blocked after β-lactam drug binding to one<br />

or more receptors. The next step involves the removal or suppression <strong>of</strong> autolytic<br />

enzymes inhibitor in the cell wall. This activates the lytic enzyme which results in<br />

lysis <strong>of</strong> cells leads to cell death (Brooks et al., 2001).<br />

1.3.7 Mechanisms <strong>of</strong> Carbapenem Resistance:<br />

There are several complex mechanisms and genetics <strong>of</strong> resistance acquired by this<br />

species which involve several pl<strong>as</strong>mid-borne β-lactam<strong>as</strong>es and aminoglycoside<br />

modifying enzymes, <strong>as</strong> well <strong>as</strong> variation in membrane permeability and alteration in<br />

penicillin-binding proteins. The possession <strong>of</strong> these multiple mechanisms may be due<br />

to the physiological capability <strong>of</strong> Acinetobacter and it can obtain DNA by<br />

transformation in vivo (Finch 2003)<br />

48


In the l<strong>as</strong>t few years resistant strain <strong>of</strong> A. baumannii to carbapenem have been<br />

reported globally and known <strong>as</strong> imipenem and meropenem resistant A. baumannii<br />

(IMRAB). The epidemic strains <strong>of</strong> IMRAB demonstrated three different mechanisms<br />

<strong>of</strong> β-lactam<strong>as</strong>es, these are: pl<strong>as</strong>mid mediated enzymes (TEM-1), chromosomal<br />

mediated enzyme (Noval OXA-type) and cephalosporin<strong>as</strong>e ampC-type enzyme (Bou<br />

et al., 2000)<br />

Major carbapenem-resistant Acinetobacter species have metallo-enzyme and OXA-<br />

type enzymes (Bou et al., 2000). However, OXA-type β-lactam<strong>as</strong>es which belong to<br />

cl<strong>as</strong>s D β-lactam<strong>as</strong>e have a poor activity against carbapenems; such enzymes<br />

discovered in A. baumannii isolates from Argentina, Belgium, Kuwait, Scotland,<br />

Spain and Singapore. Many <strong>of</strong> this group <strong>of</strong> enzymes have been cl<strong>as</strong>sified to form a<br />

subgroup in cl<strong>as</strong>s D β-lactam<strong>as</strong>es, currently including the OXA-23, -24, -25, -26, -27<br />

and -40 types (Song et al., 2004).<br />

There are several factors that determine the acquisition <strong>of</strong> multi- resistance in<br />

A. baumannii. One is the intrinsic resistance <strong>of</strong> microorganisms, due to low level<br />

diffusion <strong>of</strong> certain antibiotics through the outer membrane because <strong>of</strong> low number<br />

<strong>of</strong> porins present (Levin 2002). Another is due to the acquisition genetic elements;<br />

there are 3 types <strong>of</strong> mobile genetic elements have been found in Acinetobacter.<br />

These are pl<strong>as</strong>mids, transposons and integrons (Vila 1998). The pl<strong>as</strong>mids contains 3<br />

resistance genes; genes encoding ß-lactam<strong>as</strong>e TEM-1, TEM-2, and CARB-5 (Bou et<br />

al., 2000). The pl<strong>as</strong>mid encoded β-lactam<strong>as</strong>es have attracted great attention in which<br />

the resistance <strong>of</strong> this bacterium occurs by a single genetic event. However, this type<br />

<strong>of</strong> resistance occurs mostly in the highly selective environment <strong>of</strong> the hospital<br />

(Greenwood 2000). Integrons, which are chains <strong>of</strong> genes with a greater mobility to<br />

transfer from one location <strong>of</strong> A.baumannii chromosome to another with help <strong>of</strong> a<br />

transposon, carry this component (Vila 1998). This may be an important factor in the<br />

49


ability <strong>of</strong> Acinetobacter species to survive in human and environmental reservoirs in<br />

which the genes <strong>of</strong> resistance may be transferred (Vila 1998).<br />

For multi-drug resistant Acinetobacter infections, several studies have demonstrated<br />

clinical effectiveness <strong>of</strong> sulbactam in combination with ampicillin or cefoperazone.<br />

The only effective antibacterial <strong>agent</strong> to this bacterium is colistin (Winn et al., 2006).<br />

One report h<strong>as</strong> demonstrated the efficiency and safety <strong>of</strong> colistin in patients with<br />

Acinetobacter infection that w<strong>as</strong> not susceptible to carbapenem and shown that 57%<br />

<strong>of</strong> patients cured with colistin therapy, without prolonged neuro-muscular blockade<br />

<strong>as</strong> a side effect <strong>of</strong> therapy (Torres et al., 2007)<br />

Recently Enoch and his colleges (2008) completed a six month study <strong>of</strong> the outbreak<br />

caused by multi-drug-carbapenem-resistant Acinetobacter baumanii (MRAB-C)<br />

strain in UK hospitals. This outbreak would have incre<strong>as</strong>ed the rate <strong>of</strong> mortality if<br />

not controlled properly. However, isolation <strong>of</strong> the patients infected with MRAB-C,<br />

education <strong>of</strong> staff dealing with those patients, early patient and environmental<br />

screening, and effective hygiene all helped to control this outbreak. Also, patients<br />

infected with MRAB-C were treated with colistin and tigecycline and improved.<br />

1.3.8 Acinetobacter Treatment with Honey:<br />

Due to the emergence <strong>of</strong> bacteria resistant to antibiotics, the bactericidal properties <strong>of</strong><br />

manuka <strong>honey</strong> have been extensively researched. Currently, few studies have<br />

reported the antibacterial activity <strong>of</strong> <strong>honey</strong> against Acinetobacter. George & Cutting,<br />

(2007) initiated an in-vitro study <strong>of</strong> the antibacterial activity <strong>of</strong> Medi<strong>honey</strong> against<br />

130 clinical isolates <strong>of</strong> multi-drug resistant organisms including Acinetobacter. The<br />

study showed that the concentration needed to inhibit the resistant Acinetobacter<br />

strains w<strong>as</strong> 8% (v/v) <strong>of</strong> <strong>honey</strong>.<br />

50


More recently, a study w<strong>as</strong> carried out using Malaysian tualang <strong>honey</strong> in comparison<br />

to manuka <strong>honey</strong> against wound pathogens including Acinetobacter. The<br />

antibacterial activity for both <strong>honey</strong>s w<strong>as</strong> same with the MIC ranges between (11.25<br />

& 12.5% w/v) (Tan et al., 2009).<br />

Although Acinetobacter spp. have been found to be susceptible to <strong>honey</strong>, more<br />

objective evidence derived from clinical trails and animal models to determine<br />

whether <strong>honey</strong> h<strong>as</strong> a similar <strong>antimicrobial</strong> effect in-vivo are required.<br />

1.4 Alternative Antimicrobial Therapies:<br />

As concerns about <strong>antimicrobial</strong> resistance incre<strong>as</strong>e, efforts <strong>of</strong> the pharmaceutical<br />

industry to develop new drugs have diminished and the possibility <strong>of</strong> running out <strong>of</strong><br />

effective <strong>antimicrobial</strong> <strong>agent</strong>s h<strong>as</strong> incre<strong>as</strong>ed. It takes 10-12 years for new antibiotic to<br />

be developed and costs approximately £250 million for each one (Greenwood 2003).<br />

Yet the chance <strong>of</strong> developing a new drug that will have excellent bactericidal<br />

properties without causing the emergence <strong>of</strong> resistance is impossible. The<br />

introduction and application <strong>of</strong> <strong>honey</strong> into clinical practice several years ago h<strong>as</strong> put<br />

attention on using it for treatment <strong>of</strong> various infections. An extensive review <strong>of</strong><br />

clinical studies by Molan suggested that <strong>honey</strong> might be success<strong>full</strong>y used <strong>as</strong> an<br />

<strong>antimicrobial</strong> <strong>agent</strong> and also in promoting healing <strong>of</strong> wounds (Molan 2006).<br />

1.4.1 Ancient Use <strong>of</strong> Honey <strong>as</strong> a Medicine:<br />

Honey h<strong>as</strong> been used for many thousands <strong>of</strong> years <strong>as</strong> a food, a medicine and it h<strong>as</strong><br />

been incorporated into cosmetics. A large number <strong>of</strong> different cultures have<br />

extensively used <strong>honey</strong> <strong>as</strong> a medicine for many disorders. It h<strong>as</strong> been used in wound<br />

51


care since the time <strong>of</strong> ancient Egyptians, <strong>as</strong> suggested by an inscription on a<br />

Sumarian clay tablet. Honey w<strong>as</strong> also mentioned in the Holy Koran, the Talmud, the<br />

Bible, <strong>as</strong> well <strong>as</strong> the sacred books <strong>of</strong> India, China, Persia and Egypt (Zumla & Lulat<br />

1989). All the clues point to the medicinal use <strong>of</strong> <strong>honey</strong> throughout human history.<br />

1.4.2 Honey <strong>as</strong> a Modern Medicine:<br />

In the l<strong>as</strong>t 30 years interest in the use <strong>of</strong> <strong>honey</strong> <strong>as</strong> a treatment <strong>agent</strong> h<strong>as</strong> incre<strong>as</strong>ed.<br />

Most research that h<strong>as</strong> been undertaken h<strong>as</strong> focused on the employment <strong>of</strong> <strong>honey</strong> in<br />

wound treatment (Ahmed et al., 2003; Berguman et al., 1983; Dumronglert, 1983;<br />

Emarah, 1982; Haffejee & Moosa, 1985; Ingle et al., 2006; Wadi et al., 1987).<br />

In 1988, Efem reported the first large clinical cohort study involving 59 patients who<br />

had a variety <strong>of</strong> wounds such <strong>as</strong> Fournier‟s gangrene, burns and ulcers. The use <strong>of</strong><br />

<strong>honey</strong> on these patients resulted in successful wound healing and the clearance <strong>of</strong><br />

infection. In addition Subrahmanyam (1993, 1994, 1996, 1998) and Subrahmanyam<br />

et al., 2001, 2003, 2007) reported several clinical trials on burns patients with <strong>honey</strong><br />

compared to various different treatments.<br />

Clinically, many researchers have studied the uses <strong>of</strong> <strong>honey</strong> in wound management<br />

and it h<strong>as</strong> been reported to clear wound pathogens rapidly (Al-Waili and Saloom,<br />

1999; Lusby et al., 2002), to stimulate immune response and to reduce inflammation<br />

(Molan and Betts 2001; Tonks et al., 2003) and to support the debridement <strong>of</strong><br />

wounds by autolysis (Stephen-Haynes 2004). In addition, <strong>honey</strong> h<strong>as</strong> been reported to<br />

have a deodorising property on wounds, due to the oxidation <strong>of</strong> glucose by bacteria<br />

resulting in production <strong>of</strong> lactic acid rather than malodorous compounds such <strong>as</strong><br />

ammonia, sulphur compounds and amines produced by the breakdown <strong>of</strong> amino<br />

acids (Cooper 2005; Molan 2002; Stephen-Haynes 2004). Moreover, <strong>honey</strong> h<strong>as</strong> been<br />

used effectively on skin grafts (Schumacher 2004), diabetic foot ulcers (Eddy &<br />

52


Gideonsen 2005), malignant ulcers (Simon et al., 2005) and abscesses (Okeniyi et<br />

al., 2005). Some researchers have observed that <strong>honey</strong> promotes tissue regeneration<br />

through the stimulation <strong>of</strong> angiogenesis and the growth <strong>of</strong> fibrobl<strong>as</strong>ts and epithelial<br />

cells (Efem, 1988, 1993; Stephen-Haynes 2004; Subrahmanyam, 1994, 1998). F<strong>as</strong>t<br />

healing can therefore minimise the need for skin grafts (Subrahmanyam 1998).<br />

Recently, (Gethin et al., 2008) observed that the use <strong>of</strong> manuka <strong>honey</strong> <strong>as</strong> a wound<br />

dressing reduced wound pH which in turn decre<strong>as</strong>ed prote<strong>as</strong>e activity, incre<strong>as</strong>ed<br />

fibrobl<strong>as</strong>t activity and rele<strong>as</strong>ed more oxygen from haemoglobin to promote rapid<br />

wound healing.<br />

Furthermore, after <strong>honey</strong> is applied to the wound, it forms a film <strong>of</strong> liquid between<br />

the wound and the dressing that prevents the dressing from sticking to the wound,<br />

reducing pain and not damaging the newly formed cells. As <strong>honey</strong> h<strong>as</strong> no adverse<br />

effects on tissue, it can be used on wounds safely and introduced into cavities and<br />

sinuses to clear infection (Molan 2000).<br />

Despite, extensive anecdotal evidence to support the topical used <strong>of</strong> <strong>honey</strong> in treating<br />

wounds, systematic review <strong>of</strong> the clinical evidence h<strong>as</strong> not been so supportive (Bardy<br />

et al., 2008; Jull et al., 2008; Moore et al., 2001).<br />

1.4.3 Antimicrobial Activity <strong>of</strong> Honey:<br />

The antibacterial activity <strong>of</strong> <strong>honey</strong> w<strong>as</strong> first identified by Van Ketel in 1892<br />

(Dustmann 1979). After that several studies established the antibacterial activity <strong>of</strong><br />

<strong>honey</strong> against various bacterial pathogens and fungi (Cooper et al., 1999; Efem, et<br />

al., 1992; Molan, 1992a; Molan, 1992b; Mulu et al., 2004; Lusby et al., 2005;<br />

Wilkinson and Cavonagh 2005). It w<strong>as</strong> shown that <strong>honey</strong> inhibits various bacterial<br />

53


species. There are many reports <strong>of</strong> it being bacteriostatic and bactericidal (Alandejani<br />

et al., 2009; Henriques et al., 2009; Molan 1992b).<br />

Undiluted <strong>honey</strong> w<strong>as</strong> shown to prevent the growth <strong>of</strong> Candida albicans and<br />

demostrated potential <strong>as</strong> a topical treatment <strong>of</strong> external fungal infections such <strong>as</strong><br />

ringworm and superficial candidi<strong>as</strong>es (Brady et al., 1997; Efem 1992; Irish et al.,<br />

2006; Wahdan 1998).<br />

Cooper et al., (1999) showed that <strong>honey</strong> h<strong>as</strong> an effective antibacterial activity against<br />

the major wound infecting species including Staphylococcus aureus. A year later the<br />

sensitivity <strong>of</strong> multi-resistant strains <strong>of</strong> Burkholderia cepacia isolated from cystic<br />

fibrosis patients to manuka <strong>honey</strong> at concentrations below 6% (v/v) w<strong>as</strong> reported<br />

(Cooper et al.,2000).<br />

Cooper et al., (2002a) compared the <strong>antimicrobial</strong> activity <strong>of</strong> artificial <strong>honey</strong> ( a<br />

sugar solution) and two natural <strong>honey</strong>s (manuka and p<strong>as</strong>ture <strong>honey</strong>) against 18<br />

strains <strong>of</strong> MRSA isolated from wounds, 20 strains <strong>of</strong> vancomycin-resistant<br />

enterococci (VRE) and 7 strains <strong>of</strong> vancomycin-sensitive enterococci (VSE). The<br />

study showed the minimum inhibitory concentration (MIC) which is the<br />

concentration required to inhibit the growth for the natural <strong>honey</strong>s w<strong>as</strong> below 10%<br />

(v/v) and < 30% (v/v) with artificial <strong>honey</strong> for all strains. This means the<br />

antibacterial activity <strong>of</strong> <strong>honey</strong> is not limited to osmolarity. Also in the same study it<br />

w<strong>as</strong> concluded that there w<strong>as</strong> no difference in the MIC values between the sensitive<br />

and resistant Gram positive strains. Furthermore, the <strong>antimicrobial</strong> activity <strong>of</strong> the<br />

same two natural <strong>honey</strong>s w<strong>as</strong> tested against 17 strains <strong>of</strong> Pseudomon<strong>as</strong> aeruginosa<br />

isolated from burns. Both <strong>honey</strong>s maintained bactericidal activity when diluted more<br />

than 10-fold (Cooper et al., 2002b). In addition, a study showed that manuka <strong>honey</strong><br />

(MIC 3.4±0.5% (v/v)) h<strong>as</strong> a potential activity to control and prevent infection with<br />

coagul<strong>as</strong>e-negative staphylococci (French et al., 2005)<br />

54


Nezeako & Hamdi, (2000) tested six commercial <strong>honey</strong> samples against control<br />

organisms, Staph.aureus, E. coli, P. aeruginosa and various clinical isolates. They<br />

found that some samples had high broad-spectrum <strong>antimicrobial</strong> activity which<br />

resisted refrigeration temperature for six months and being boiled for 15 minutes. In<br />

addition Mullai and Menon (2007) tested the antibacterial effect <strong>of</strong> different types <strong>of</strong><br />

<strong>honey</strong> against 150 strains <strong>of</strong> P. aeruginosa isolated from otitis media, diabetic foot<br />

ulcers and burns wound. MIC <strong>of</strong> 20% (v/v), 11% (v/v) and 20% (v/v) were<br />

determined from manuka, khadikraft and heather <strong>honey</strong> respectively.<br />

Recently, Blair et al., (2009) studied the <strong>antimicrobial</strong> activity <strong>of</strong> Leptospermum<br />

Medi<strong>honey</strong> with high levels <strong>of</strong> hydrogen peroxide-dependent activity or Comvita<br />

manuka woundcare 18+ against MRSA, Acinetobacter and 6 strains <strong>of</strong> multi-drug<br />

resistant Enterobacteriaceae. MICs ranged from 4 to 5% w/v, 6.0 to 9.3% w/v and<br />

6.3 to 14.8% w/v respectively. This indicated that active Leptospermum <strong>honey</strong> is<br />

potentially active against antibiotic-resistant clinical pathogens.<br />

A survey w<strong>as</strong> carried out with regard to 345 samples <strong>of</strong> New Zealand <strong>honey</strong> to <strong>as</strong>sess<br />

the antibacterial activity <strong>of</strong> <strong>honey</strong>. Four types <strong>of</strong> <strong>honey</strong> were shown to have high<br />

antibacterial activity equivalent to phenol standard. In this <strong>as</strong>say, antibacterial<br />

activity w<strong>as</strong> not detected in almost all <strong>honey</strong>s when catal<strong>as</strong>e w<strong>as</strong> added to remove<br />

hydrogen peroxide. However, manuka and vipers bugloss <strong>honey</strong>s showed<br />

me<strong>as</strong>urable amount <strong>of</strong> hydrogen peroxide which is believed to aid antibacterial<br />

activity (Allen et al., 1991).<br />

1.4.4 Chemical composition <strong>of</strong> <strong>honey</strong>:<br />

Honey is a combination <strong>of</strong> sugars, water, and other compounds (Table 1.1), the<br />

specific composition depending largely on the mix <strong>of</strong> flowers foraged by the bees. It<br />

55


h<strong>as</strong> been reported that approximately 181 substances are present in <strong>honey</strong> (Terrab et<br />

al., 2003).<br />

Table 1.1: Honey composition<br />

Substance Percentages %<br />

Water 17.1<br />

Fructose 38.5<br />

Glucose 31.0<br />

Maltose and other reducing disaccharides 7.2<br />

Sucrose 1.5<br />

Tri-saccharides and other carbohydrates 4.2<br />

Minerals, Vitamins & Enzymes 0.5<br />

Sammataro and Avitabile (1998)<br />

The composition <strong>of</strong> <strong>honey</strong> varies from one <strong>honey</strong> to another depending on several<br />

factors. A major factor is the floral source, <strong>as</strong> the nectar from different plants will<br />

contain different compositions <strong>of</strong> the main sugars and trace elements. These<br />

compositions are influenced by soil type, climatic conditions (se<strong>as</strong>ons) and the<br />

environment surrounding the plant (Crane 1979).<br />

It is recognized that some chemical changes occur when the nectar is transformed to<br />

<strong>honey</strong>. These changes are mainly because <strong>of</strong> some bee enzymes deposited in the<br />

<strong>honey</strong>. These enzymes are invert<strong>as</strong>e that hydrolyses sucrose into glucose and<br />

fructose, amyl<strong>as</strong>e or di<strong>as</strong>t<strong>as</strong>e enzymes and glucose oxid<strong>as</strong>e that generate gluconic<br />

acid and hydrogen peroxide from glucose in diluted <strong>honey</strong>. Other enzymes which are<br />

also present in the <strong>honey</strong> are catal<strong>as</strong>e and acid phosphat<strong>as</strong>e (Sammataro and<br />

Avitabile 1998).<br />

56


In addition, <strong>honey</strong> contains several B vitamins such <strong>as</strong> rib<strong>of</strong>lavin, niacin, folic acid,<br />

and B6. Moreover, <strong>honey</strong> contains a number <strong>of</strong> minerals such <strong>as</strong> calcium, iron, zinc,<br />

pot<strong>as</strong>sium, phosphorus, magnesium, selenium, chromium and manganese (Atrouse et<br />

al., 2004; White 1975). Cooper and Jenkins, (2009) compared the antibacterial<br />

activity <strong>of</strong> medical grade <strong>honey</strong> with 18 table <strong>honey</strong>s collected from different<br />

regions. They found that all table <strong>honey</strong>s have less antibacterial activity compared to<br />

medical grade <strong>honey</strong>. Moreover, all table <strong>honey</strong>s were non sterile and contained<br />

various bacterial species such <strong>as</strong> mesophilic aerobic bacteria, coliforms and<br />

clostridia.<br />

1.4.5 Factors Contributing Antibacterial Properties <strong>of</strong><br />

Honey:<br />

Until 1963, it w<strong>as</strong> thought that the <strong>antimicrobial</strong> properties <strong>of</strong> <strong>honey</strong> were mainly<br />

because <strong>of</strong> hydrogen peroxide, but further studies have indicated that other physical<br />

factors like acidity, osmolarity (Molan 1992a) and electrical conductivity, and<br />

chemical factors including volatile compounds (Yao et al., 2003), antioxidant<br />

(Gheld<strong>of</strong>f et al., 2002; Henriques et al., 2006), beeswax, propolis and pollen (Viuda-<br />

Martos et al., 2008) play a considerable role in <strong>antimicrobial</strong> activity.<br />

1.4.5.1 Osmotic effect:<br />

Honey is a super saturated solution <strong>of</strong> sugar (80%) and water (17%). The osmolarity<br />

<strong>of</strong> <strong>honey</strong> inhibits microbial growth because <strong>of</strong> the strong interaction <strong>of</strong> sugar<br />

molecules with water molecules thus, insufficient water molecules are available to<br />

support microbial growth. This availability is known <strong>as</strong> water activity (aw). Water<br />

involved in many metabolic processes in many organisms. Depending on the<br />

57


permeability <strong>of</strong> cell membrane in each organism the water activity (aw) <strong>of</strong> many<br />

bacterial species is vary between 0.94-0.99. The water activity <strong>of</strong> <strong>honey</strong> is 0.6<br />

because <strong>of</strong> high sugar molecules and low water thus many species cannot grow in<br />

that environment. Fungi can tolerate a lower aw than bacteria, so reports <strong>of</strong> antifungal<br />

activity with diluted <strong>honey</strong> reveal that there are more factors involved than only the<br />

sugar content <strong>of</strong> <strong>honey</strong>. Also, Staph aureus h<strong>as</strong> a high tolerance <strong>of</strong> low aw (0.86) ie.<br />

can tolerate high NaCl level but not high sugar therefore, it considered <strong>as</strong> one <strong>of</strong> the<br />

most susceptible species to the antibacterial activity <strong>of</strong> <strong>honey</strong> (Molan 1992a).<br />

1.4.5.2 Acidity<br />

Honey is quite acidic; normally, it h<strong>as</strong> an average pH <strong>of</strong> 3.9 (with a typical range <strong>of</strong><br />

3.2 to 4.5). It h<strong>as</strong> been known that this acidity is a result the conversion <strong>of</strong> glucose to<br />

gluconic acid with help <strong>of</strong> glucose oxid<strong>as</strong>e enzyme (Molan 2001b). The optimum pH<br />

for growth <strong>of</strong> many bacterial species is 7.2 – 7.4. However, the lowest pH value for<br />

growth <strong>of</strong> some wound pathogens is 4.3 for E .coli and 4.4 for P. aeruginosa. The<br />

low pH <strong>of</strong> <strong>honey</strong> is therefore important to slow down or inhibit bacterial growth<br />

(Bogdanov 1996; Molan 2000a). Since 2001, the osmotic effect w<strong>as</strong> thought to be<br />

the main factor for <strong>antimicrobial</strong> activity (Molan 2001b). However, in 2005 a study<br />

compared <strong>honey</strong> and sugar solution <strong>of</strong> same osmotic effect on coagul<strong>as</strong>e negative<br />

staphylococci. The study confirmed that <strong>antimicrobial</strong> properties are not exclusively<br />

due to osmotic effect (French et al., 2005). It h<strong>as</strong> been noted that the pH <strong>of</strong> <strong>honey</strong><br />

also generates and maintains good environment for fibrobl<strong>as</strong>t activity (Lusby et al.,<br />

2002).<br />

1.4.5.3 Hydrogen peroxide production<br />

In 1919, Sackett reported that in diluted <strong>honey</strong> the antibacterial properties <strong>of</strong> <strong>honey</strong><br />

were incre<strong>as</strong>ed. This is because when <strong>honey</strong> is diluted, hydrogen peroxide is rele<strong>as</strong>ed<br />

58


with the help <strong>of</strong> an enzyme (glucose oxid<strong>as</strong>e) that is found in <strong>honey</strong> (Molan 1992b).<br />

This enzyme is secreted by the hypopharyngeal gland <strong>of</strong> bees and added to nectar<br />

during <strong>honey</strong> formation (Borland 2000).<br />

Glucose + H2O + O2 glucose oxid<strong>as</strong>e gluconic acid + H2O2<br />

Hydrogen peroxide (H2O2) is considered to be one <strong>of</strong> the main factors in antibacterial<br />

activity <strong>of</strong> <strong>honey</strong>. It is involved in cell multiplication in different cell types in the<br />

body <strong>as</strong> certain a concentration <strong>of</strong> H2O2 can support epithelial cells and fibrobl<strong>as</strong>t<br />

growth to repaire damage or injury (Burdon 1995). It also promotes wound healing<br />

by regeneration <strong>of</strong> new capillaries (Tur et al., 1995). The enzyme (glucose oxid<strong>as</strong>e)<br />

is inactive in <strong>full</strong> strength <strong>honey</strong> due to the low pH, so the diluting action <strong>of</strong> fluids<br />

produced by the wound is thought to activate glucose oxid<strong>as</strong>e to produce hydrogen<br />

peroxide. In addition, it stays in the <strong>honey</strong> during storage without losing activity.<br />

Hydrogen peroxide w<strong>as</strong> used for long time to disinfect wounds in hospitals. This<br />

chemical causes damage to the tissues and inflammation due to free radical that is<br />

produced. The levels <strong>of</strong> H2O2 in <strong>honey</strong> are around 1000 times lower than those<br />

applied <strong>as</strong> antiseptic on wounds (Molan 2001b). As a result it does not inflame a<br />

wound or damage the tissue (Bang et al., 2003). Weston (2000) suggested that the<br />

level <strong>of</strong> H2O2 w<strong>as</strong> related to floral source, and that it depended on the balance<br />

between the production and destruction rate <strong>of</strong> H2O2. Destruction <strong>of</strong> H2O2 is due to<br />

catal<strong>as</strong>e which derives from both the pollen and the nectar <strong>of</strong> plants, and the amount<br />

<strong>of</strong> catal<strong>as</strong>e in different sources is variable. In addition, Brudzynski (2006) studied the<br />

effect <strong>of</strong> H2O2 on the antibacterial activity <strong>of</strong> 42 <strong>honey</strong> samples from Canada. She<br />

found that the antibacterial activity w<strong>as</strong> correlated with production <strong>of</strong> H2O2 in <strong>honey</strong>.<br />

59


1.4.5.4 Non-peroxide Components<br />

Several efforts were made to identify the non-peroxide antibacterial components<br />

present in the <strong>honey</strong> (Allen et al., 1991). Weston et al., (1999) separated the<br />

antibacterial phenolic fraction (APF) from the <strong>honey</strong> which consisted <strong>of</strong> benzonic<br />

acids, cinnamic acids and flavonoids. It w<strong>as</strong> determined that APF plays a small role<br />

in manuka <strong>honey</strong> <strong>as</strong> non-peroxide antibacterial component, therefore, there are other<br />

factors which were need to be identified. Honey contains a variety <strong>of</strong> polyphenolic<br />

compounds that may be capable <strong>of</strong> chelating metal ions and decre<strong>as</strong>ing oxidation<br />

(Gheld<strong>of</strong> et al., 2002). Two important cl<strong>as</strong>ses <strong>of</strong> phenolic compounds are flavonoids<br />

and phenolic acid which are known <strong>as</strong> natural antioxidants (Molan 1992a; Pyrznska<br />

and Biesaga 2009; Yao et al., 2003). In a study performed by Wahdan (1998), two<br />

phenolic acids were extracted for the first time; these were caffeic acid and ferulic<br />

acid. Flavonoids had shown a range <strong>of</strong> biochemical and pharmacological actions,<br />

which affect the inflammatory cells and the generation <strong>of</strong> inflammatory processes<br />

(Viuda-Martos et al., 2008). The use <strong>of</strong> flavonoids in medicine is incre<strong>as</strong>ing due to<br />

their ability to trap free radicals, to stimulate hormones and neurotransmitters, and to<br />

inhibit specific enzymes which produce superoxide anions (Pyrznska and Biesaga<br />

2009).<br />

However, it h<strong>as</strong> been identified that several organic components in the ether extract<br />

<strong>of</strong> <strong>honey</strong> possess antibacterial activity; these include 3,5-dimethoxy-4-hydroxy<br />

benzoic acid (syringic acid), and methyl 3,5-dimethoxy-4- hydroxy benzoate (methyl<br />

syringate) (Russel et al., 1988). By using high performance liquid chromatography<br />

(HPLC), some other flavonoids and phenolic acids have also been identified in<br />

different <strong>honey</strong>s, for example, pinocembrin, pinobanksin and chrysin (Bogdanove et<br />

al., 1989), gallic acid and abscisic acid (Yao et al., 2003) caffeic acid and ferulic acid<br />

60


(Wahdan 1998), and vanillic acid, cinnamic acid, and benzoic acid (Weston et al.,<br />

1999; Weston et al., 2000).<br />

1.4.5.5 Antioxidant activity:<br />

Antioxidants are substances that protect wound tissues from being damaged by<br />

oxygen radicals. The free radicals may be produced by hydrogen peroxide and cause<br />

cellular damage. Free radicals are involved in cell toxicity and can alter cell<br />

biomolecules such <strong>as</strong> proteins, carbohydrates, lipids and nucleic acids causing cell<br />

death (Nagai et al., 2001).<br />

Gheld<strong>of</strong> et al., (2002) analysed the antioxidant activity in different <strong>honey</strong> fractions<br />

and determined that most <strong>of</strong> the antioxidant components were found in the water-<br />

soluble fraction. These include gluconic acid, protein, <strong>as</strong>corbic acid,<br />

hydroxymethylfuraldehyde, and the combined activities <strong>of</strong> the enzymes glucose<br />

oxid<strong>as</strong>e, catal<strong>as</strong>e and peroxid<strong>as</strong>e. The same study also showed that the phenolic<br />

compounds in <strong>honey</strong> contributed very significantly to its antioxidant capacity.<br />

When <strong>honey</strong> is diluted the rele<strong>as</strong>e <strong>of</strong> high levels <strong>of</strong> hydrogen peroxide may lead to<br />

tissue damage by formation <strong>of</strong> free radicals such <strong>as</strong> hydroxyl and superoxide. Many<br />

<strong>honey</strong>s including manuka <strong>honey</strong> have the ability to quench free radicals. This<br />

property may play a role in reducing inflammation and chronic wound infection<br />

(Henriques et al., 2006)<br />

A recent study w<strong>as</strong> completed by Van de Berg et al., (2008) with regard to the<br />

antioxidant level in buckweat <strong>honey</strong> showing that this type <strong>of</strong> <strong>honey</strong> reduced the<br />

level <strong>of</strong> reactive oxygen species (ROS) which affect the wound healing process.<br />

Also, beside the low pH and high acidity buckwheat <strong>honey</strong> w<strong>as</strong> shown to contain<br />

high amounts <strong>of</strong> phenolic components that aid the <strong>antimicrobial</strong> mechanisms and<br />

61


lock the oxidative reaction system (Inoue et al., 2005). In addition, several reports<br />

demonstrated the relationship between the antioxidant and the colour <strong>of</strong> <strong>honey</strong>,<br />

where darker <strong>honey</strong> exhibited higher antioxidant content (Bogdanov et al., 2004;<br />

Estevinho et al., 2008; Turkmen et al., 2006). It h<strong>as</strong> been thought that non-hydrogen<br />

peroxide activity in manuka <strong>honey</strong> may be due to plant derived components such <strong>as</strong><br />

flavonoids and phenolic compounds. Recently, two research groups have reported<br />

that the activity <strong>of</strong> Leptospermum <strong>honey</strong>s correlates with the presence <strong>of</strong><br />

methyglyoxal (MG), an alpha-oxoaldehyde that reacts with macromolecules such <strong>as</strong><br />

DNA, RNA and proteins (Adams et al., 2008; Mavric et al., 2008). High amount <strong>of</strong><br />

MG w<strong>as</strong> present in some manuka <strong>honey</strong> which is equivalent to the non-peroxide<br />

activity. MG w<strong>as</strong>, therefore, known <strong>as</strong> a bioactive complex responsible for the<br />

antibacterial activity in manuka <strong>honey</strong> (Mavric et al., 2008).<br />

Recently Atrott and Henle (2009) studied the presence <strong>of</strong> methylglyoxal in 61<br />

samples <strong>of</strong> manuka <strong>honey</strong>. They found that the antibacterial activity ranged between<br />

12.4% to 30.9% equivalent to phenol concentration.<br />

More recently Kwakman et al., (2010) discovered an antibacterial bee peptide called<br />

bee defensin-1 in <strong>honey</strong>. To date this peptide h<strong>as</strong> been isolated only from a <strong>honey</strong><br />

used in the production <strong>of</strong> Revamil and it w<strong>as</strong> confirmed that this protein exhibits<br />

most <strong>of</strong> the antibacterial activity. The exact mechanism <strong>of</strong> bee defensin-1 on bacteria<br />

is not yet known.<br />

62


Table 1.2: Comparisons between peroxide and non-peroxide <strong>honey</strong><br />

Peroxide <strong>honey</strong> Non-peroxide <strong>honey</strong><br />

Form H2O2 Polyphenolic compounds<br />

Effected by heat &<br />

light<br />

Effected by long<br />

storage<br />

Sensitive Thermostable<br />

Yes No<br />

Origin: Bee glucose oxid<strong>as</strong>e Honey acidity, di<strong>as</strong>t<strong>as</strong>e &<br />

invert<strong>as</strong>e enzymes<br />

Origin: Flora - Flavonoids, phenolic acids<br />

Mode <strong>of</strong> action active only when diluted,<br />

tissue repair<br />

63<br />

Antioxidant, MGO<br />

High level Bad- rele<strong>as</strong>e <strong>of</strong> free radical Good- more scavenging<br />

(quenching properties)<br />

Example Almost all <strong>honey</strong> Manuka, Jelly bush <strong>honey</strong>


1.4.5 Manuka Honey:<br />

Manuka <strong>honey</strong> comes from the tree (Leptospermum scoparium) and is currently<br />

approved for therapeutic use in many countries. The <strong>honey</strong> mainly derived from<br />

nectar taken from trees <strong>of</strong> L.scoparium variety is referred to <strong>as</strong> manuka; a closely<br />

related shrub is Kunzea ericoides commonly referred to <strong>as</strong> kanuka. These are shrubs<br />

which form bushes with height <strong>of</strong> 12-15 ft. They belong to the Myrtaceae family and<br />

are native to New Zealand although other species <strong>of</strong> Leptospermum are found in<br />

Australia.<br />

A survey <strong>of</strong> 345 New Zealand <strong>honey</strong>s found that antibacterial activity <strong>of</strong> manuka<br />

<strong>honey</strong> w<strong>as</strong> retained in the presence <strong>of</strong> catal<strong>as</strong>e, and w<strong>as</strong> called non-peroxide activity,<br />

while most other types <strong>of</strong> <strong>honey</strong> were found to be inactive when catal<strong>as</strong>e w<strong>as</strong> added.<br />

The later <strong>honey</strong>s were called peroxide <strong>honey</strong>s. In this study antibacterial <strong>as</strong>says were<br />

performed using phenol <strong>as</strong> a standard and Staphylococcus aureus <strong>as</strong> the test strain.<br />

This developed a me<strong>as</strong>ure <strong>of</strong> activity relating to percentage <strong>of</strong> phenol which had the<br />

same degree <strong>of</strong> antibacterial activity (Allen et al., 1991). This system w<strong>as</strong> then<br />

granted by New Zealand government <strong>as</strong> the UMF or „Unique Manuka Factor‟ to give<br />

a standard for customers. It is now recommended to use <strong>honey</strong> with a UMF <strong>of</strong> 10 or<br />

higher (equivalent activity <strong>of</strong> 10% phenol) to ensure sufficient activity circulate into<br />

deeper tissues where severe wounds are involved. Higher UMF ratings indicate<br />

higher levels <strong>of</strong> antibacterial activity (Molan 2001b). Recently, several rating<br />

systems developed by incre<strong>as</strong>ing the companies that produce manuka <strong>honey</strong>.<br />

Methylglyoxal (MG) and Molan Gold Standard are new registered trademarks from<br />

different organizations to rate the potency <strong>of</strong> manuka <strong>honey</strong><br />

(http://www.molangoldstandard.co.nz/article, 2010).<br />

64


Another study compared the effectiveness <strong>of</strong> manuka <strong>honey</strong> with other peroxide<br />

<strong>honey</strong> against 7 Helicobacter pylori strains isolated from biopsies <strong>of</strong> stomach ulcers.<br />

The MIC w<strong>as</strong> 5% (v/v) for manuka <strong>honey</strong> & >40% (v/v) for other <strong>honey</strong> (Al Somal<br />

et al., 1994).<br />

Some studies have reported the in vitro effectiveness <strong>of</strong> manuka <strong>honey</strong> against<br />

bi<strong>of</strong>ilms produced by 3 species; Pseudomon<strong>as</strong> aeruginosa, methicillin resistant<br />

Staphylococcus aureus (MRSA) & methicillin sensitive Staphylococcus aureus<br />

(MSSA) with killing rate <strong>of</strong> 91%, 63% and 82%, respectively (Alandejani et al.,<br />

2009; Okhiria et al., 2009).<br />

The high level <strong>of</strong> methyl syringate in manuka <strong>honey</strong> w<strong>as</strong> proved to scavenge<br />

superoxide anion radicals (Inoue et al., 2005). In addition, Stephens and his<br />

colleagues (2009) have analysed the phenolic components in manuka and kanuka<br />

<strong>honey</strong>s. Both <strong>honey</strong>s have six phenolic acids in common <strong>as</strong> a primary component. In<br />

the same study elevated levels <strong>of</strong> MG in manuka <strong>honey</strong> acted <strong>as</strong> a source <strong>of</strong><br />

antibacterial activity. Both <strong>of</strong> these components (MG and phenolic compounds)<br />

could contribute to the bacterial effect <strong>of</strong> <strong>honey</strong> in medical use.<br />

The conclusion from these observations is that certain <strong>honey</strong>s contain <strong>antimicrobial</strong><br />

factors such <strong>as</strong> methylglyoxal and bee defensin-1 in addition to sugar content, low<br />

pH and hydrogen peroxide generation.<br />

However, in order for <strong>honey</strong> to be accepted <strong>as</strong> an alternative to <strong>antimicrobial</strong>, it is<br />

desirable to characterise the components that are responsible for its activity.<br />

Although many constituents <strong>of</strong> the antibacterial activity <strong>of</strong> <strong>honey</strong> are known, such <strong>as</strong><br />

the sugars, pH, hydrogen peroxide, and more recently MG and bee defensin-1, these<br />

do not account for the total antibacterial activity observed in many <strong>honey</strong>s. However,<br />

complete identification <strong>of</strong> the antibacterial components h<strong>as</strong> not been achieved<br />

65


ecause <strong>of</strong> the complexity <strong>of</strong> <strong>honey</strong> itself and the possible interaction between<br />

different substances present in the <strong>honey</strong>. Recently, a few studies concentrated on the<br />

effect <strong>of</strong> <strong>honey</strong> against certain bacteria by looking into the intracellular change in<br />

order to find the mechanism <strong>of</strong> action and the target site <strong>of</strong> the <strong>honey</strong>. They found<br />

that 10% (w/v) <strong>of</strong> manuka <strong>honey</strong> w<strong>as</strong> able to cause marked structural changes to<br />

Gram positive S.aureus. The mechanism w<strong>as</strong> not <strong>full</strong>y understood however; the<br />

inhibition <strong>of</strong> the cell cycle with incre<strong>as</strong>ed septa formation is obvious in this study and<br />

is considered <strong>as</strong> one mechanism leading to cell death (Henriques et al., 2009). A<br />

similar study with P. aeruginosa h<strong>as</strong> demonstrated that manuka <strong>honey</strong> induced cell<br />

lysis (Henriques et al., 2010).<br />

1.4.6 Omani Honey:<br />

Beekeeping h<strong>as</strong> been practised since ancient times in Oman. Oman h<strong>as</strong> a wide-<br />

ranged landscape characterized in its dried-up river beds, hills, plains and deserts in<br />

which grow the plants and trees that supply the <strong>honey</strong> bee with the nutrients it<br />

requires. Such trees are palm trees, coconut palms, cereals, limes, vegetables, sugar<br />

cane, frankincense and gum trees (Al-Taie et al., 1999)<br />

In particular, trees such <strong>as</strong> the Simr (Acacia tortilis), Sidr (Ziziphus spina-christi),<br />

Ghaf (Prosopis cineraria), coconut palm, prickly pear and papaya trees make the<br />

primary ingredients that give Omani <strong>honey</strong> its distinctive flavour (Al-Taie et al.,<br />

1999). Honeycombs are formed within one and half months and harvested twice a<br />

year, which is long enough not to be mixed with the date formation period. Acacia<br />

tortilis take part in the production <strong>of</strong> <strong>honey</strong>, because it flowers at a different time <strong>of</strong><br />

year compared to other trees. This expands the se<strong>as</strong>on in which bees can collect<br />

nectar and produce their <strong>honey</strong>. Total polyphenols, flavonoids and antioxidant levels<br />

66


<strong>of</strong> raw <strong>honey</strong> samples from Acacia, have been evaluated. Phenolic content, expressed<br />

<strong>as</strong> caffeic acid equivalents, ranged from 3 to 11 mg/100 g in Acacia. Total flavonoids<br />

in 100 g Acacia <strong>honey</strong> were in the range <strong>of</strong> 0.45–1.01 mg CE (Bl<strong>as</strong>a et al., 2006)<br />

Two types <strong>of</strong> bees have been identified in Oman these are Apis mellifera and Apis<br />

florae. The specialist beekeepers <strong>of</strong> northern Oman have build up a huge skill in<br />

gaining <strong>honey</strong> and proliferating bee colonies in a controlled approach (Sajwani et al.,<br />

2007a).<br />

The distribution area <strong>of</strong> A. florae is commonly restricted to warm climates. In the<br />

west, this species is found in the warmer parts <strong>of</strong> Oman, Iran and Pakistan, through<br />

India and Sri Lanka (Akratanakul 1990). A recent 3 year melissopalynological study<br />

<strong>of</strong> 48 Omani <strong>honey</strong> samples collected from 14 locations in the Muscat and Al Batina<br />

regions <strong>of</strong> Oman w<strong>as</strong> conducted. It w<strong>as</strong> found that 32 <strong>honey</strong> samples among 48 are<br />

mon<strong>of</strong>loral types and the other 16 were multifloral. Overall 122 pollen types from 50<br />

plant families were recognized (Sajwani et al., 2007a). In addition, sugar and protein<br />

pr<strong>of</strong>iles <strong>of</strong> same Omani <strong>honey</strong>s were me<strong>as</strong>ured (Sajwani et al., 2007b)<br />

Al-Jabri et al., (2003) compared the antibacterial activity <strong>of</strong> 16 <strong>honey</strong>s from different<br />

parts <strong>of</strong> Oman, and 8 from different countries in Africa, against three organisms<br />

Staph.aureus, E. coli, P. aeruginosa. It w<strong>as</strong> found that Dh<strong>of</strong>ar <strong>honey</strong> (Oman) and<br />

eucalyptus <strong>honey</strong> (Uganda) had the highest level <strong>of</strong> activity against the three test<br />

organisms. Another study w<strong>as</strong> performed by the same group on the interaction <strong>of</strong><br />

<strong>honey</strong> and bovine milk against S.aureus (Al-Jabri et al., 2005b).<br />

In addition, anti-Staphylococcal activity <strong>of</strong> thirty types <strong>of</strong> Omani <strong>honey</strong> w<strong>as</strong> tested<br />

alone and in combination with gentamicin. It w<strong>as</strong> observed that thirteen <strong>of</strong> the Omani<br />

<strong>honey</strong>s had high anti-Staphylococcus aureus activity. The killing rate for the best<br />

<strong>honey</strong> w<strong>as</strong> 38% <strong>of</strong> Staphylococcus aureus using 50% (w/v) concentration <strong>honey</strong> in<br />

67


30 minutes. Gentamicin (4µg/ml) killed 70% <strong>of</strong> isolate; <strong>honey</strong> and gentamicin were<br />

combined to give excellent killing rate at 92% in the same duration (Al-Jabri et al.,<br />

2005a).<br />

The first study <strong>of</strong> the ability <strong>of</strong> <strong>honey</strong> to prevent bacterial adherence in-vitro w<strong>as</strong><br />

done by Al-Naqdy et al., (2005). Four different types <strong>of</strong> Omani <strong>honey</strong> were used in<br />

this study <strong>of</strong> growth inhibition. Bacterial adherence w<strong>as</strong> examined using Salmonella<br />

enteritidis that had been incubated first with <strong>honey</strong> and then with intestinal epithelial<br />

cells. Results showed decre<strong>as</strong>es in the number <strong>of</strong> bacteria attached to the treated<br />

epithelial cells from 25.6±6.5 to 6.7±3.3 bacteria per epithelial cell (P


1.5 Aims and Objectives:<br />

The aim <strong>of</strong> this study w<strong>as</strong> to investigate the inhibition <strong>of</strong> MDR bacteria including<br />

ESBLs by a medical grade <strong>honey</strong> and a selection <strong>of</strong> Omani <strong>honey</strong>s. The following<br />

objectives were identified:<br />

1. To investigate the effect <strong>of</strong> a medical grade manuka <strong>honey</strong> on MDR bacteria<br />

possessing ESBLs by determining:<br />

� The MIC and MBC <strong>of</strong> manuka <strong>honey</strong> against selected organisms<br />

� The bactericidal activity <strong>of</strong> manuka <strong>honey</strong> using time-to-kill curves <strong>as</strong>say<br />

� The surface and intra-structural morphological changes <strong>of</strong> bacterial cells after<br />

treatment with manuka <strong>honey</strong> using scanning and transmission electron<br />

microscopy.<br />

� the physiological effect <strong>of</strong> manuka <strong>honey</strong> against selected organisms using<br />

two-dimensional electrophoresis<br />

2. To investigate the potential <strong>of</strong> Omani <strong>honey</strong>s for their use in wound care in<br />

Oman by:<br />

� Characterizing the physical and chemical properties <strong>of</strong> selected Omani <strong>honey</strong>.<br />

� Investigating the <strong>antimicrobial</strong> activity <strong>of</strong> Omani <strong>honey</strong> against MDR and<br />

ESBLs bacteria<br />

� Investigating the antioxidant potential activity <strong>of</strong> selected Omani <strong>honey</strong><br />

69


Chapter 2<br />

Materials &<br />

Methods<br />

70


The materials used throughout this study are listed <strong>as</strong> chemicals and re<strong>agent</strong>s (Table<br />

2.1, 2.2 & 2.3), equipment and apparatus (Table 2.4 & 2.5), and MDR and ESBL<br />

clinical isolates (Table 2.3 and 2.4, respectively).<br />

Table 2.1: Media, Chemicals and Re<strong>agent</strong>s used for general experiment in the<br />

<strong>project</strong>:<br />

Name <strong>of</strong> Chemical Suppliers<br />

Nutrient agar CM 0003 Oxoid<br />

Mueller-Hinton agar (MH) CM0337 Oxoid<br />

Eosin Methylene Blue agar (EMB) CM0069 Oxoid<br />

Iso-sensitest broth (ISB) CM 0473 Oxoid<br />

Tryptone Soya Broth (TSB) Oxoid<br />

Maximum recovery diluent (MRD) Oxoid<br />

Catal<strong>as</strong>e 010M7010 Sigma-Aldrich<br />

Nutrient broth (NB) Oxoid<br />

Phenol Fisher<br />

Staph aureus – Oxford strain- NCTC 6571 Selectrol<br />

Bovine albumin serum (BSA) Sigma<br />

DPPH (2,2-Di(4-tert-octylphenyl)-1-picryl-hydrazyl)<br />

Free radical – 257621<br />

71<br />

Sigma-Aldrich


Table 2.2: Chemicals and Re<strong>agent</strong>s used for electron microscopy experiment in<br />

the <strong>project</strong>:<br />

Name <strong>of</strong> chemicals / re<strong>agent</strong>s Suppliers<br />

Phosphate Buffered Saline (PBS) Oxoid<br />

Glutaraldehyde 3% Fluka-Sigma<br />

Osmium tetroxide 1% (OsO4)<br />

72<br />

Agar scientific<br />

Ethanol Sigma-Aldrich<br />

Filter membrane (0.02mm pore size) Agar scientific<br />

Resin (no propylene oxide) Agar scientific<br />

Araldite resin CY212 Agar scientific<br />

DDSA Agar scientific<br />

BDMM/propylene oxide Agar scientific<br />

Uranyl acetate Sigma<br />

Reynolds lead citrate Sigma


Table 2.3: Chemicals and Re<strong>agent</strong>s used for 2- dimensional electrophoresis<br />

experiment in the <strong>project</strong><br />

Name <strong>of</strong> chemicals / re<strong>agent</strong>s Suppliers<br />

DC protein <strong>as</strong>say re<strong>agent</strong> A - 8473 BioRad<br />

DC protein <strong>as</strong>say re<strong>agent</strong> B - 500-0114 BioRad<br />

2,4,6-Tris(2-pyridyl)-s-triazine- T1253 Sigma-Aldrich<br />

Trizma ® b<strong>as</strong>e (C4H11NO3) BCBC0475 Sigma-Aldrich<br />

Iodoacetamide (C2H4INO) 030M5300 Sigma-Aldrich<br />

MOPS (3-Morpholino-Propansulfonsaure acid – 69947 Fluka, Sigma-Aldrich<br />

DL-Dithiothreitol (C4H10O2S2) D9163 Sigma-Aldrich<br />

Trizma ® hydrochloride (C4H11NO3) T3253 Sigma-Aldrich<br />

Sodium dodecylsulfate (C12H25NaO4S) L3771 Sigma-Aldrich<br />

CHAPS (C32H58N2O75) C9426 Sigma-Aldrich<br />

Simply Blue Safe stain LC6060 Invitrogen<br />

Ethylenediaminetetraacetic acid tetr<strong>as</strong>odium sodium<br />

salt dihydrate E6511<br />

73<br />

Sigma-Aldrich<br />

Glycerol G8773 Sigma-Aldrich<br />

Sodium chloride NaCl Fisher scientific<br />

Urea (CH4 N2O) 46504 Sigma-Aldrich<br />

Bromophenol Blue B0126 Sigma-Aldrich<br />

Agarose- low melting point A9414 Sigma-Aldrich


Table 2.4: Equipment used in general experiments in the <strong>project</strong>:<br />

Name <strong>of</strong> Materials Suppliers<br />

Automatic pipettes Eppendorf<br />

Volumetric fl<strong>as</strong>ks Volac-UK<br />

Conical fl<strong>as</strong>ks PYREX-UK<br />

Autoclave LTE scientific<br />

Micro-centrifuge Sanyo-MSE<br />

Weighing scale DP-300 Fisher brand<br />

Microtitre plate/96wells Nunc (flat bottomed) Fisher scientific Nuclon <br />

74<br />

Surface<br />

Bio<strong>as</strong>say plate CORNING 43110<br />

Multipoint inoculator MAST<br />

Spectrophotometer/Plate reader MRX Revelation, Dynex<br />

BBL Sens-Disc for ESBL BBL<br />

Oxoid Sens-Disc Oxoid<br />

Spectrophotometer JENWAY 6705UV Vis<br />

Disc dispenser Oxoid<br />

AmpC & ESBL ID set D68C MAST Diagnostic-UK<br />

Incubator 37 o C BINDER<br />

pH meter 3510 JENWAY<br />

Water refractometer Atago HHR-2N<br />

Sugar refractometer (40-85%) Bellingham & Stanley<br />

Water bath 37 o C/100 OLS200-Grant<br />

BioPak (Synergy UV) B0901


Table 2.5: Equipment used in electron microscopy and 2-dimentional<br />

electrophoresis experiments in the <strong>project</strong>:<br />

Name <strong>of</strong> materials Suppliers<br />

Critical point dryer (CPD) Balzers CPD-030<br />

Gold sputter coater AE 1232 EM Scope UK<br />

Scanning electron microscopy (SEM) JEOL 5600V SEM- UK<br />

Transmission electron microscopy (TEM) Leica-Reichert- Jung UK &<br />

Ready strip 11cm pH 3-10 BioRad<br />

Protean IEF cell & Tank BioRad<br />

75<br />

JEOL 1230 TEM- UK<br />

Refrigerated superspeed centrifuge Sorvell ® RC-5B<br />

Thirty isolates <strong>of</strong> Acinetobacter baumannii and Acinetobacter<br />

baumannii/calcoaceticus complex that were resistant to third generation<br />

cephalosporins (3 rd G cephalosporin) were kindly provided by the Department <strong>of</strong><br />

Medical Microbiology in the <strong>University</strong> <strong>of</strong> Wales Hospital at Heath Park, Cardiff.<br />

Antibiotic sensitivities had already been evaluated in the hospital using antibiotic<br />

sensitivity discs for cefotaxime (CTX 30µg), ceftazidime (CAZ 30µg), ceftriaxone<br />

(CRO 30µg) & cefoxitin (FOX30µg). This information w<strong>as</strong> provided with the<br />

isolates (Table 2.6).


Table 2.6: Thirty isolates <strong>of</strong> MDR Acinetobacter<br />

S. no. Identification <strong>of</strong> isolates<br />

1. Acinetobacter baumannii /calcoaceticus complex<br />

2. Acinetobacter baumannii/calcoaceticus complex<br />

3. Acinetobacter baumannii<br />

4. Acinetobacter baumannii<br />

5. Acinetobacter baumannii<br />

6. Acinetobacter baumannii/calcoaceticus complex<br />

7. Acinetobacter baumannii/calcoaceticus complex<br />

8. Acinetobacter baumannii/calcoaceticus complex<br />

9. Acinetobacter baumannii/calcoaceticus complex<br />

10. Acinetobacter baumannii/calcoaceticus complex<br />

11. Acinetobacter baumannii<br />

12. Acinetobacter baumannii/calcoaceticus complex<br />

13. Acinetobacter baumannii<br />

14. Acinetobacter baumannii<br />

15. Acinetobacter baumannii/calcoaceticus complex<br />

16. Acinetobacter baumannii/calcoaceticus complex<br />

17. Acinetobacter baumannii/calcoaceticus complex<br />

18. Acinetobacter baumannii<br />

19. Acinetobacter baumannii<br />

20. Acinetobacter baumannii<br />

21. Acinetobacter baumannii<br />

22. Acinetobacter baumannii<br />

23. Acinetobacter baumannii<br />

24. Acinetobacter baumannii<br />

25. Acinetobacter baumannii<br />

26. Acinetobacter baumannii<br />

27. Acinetobacter baumannii<br />

28. Acinetobacter baumannii<br />

29. Acinetobacter baumannii<br />

30. Acinetobacter baumannii<br />

76


Fifty five clinical isolates belonging to the Enterobacteriaceae family that were resistant<br />

to third generation cephalosporins (3 rd G cephalosporin) with their mechanisms <strong>of</strong> ESBL<br />

production by molecular method were also provided for this study (Table 2.7a & 2.7b).<br />

Isolates were stored at -80ºC on Protect beads until required. When needed, one bead<br />

w<strong>as</strong> introduced into 10 ml nutrient broth and incubated overnight at 37 o C. The broth<br />

culture w<strong>as</strong> either used directly or streaked onto nutrient agar and incubated at 37ºC<br />

overnight.<br />

Table 2.7a: Clinical isolates <strong>of</strong> E.coli & Klebsiella provided for this study<br />

Identification <strong>of</strong> isolates Mechanisms<br />

31. Escherichia coli CTX-M<br />

32. Escherichia coli CTX-M<br />

33. Escherichia coli CTX-M<br />

34. Escherichia coli CTX-M<br />

35. Escherichia coli CTX-M<br />

36. Escherichia coli TEM<br />

37. Escherichia coli TEM<br />

38. Escherichia coli AMPC<br />

39. Escherichia coli N.A<br />

40. Escherichia coli N.A<br />

41. Klebsiella pneumoniae SHV<br />

42. Klebsiella pneumoniae CTX-M<br />

43. Klebsiella pneumoniae CTX-M<br />

44. Klebsiella pneumoniae CTX-M<br />

45. Klebsiella oxytoca CTX-M<br />

46. Klebsiella pneumoniae SHV<br />

47. Klebsiella pneumoniae SHV<br />

48. Klebsiella oxytoca SHV<br />

49. Klebsiella pneumoniae Pl<strong>as</strong>midic ampC<br />

50. Klebsiella oxytoca K1<br />

51. Klebsiella pneumoniae N.A<br />

NA = not available<br />

77


Table 2.7b: Clinical isolates <strong>of</strong> Citrobacter, Enterobacter & Serratia provided for<br />

this study<br />

Identification <strong>of</strong> isolates Mechanisms<br />

52. Citrobacter farmeri CTX-M<br />

53. Citrobacter farmeri Hyper ampC expression<br />

54. Citrobacter farmeri Hyper ampC expression +CTX-M<br />

55. Citrobacter farmeri Hyper ampC expression +TEM<br />

56. Citrobacter farmeri Hyper ampC expression +CTX-M<br />

57. Citrobacter farmeri Hyper ampC expression<br />

58. Citrobacter farmeri Hyper ampC expression<br />

59. Citrobacter farmeri Hyper ampC expression<br />

60. Citrobacter farmeri Hyper ampC expression<br />

61. Citrobacter farmeri Hyper ampC expression<br />

62. Citrobacter farmeri Hyper ampC expression +TEM<br />

63. Citrobacter farmeri Hyper ampC expression +CTX-M<br />

64. Enterobacter cloacae CTX-M<br />

65. Enterobacter cloacae CTX-M<br />

66. Enterobacter cloacae CTX-M<br />

67. Enterobacter cloacae CTX-M<br />

68. Enterobacter hormaechi CTX-M<br />

69. Enterobacter cloacae CTX-M<br />

70. Enterobacter cloacae CTX-M<br />

71. Enterobacter aerogenes AMPC<br />

72. Enterobacter cloacae TEM<br />

73. Enterobacter cloacae Hyper ampC expression<br />

74. Enterobacter cloacae Hyper ampC expression<br />

75. Enterobacter cloacae Hyper ampC expression<br />

76. Enterobacter cloacae Hyper ampC expression<br />

77. Enterobacter cloacae Hyper ampC expression<br />

78. Serratia plymuthica CTX-M<br />

79. Serratia marcescens Hyper ampC expression<br />

80. Serratia marcescens Hyper ampC expression<br />

81. Serratia marcescens Hyper ampC expression<br />

82. Serratia marcescens Hyper ampC expression<br />

83. Serratia marcescens Hyper ampC expression<br />

84. Serratia marcescens Hyper ampC expression +SHV<br />

85. Serratia marcescens Hyper ampC expression<br />

78


2.1 Characterization <strong>of</strong> test organisms:<br />

2.1.1 Confirmation <strong>of</strong> the identity <strong>of</strong> test organisms:<br />

To confirm the identity <strong>of</strong> these isolates, each culture w<strong>as</strong> tested with API 20E<br />

(BioMerieux, Becton & Dickinson) according to the manufacturer‟s instructions. To<br />

verify that the kits gave reliable results, two known organisms (Escherichia coli<br />

NCTC 10410 and Klebsiella oxytoca NCTC 8167) were tested on each occ<strong>as</strong>ion.<br />

2.1.2 Antibiotics sensitivities: Extended Spectrum Beta-<br />

lactam<strong>as</strong>es test (ESBLs):<br />

Antibiotic sensitivity <strong>of</strong> the clinical isolates w<strong>as</strong> also confirmed and the method used<br />

w<strong>as</strong> that <strong>of</strong> Clinical and Laboratory Standard Institute (CLSI 2006).<br />

2.1.2.1 Inoculum preparation:<br />

Three to four similar colonies were selected from a pure overnight nutrient agar plate<br />

culture and transferred with inoculation loop into 4-5 ml <strong>of</strong> nutrient broth and<br />

incubated at 37°C for 2-6 h. The inocula were standardized photometrically to obtain<br />

turbidity equivalent to the 0.5 McFarland standards.<br />

2.1.2.2 Disc application:<br />

A sterile cotton wool swab w<strong>as</strong> dipped into the standardised inocula and excess<br />

culture removed by pressing against the edge <strong>of</strong> the culture bottle. The entire agar<br />

surface <strong>of</strong> a Mueller Hinton plate w<strong>as</strong> streaked in three directions to produce an even<br />

lawn plate. Then, the appropriate discs (Oxoid Sens-Disc) <strong>of</strong> carbapenem (imipenem<br />

10µg and meropenem 10µg), third and fourth generation cephalosporins (cefotaxime<br />

CTX 30µg, ceftazidime CAZ 30µg and cefepime FEP 30µg) respectively,<br />

aminoglycosides (amikacin AK 30µg & gentamicin CN 10µg) and piperacillin-<br />

tazobactam TZP 100/10 µg were applied for all isolates by disc dispenser.<br />

79


Appropriate discs (BD BBL TM Sens-Disc) <strong>of</strong> ceftazidime (CAZ 30µg), cefotaxime<br />

(CTX 30µg), ceftazidime with clavulanic acid (CAZ/CLA 30/10µg) and cefotaxime<br />

with clavulanic acid (CTX/CLA 30/10µg) were also dispensed for the 55<br />

Enterobacteriaceae isolates. The plates were placed in the 37°C incubator within 15<br />

minutes and incubated for 16-18 h. After incubation, the plates were examined and<br />

zone diameters were recorded. Antibiotic sensitivity w<strong>as</strong> determined by reference to<br />

the (CLSI).<br />

2.1.2.3 Screening test for ESBL:<br />

If the zones for each <strong>of</strong> CAZ 30µg ≤ 22mm, CTX 30µg ≤ 27mm and CRO 30µg ≤<br />

25mm, ESBL production w<strong>as</strong> indicated.<br />

2.1.2.4 Phenotypic confirmatory test for ESBL:<br />

A confirmatory test w<strong>as</strong> required for each <strong>of</strong> ceftazidime and cefotaxime in<br />

combination with clavulanic acid. For comparison with zone sizes without<br />

clavulanic acid a ≥ 5 mm incre<strong>as</strong>e in zone diameter for either <strong>antimicrobial</strong> <strong>agent</strong><br />

tested in combination with clavulanic acid versus its zone when tested alone<br />

confirmed an ESBL producer.<br />

A positive control (ESBL producer) <strong>of</strong> Escherichia coli ATCC 13353 and a negative<br />

control (non-ESBL producer) Escherichia coli NCTC 10418 were used for quality<br />

control <strong>of</strong> this method.<br />

2.1.2.5 ESBL/AmpC * confirmation test:<br />

This test w<strong>as</strong> used for the detection <strong>of</strong> AmpC and/or ESBL enzyme production. Four<br />

discs were applied: cefpodoxime (10 µg), cefpodoxime (10µg) with ESBL inhibitor,<br />

cefpodoxime (10µg) with AmpC inhibitor and cefpodoxime (10µg) with ESBL and<br />

AmpC inhibitor. The interpretation <strong>of</strong> results w<strong>as</strong> determined using s<strong>of</strong>tware product<br />

<strong>of</strong> ESBL/AmpC calculator (EAC) programme provided by MAST manufacturer.<br />

80


2.2 Characterization <strong>of</strong> <strong>honey</strong> samples:<br />

2.2.1 Honey sample collection:<br />

A medical grade <strong>honey</strong> (manukacare 18+), which w<strong>as</strong> kindly provided by Comvita<br />

UK, w<strong>as</strong> used throughout this study. Eight samples <strong>of</strong> raw and unprocessed Omani<br />

<strong>honey</strong> (labelled OH-B to OH-I) were collected from different regions <strong>of</strong> Oman<br />

(Table 2.8). The floral source w<strong>as</strong> estimated by beekeepers. All samples were stored<br />

at 4°C in the dark until tested.<br />

Table 2.8: List <strong>of</strong> <strong>honey</strong>s used in this <strong>project</strong>:<br />

Batch Number Flora Source Country Regions<br />

A Manuka New Zealand -<br />

OH-B Multi-flora Oman Batina<br />

OH-C Honey dew Oman Dakhilyia<br />

OH-D Honey dew Oman Batina<br />

OH-E Honey dew Oman Dhahira<br />

OH-F Citrus Oman Sumail<br />

OH-G Honey dew Oman Batina<br />

OH-H Acacia Oman Sharqiya<br />

OH-I Market <strong>honey</strong> Oman Not known<br />

2.2.2 Bio<strong>as</strong>say <strong>of</strong> antibacterial activity:<br />

The antibacterial activity <strong>of</strong> all <strong>honey</strong> samples w<strong>as</strong> determined by the method<br />

described by Allen et al., (1991).<br />

81


2.2.2.1 Phenol standards preparation for bio<strong>as</strong>say:<br />

10% (w/v) <strong>of</strong> phenol (a reference antiseptic) (Fisher) w<strong>as</strong> prepared by weighing 5 g <strong>of</strong><br />

phenol made up to 50 ml with deionised water. This solution <strong>of</strong> phenol w<strong>as</strong> used to<br />

prepare the standard solutions <strong>of</strong> 2%, 3%, 4%, 5%, 6% and 7% (Table 2.9).These<br />

standards were stored at 4 o C till one month.<br />

Table 2.9: Preparation <strong>of</strong> phenol standards<br />

Final Phenol Conc (%w/v) Volume <strong>of</strong> 10%<br />

Phenol/ml<br />

82<br />

Volume <strong>of</strong> H2O/ml<br />

2% 2 8<br />

3% 3 7<br />

4% 4 6<br />

5% 5 5<br />

6% 6 4<br />

7% 7 3<br />

2.2.2.3 Inoculum preparation:<br />

One bead <strong>of</strong> a freeze dried culture <strong>of</strong> Staph.aureus NCTC 6571(Selectrol) w<strong>as</strong><br />

dispensed into 10 ml nutrient broth (Oxoid) using <strong>as</strong>eptic technique and incubated at<br />

37 o C for 18 h. Staph. aureus w<strong>as</strong> universally used in antibiotic sensitivity testing.<br />

2.2.2.4 Plate preparation:<br />

Nutrient agar (Oxoid) w<strong>as</strong> prepared by weighing 3.45g <strong>of</strong> nutrient agar and dissolving<br />

in 150 ml <strong>of</strong> deionised water. The agar w<strong>as</strong> mixed and sterilised by autoclaving at<br />

120 o C. After autoclaving, the agar w<strong>as</strong> kept at 50 o C in water bath for 30 minutes.<br />

While waiting for the agar, the nutrient broth with Staph. aureus (working culture)<br />

w<strong>as</strong> adjusted to an absorbance <strong>of</strong> 0.5 me<strong>as</strong>ured at 540 nm in a Cecil spectrophotometer<br />

using sterile nutrient broth <strong>as</strong> a blank. 100 µl <strong>of</strong> working culture <strong>of</strong> Staph. aureus w<strong>as</strong><br />

added to the molten nutrient agar at 50 o C. Then the agar w<strong>as</strong> mixed thoroughly and


transfered into a large square <strong>as</strong>say plate (Corning 43110) on flat surface. When the<br />

agar had set, the plate w<strong>as</strong> layed upside down at 4 o C and left overnight.<br />

2.2.2.5 Sample preparation:<br />

Eight universal containers were labelled <strong>as</strong> A1, B1, C1, D1, E1, F1, G1, H1 and I1 one<br />

for each <strong>of</strong> the eight types <strong>of</strong> Omani <strong>honey</strong> used in this <strong>project</strong>. Stock <strong>honey</strong> solution<br />

w<strong>as</strong> prepared by adding 10g <strong>of</strong> well mixed <strong>honey</strong> to 10 ml <strong>of</strong> deionised water,<br />

therefore the <strong>final</strong> <strong>honey</strong> concentration w<strong>as</strong> 50% w/v. The solution w<strong>as</strong> then placed in<br />

a water bath at 37 o C for 30 minutes to aid dissolving and mixing. To prepare further<br />

dilutions, another eight universal containers were labelled <strong>as</strong> A2, B2, C2, D2, E2, F2,<br />

G2, H2 and I2 and w<strong>as</strong> transferred 1 ml <strong>of</strong> 50% <strong>honey</strong> (stock solution) to 1 ml <strong>of</strong><br />

deionised water to make the <strong>final</strong> concentration <strong>of</strong> 25% <strong>honey</strong>.<br />

For non peroxide activity testing 0.02g <strong>of</strong> catal<strong>as</strong>e from bovine liver (Sigma) w<strong>as</strong><br />

made up to 10 ml with deionised water. The solution w<strong>as</strong> mixed gently. Then eight<br />

universal containers were labelled <strong>as</strong> A3, B3, C3, D3, E3, F3, G3, H3 and I3. 1 ml <strong>of</strong><br />

50% <strong>honey</strong> (stock <strong>honey</strong>) w<strong>as</strong> transferred to these tubes <strong>as</strong>eptically and 1 ml <strong>of</strong><br />

catal<strong>as</strong>e solution w<strong>as</strong> added to these tubes to make 25% (w/w) <strong>honey</strong> and catal<strong>as</strong>e<br />

solution.<br />

2.2.2.6 Samples and standards application:<br />

The bio<strong>as</strong>say plate w<strong>as</strong> placed over a qu<strong>as</strong>i-latin square <strong>as</strong> a template after it w<strong>as</strong><br />

removed from the cold room (4 o C). A flamed, cooled 8 mm cork borer w<strong>as</strong> used to cut<br />

out 64 wells <strong>of</strong> the agar. Cut agar w<strong>as</strong> removed with a sterile blade into a discard pot.<br />

A 100 µl <strong>of</strong> each sample w<strong>as</strong> added to each <strong>of</strong> 4 wells with the same number <strong>as</strong>signed<br />

on the <strong>as</strong>say plate. Thus, each sample w<strong>as</strong> tested in quadruplicate. Similarly, 100 µl<br />

phenol standards (2%-7%) were tested in duplicate. The plate w<strong>as</strong> then incubated at<br />

37 o C for 18 h.<br />

83


2.2.2.7 Zone me<strong>as</strong>urement:<br />

After incubation, the plate w<strong>as</strong> placed back over black paper to me<strong>as</strong>ure the diameter<br />

<strong>of</strong> the zones <strong>of</strong> inhibition with digital calipers. Each zone <strong>of</strong> inhibition w<strong>as</strong> me<strong>as</strong>ured<br />

twice (horizontal and vertical reading) at 90°angles.<br />

2.2.2.8 Calculation <strong>of</strong> antibacterial activity <strong>of</strong> <strong>honey</strong>:<br />

After me<strong>as</strong>urement <strong>of</strong> the clear zone around each phenol standard twice, the mean<br />

diameter w<strong>as</strong> calculated and squared. A standard graph w<strong>as</strong> plotted <strong>of</strong> % (w/v) phenol<br />

against the square <strong>of</strong> the mean diameter <strong>of</strong> the clear zone. A best straight line shape<br />

w<strong>as</strong> plotted using Cricket graph s<strong>of</strong>tware and the equation <strong>of</strong> this line w<strong>as</strong> used to<br />

obtain the activity <strong>of</strong> each diluted <strong>honey</strong> sample. The diameter <strong>of</strong> the clear zone w<strong>as</strong><br />

me<strong>as</strong>ured and squared. However, the dilution factor is correlation to the density <strong>of</strong><br />

<strong>honey</strong> thus this value w<strong>as</strong> multiplied by a factor <strong>of</strong> 4.69. In short to obtain the dilution<br />

from 50% w/w solution <strong>of</strong> <strong>honey</strong>, 10 ml <strong>of</strong> water were added to 10 g <strong>of</strong> <strong>honey</strong>. Ten g<br />

<strong>of</strong> <strong>honey</strong> is actually equal to 7.41 ml where the average density <strong>of</strong> <strong>honey</strong> is 1.35 g/ml<br />

i.e. 10/1.35 = 7.41. Therefore, 50% <strong>honey</strong> solution should be made by adding 10 ml <strong>of</strong><br />

water to 7.41 ml <strong>of</strong> <strong>honey</strong> which w<strong>as</strong> actually equal to 42.56% (v/v) and 25% <strong>honey</strong><br />

solution w<strong>as</strong> equal to 21.28% (v/v). To obtain the phenol percentage equivalent <strong>of</strong> <strong>full</strong><br />

strength <strong>honey</strong> it required to multiply by a factor <strong>of</strong> 4.69 i.e 100/21.28. Then the<br />

activity <strong>of</strong> <strong>honey</strong> obtained from the <strong>as</strong>say w<strong>as</strong> expressed <strong>as</strong> the equivalent phenol<br />

concentration (% w/v) (Molan, personal communication).<br />

84


2.2.3 Determination <strong>of</strong> pH:<br />

This method w<strong>as</strong> derived from the method described by the International Honey<br />

Commission (Bogdanov, 2002).<br />

All <strong>honey</strong> samples were removed from the fridge and kept at 20°C for 24 hrs in an air-<br />

conditioned laboratory prior to testing. A 50% (w/v) solution <strong>of</strong> <strong>honey</strong> w<strong>as</strong> made by<br />

weighing 5g <strong>of</strong> <strong>honey</strong> and dissolved in ultrapure water (BioPak- B0901) to 10 ml. The<br />

solution w<strong>as</strong> gently shaken until the <strong>honey</strong> sample had completely dissolved. A<br />

JENWAY instrument 3510 pH meter w<strong>as</strong> calibrated at pH 4, 7 and 10 using reference<br />

buffers (Fisher). After calibration the probe w<strong>as</strong> inserting into the diluted sample. Each<br />

sample w<strong>as</strong> tested in triplicate. Between each sample the probe w<strong>as</strong> rinsed under<br />

running water and dried. Before testing the next sample, the probe w<strong>as</strong> then placed in<br />

deionised water.<br />

2.2.4 Sugar and water content:<br />

This method w<strong>as</strong> established from the method described by the International Honey<br />

Commission (Bogdanov 2002).<br />

Sugar and water content were determined using a Bellingham & Stanley 40-85% sugar<br />

refractometer and an Atago HHR-2N moisture refractometer. Sugar and moisture<br />

content are usually tested at 20°C. Each <strong>of</strong> the <strong>honey</strong> samples were allowed to set to<br />

20°C for 24 hours before the testing started. The whole <strong>honey</strong> sample w<strong>as</strong> mixed with<br />

a sterile spatula then a drop <strong>of</strong> <strong>honey</strong> w<strong>as</strong> placed in the lens <strong>of</strong> each <strong>of</strong> the<br />

refractometers. To confirm an equal distribution <strong>of</strong> the sample over the lens with no<br />

air bubbles, the lids above the lens were care<strong>full</strong>y closed. To read the scale, the<br />

refractometers were held directed to the light and the spot <strong>of</strong> the line w<strong>as</strong> recorded.<br />

The refractometers were cleaned with running water and dried with clean tissue<br />

between each sample.<br />

85


2.2.5 Hydroxymethylfurfural (HMF) concentration:<br />

The method used w<strong>as</strong> that <strong>of</strong> White & Rudji (1978).<br />

Into 50 ml volumetric fl<strong>as</strong>ks, five grams from each <strong>of</strong> the <strong>honey</strong> samples were<br />

weighed with a total <strong>of</strong> 25 ml <strong>of</strong> deionised water. Then 0.50 ml <strong>of</strong> Carrez solution I<br />

(15 g pot<strong>as</strong>sium ferrocyanide in 100 ml <strong>of</strong> water) w<strong>as</strong> added to each fl<strong>as</strong>k and mixed<br />

well. After mixing, 0.50 ml <strong>of</strong> Carrez solution II (30 g zinc acetate in 100 ml <strong>of</strong> water)<br />

w<strong>as</strong> added to each fl<strong>as</strong>k. The solutions were diluted to volume with water and filtered<br />

through Whatman filter paper, discarding the first 10 ml. Then 5 ml <strong>of</strong> the remaining<br />

filtrate were added to each <strong>of</strong> two test tubes (18 x 150 mm). 5 ml <strong>of</strong> water were added<br />

to the first tube labelled <strong>as</strong> sample, and 5 ml <strong>of</strong> 0.20% sodium bisulphite (breaks down<br />

HMF) were added into second tube lablled <strong>as</strong> blank. Then a vortex mixer w<strong>as</strong> used to<br />

mix the solutions and the absorbance w<strong>as</strong> read in a Cecil spectrophotometer at 284 and<br />

336 nm. The HMF value w<strong>as</strong> calculated using the following formula:<br />

HMF (mg/Kg <strong>honey</strong>) = (A284 - A336) x 14.97 x 5/weight <strong>of</strong> sample in g.<br />

2.2.6 Protein content:<br />

This method w<strong>as</strong> established from the DC Protein <strong>as</strong>say kit (BioRad) that is b<strong>as</strong>ed on<br />

the Lowry method for the determination <strong>of</strong> protein content in <strong>honey</strong> (White and Rudji<br />

1978).<br />

Cellulose dialysis tubing is a selective membrane which allows low molecular weight<br />

molecules such <strong>as</strong> sugar to p<strong>as</strong>s through this membrane. However, protein molecules<br />

cannot cross the membrane and remain in the tubing because <strong>of</strong> the pore size <strong>of</strong><br />

membrane is smaller than the size <strong>of</strong> protein. The diameter <strong>of</strong> the inflated tube w<strong>as</strong> 16<br />

mm and a cut <strong>of</strong>f <strong>of</strong> 12 kDa MWCO. The tubing w<strong>as</strong> cut into lengths <strong>of</strong> 30-33 cm and<br />

placed under running tap water to hydrate and open the tubing. Two knots were tied<br />

86


once the tubing opened at one end and it w<strong>as</strong> stored until required in a beaker <strong>of</strong> tap<br />

water. To prepare the <strong>honey</strong> solution for dialysis, 5 g <strong>of</strong> <strong>honey</strong> w<strong>as</strong> dissolved with 10<br />

ml <strong>of</strong> deionised water into a small beaker. Honey solution w<strong>as</strong> poured into a section <strong>of</strong><br />

the inflated tubing. To facilitate the transfer <strong>of</strong> the solution, the tubing w<strong>as</strong> clipped to a<br />

funnel. Then total volume <strong>of</strong> 10 ml <strong>of</strong> water were used to rinse the beaker twice and<br />

w<strong>as</strong>hed out any residue. This water w<strong>as</strong> transferred into the tubing. The air w<strong>as</strong><br />

expelled from the dialysis sac and two knots tied above the liquid. The tubing w<strong>as</strong><br />

inverted several times to ensure mixing <strong>of</strong> the solution. The dialysis sac w<strong>as</strong> then kept<br />

in a beaker with running tap water for 16 h. After dialysis w<strong>as</strong> completed, A<br />

Erlenymeyer fl<strong>as</strong>k (50 ml capacity) w<strong>as</strong> weighed to 0.01 g and the weight recorded. A<br />

dialysis sac w<strong>as</strong> removed from the beaker and held over a funnel directed into a fl<strong>as</strong>k.<br />

The lower end <strong>of</strong> the sac w<strong>as</strong> cut with a blade, ensuring that the flow <strong>of</strong> solution<br />

rele<strong>as</strong>ed w<strong>as</strong> lead into the funnel. The remaining residual liquid w<strong>as</strong> removed from the<br />

tubing using fingers. To obtain the weight <strong>of</strong> the protein solution the fl<strong>as</strong>k w<strong>as</strong><br />

reweighed. Five µl <strong>of</strong> these solutions (protein) were distributed into the wells <strong>of</strong> a 96<br />

well microtitre plate. Then 25 µl <strong>of</strong> re<strong>agent</strong> A (alkaline copper tartrate) were added to<br />

the wells. After that 200 µl <strong>of</strong> re<strong>agent</strong> B (dilute Folin re<strong>agent</strong>) were also added to the<br />

same wells. The plate w<strong>as</strong> then incubated at room temperature for 15 minutes and read<br />

in a microtiter plate reader (DYNEX Revelation 4.21) at 620 nm. A calibration curve<br />

<strong>of</strong> the best fit line w<strong>as</strong> obtained from the standard solutions <strong>of</strong> bovine serum albumin<br />

(Sigma) (Fig. 2.1). The concentration <strong>of</strong> proteins in the <strong>honey</strong> samples w<strong>as</strong><br />

determined from the equation <strong>of</strong> the calibration curve line. Each sample w<strong>as</strong> repeated<br />

in triplicate.<br />

87


Figure 2.1: Calibration curve for protein determination in <strong>honey</strong> samples<br />

2.2.7 Colour:<br />

The colour <strong>of</strong> all <strong>honey</strong> samples w<strong>as</strong> established using the optical density method<br />

recommended by the National Honey Board. The absorbance <strong>of</strong> a 50% (w/v) <strong>honey</strong><br />

solution w<strong>as</strong> determined at 560 nm in a Cecil spectrophotometer. To obtain the actual<br />

absorbance value <strong>of</strong> the undiluted solution, the absorbance w<strong>as</strong> multiplied by 2. Honey<br />

were then categorised according to a Townsend cl<strong>as</strong>sification system (1969).<br />

2.2.8 Pollen analysis:<br />

This method w<strong>as</strong> derived from that <strong>of</strong> Loveaux et al. (1978).<br />

Ten grams <strong>of</strong> <strong>honey</strong> were dissolved in 20 ml <strong>of</strong> deionised water and centrifuged in a<br />

Sanyo-MSE centrifuge at 2500 rpm (3.8 G) for 10 mins. Then the supernatant w<strong>as</strong><br />

decanted and re-suspended in 10 ml <strong>of</strong> deionised water. The solution w<strong>as</strong> re-<br />

centrifuged <strong>as</strong> above and the supernatant w<strong>as</strong> decanted and discarded. The sediment<br />

88


w<strong>as</strong> re-suspended in 1 ml liquefied glycerine-gelatine, transferred to a gl<strong>as</strong>s slide using<br />

a P<strong>as</strong>teur pipette and mounted with a cover slip. Using a light microscope at le<strong>as</strong>t 100<br />

pollen grains in each sample were identified, examined and counted. The samples<br />

were cl<strong>as</strong>sified according to the predominant pollen present in the sample (›45%<br />

pollen grains). Advice about the identity <strong>of</strong> the pollen grains w<strong>as</strong> kindly provided by<br />

National Pollen And Research Unit (NPARU) at Worcester <strong>University</strong>.<br />

2.2.9 Total phenolic content:<br />

This method w<strong>as</strong> used that described by Berreta et al. (2005).<br />

2.2.9.1 Re<strong>agent</strong>/standard preparation:<br />

A 10% (w/v) stock <strong>honey</strong> solution w<strong>as</strong> prepared by weighing 0.5 g <strong>of</strong> each <strong>honey</strong><br />

sample and diluted with 5ml <strong>of</strong> warm (45 o C) deionised water, then mixed for 5 mins.<br />

A Folin-Ciocalteau re<strong>agent</strong> (1:10) w<strong>as</strong> prepared by adding 10 ml <strong>of</strong> Folin re<strong>agent</strong> to<br />

90 ml <strong>of</strong> deionised water.<br />

Gallic acid standards (10-250 µg/ml): A stock solution <strong>of</strong> 0.005 M gallic acid (Sigma)<br />

w<strong>as</strong> prepared by weighing 0.017g <strong>of</strong> gallic acid and made up to 20 ml using<br />

water:methanol solution (1:1).<br />

A range <strong>of</strong> gallic acid standards (10-250 µg/ml) w<strong>as</strong> prepared (Table 2. 10)<br />

89


Table 2.10: Preparation <strong>of</strong> gallic acid standard solutions<br />

Working standard<br />

solution (µg/ml)<br />

Standard stock<br />

solution (µl)<br />

90<br />

Deionised water:<br />

methanol (µl)<br />

0 0 4000<br />

10 47 3953<br />

25 118 3882<br />

50 235 3765<br />

100 470 3530<br />

150 705 3295<br />

200 940 3060<br />

250 1176 2824<br />

Figure 2.2: Standard curve for total phenolic content in <strong>honey</strong> samples<br />

Absorbance at 750nm<br />

0.4<br />

0.35<br />

0.3<br />

0.25<br />

0.2<br />

0.15<br />

0.1<br />

0.05<br />

0<br />

2.2.9.2 Assay method:<br />

Standard curve for total phenolic content<br />

y = 0.0013x<br />

R² = 0.9974<br />

0 50 100 150 200 250 300<br />

Concentration <strong>of</strong> gallic acid standard (ug/ml)<br />

100 µl <strong>of</strong> 10% <strong>of</strong> each <strong>honey</strong> solution w<strong>as</strong> added to 1 ml <strong>of</strong> Folin-phenol re<strong>agent</strong><br />

(1:10) in a test tube. The mixture w<strong>as</strong> mixed for 2 mins and incubated for 20 mins at<br />

room temperature. After incubation, the absorbance w<strong>as</strong> read at 750 nm against<br />

re<strong>agent</strong> blank (100 ml <strong>of</strong> water:methanol) on a Cecil spectrophotometer. The


difference between this absorbance and the sample blank (<strong>honey</strong> solution with<br />

deionised water) w<strong>as</strong> calculated to get the net absorbance. The solutions with gallic<br />

acid in the range <strong>of</strong> 10-250 µg/ml were used for calibration (standard graph). Total<br />

phenolic content w<strong>as</strong> expressed <strong>as</strong> µmol <strong>of</strong> gallic acid eq/kg <strong>honey</strong>. Each <strong>honey</strong><br />

sample w<strong>as</strong> tested in duplicate and the average net absorbance w<strong>as</strong> calculated (A).<br />

Determination for <strong>honey</strong> colour interference with the <strong>as</strong>say:<br />

Two <strong>honey</strong> solutions were made for each sample <strong>as</strong> follows:<br />

A: 1000 µl distilled water: methanol + 100 µl <strong>honey</strong> solution<br />

B: 1100 µl distilled water: methanol<br />

The <strong>honey</strong> colour (H) w<strong>as</strong> calculated <strong>as</strong> the difference in absorbance between A and B<br />

at 750nm. Net absorbance (N) for each sample w<strong>as</strong> calculated <strong>as</strong> follows: N = (A) –<br />

(H), and this value (N) w<strong>as</strong> used to determine phenolic content from the calibration<br />

curve. The standard graph w<strong>as</strong> plotted and phenolic content value for test w<strong>as</strong> obtained<br />

from the standard graph.<br />

2.2.10 Free radical activity <strong>of</strong> <strong>honey</strong>:<br />

This method w<strong>as</strong> established from the method described by Chen et al. (2000) and<br />

Aljadi & Kamaruddish (2004).<br />

2.2.10.1 Re<strong>agent</strong> / Standard Preparations<br />

DPPH (2,2-Di(4-tert-octylphenyl)-1-picryl-hydrazyl) 0.09 mg/ ml (Sigma) w<strong>as</strong><br />

prepared by weighing 0.009 g <strong>of</strong> DPPH and made up to 100 ml with methanol.<br />

Deionised water: methanol (1:1) w<strong>as</strong> prepared by adding 5 ml <strong>of</strong> deionised water to 5<br />

ml <strong>of</strong> methanol.<br />

91


A 10 % (w/v) <strong>honey</strong> solution w<strong>as</strong> made by weighing 5 g <strong>of</strong> <strong>honey</strong> and added to 5 ml<br />

<strong>of</strong> warmed deionised water. The solution then mixed for 5 minutes.<br />

2.2.10.2 Assay procedure:<br />

To <strong>as</strong>say free radical activity 0.75 ml <strong>of</strong> each 10 % (w/v) <strong>honey</strong> solution w<strong>as</strong> mixed<br />

with 1.5 ml <strong>of</strong> 0.09 mg/ml DPPH in methanol in a test tube. Mixture w<strong>as</strong> then<br />

incubated at 25°C for 5 minutes. The absorbance w<strong>as</strong> me<strong>as</strong>ured at 517 nm against a<br />

test blank (<strong>honey</strong> solution + 1.5 <strong>of</strong> deionised water:methanol). The absorbance <strong>of</strong> 1.5<br />

ml <strong>of</strong> DPPH radical control w<strong>as</strong> me<strong>as</strong>ured with 0.75 ml <strong>of</strong> distilled water: methanol<br />

against DPPH radical control blank (2.25 ml <strong>of</strong> distilled water: methanol). The anti<br />

radical activity (ARA) w<strong>as</strong> expressed <strong>as</strong> a percentage inhibition <strong>of</strong> DPPH radical by<br />

<strong>honey</strong> and calculated <strong>as</strong> follows: Anti –radical Activity (ARA%)= {(A-B) / A} x 100<br />

Where A= Average absorbance <strong>of</strong> DPPH radical without <strong>honey</strong> (DPPH control<br />

absorbance) and B = Average absorbance <strong>of</strong> DPPH radical with <strong>honey</strong> (Test<br />

absorbance).<br />

2.3 Determination <strong>of</strong> antibacterial activity <strong>of</strong> <strong>honey</strong><br />

samples against test cultures:<br />

2.3.1 Minimum Inhibitory Concentration (MIC) method:<br />

2.3.1.1 Agar incorporation method:<br />

2.3.1.1.1 Honey selection:<br />

From the bio<strong>as</strong>say results, two <strong>honey</strong>s were selected according to their antibacterial<br />

activity: Manuka <strong>honey</strong> with non peroxide activity equivalent to 18.0% (w/v) phenol<br />

92


and Omani <strong>honey</strong> (B) with hydrogen peroxide activity equivalent to 17.7% (w/v)<br />

phenol<br />

2.3.1.1.2 Inoculum preparation:<br />

One to five colonies <strong>of</strong> test organism were inoculated into 10 ml nutrient broth and<br />

incubated at 37 o C overnight. Subculture w<strong>as</strong> performed for each isolate onto nutrient<br />

agar plates to confirm the purity.<br />

2.3.1.1.3 Plates preparation:<br />

300 ml <strong>of</strong> double strength nutrient agar w<strong>as</strong> prepared by weighing 14.8 g <strong>of</strong> nutrient<br />

agar into 300 ml <strong>of</strong> deionised water. The mixture w<strong>as</strong> mixed and boiled to dissolve the<br />

agar then it w<strong>as</strong> kept in a water bath at 50 o C for 15 minutes. An automatic pipette w<strong>as</strong><br />

used to dispense 10 ml <strong>of</strong> nutrient agar into each <strong>of</strong> 22 universal containers and<br />

autoclaved at 121 o C for 15 minutes. After autoclaving, it w<strong>as</strong> put in water bath at 50 o C<br />

for 20 minutes.<br />

2.3.1.1.4 Honey preparation:<br />

A stock <strong>honey</strong> solution <strong>of</strong> 40% (w/v) w<strong>as</strong> prepared either by adding 20 g <strong>of</strong> manuka<br />

<strong>honey</strong> (A) to a volumetric fl<strong>as</strong>k, and making up to 50 ml with sterile deionised water,<br />

or by adding 12 g <strong>of</strong> <strong>honey</strong> (B) to a volumetric fl<strong>as</strong>k, and making up to 30 ml with<br />

sterile deionised water. Nutrient agar plates containing <strong>honey</strong> varying from 0 -10%<br />

(w/v) were prepared (Table 2.11).<br />

93


Table 2.11: Preparation <strong>of</strong> varying concentration <strong>of</strong> <strong>honey</strong> solution from 40% (w/v)<br />

stock <strong>honey</strong> for MIC method<br />

% <strong>of</strong> <strong>honey</strong><br />

(w/v)<br />

Quantity <strong>of</strong> double<br />

strength N.A (ml)<br />

94<br />

Quantity <strong>of</strong> 40%<br />

stock <strong>honey</strong> (ml)<br />

Quantity <strong>of</strong> sterile<br />

water (ml)<br />

0% 10 0 10.0<br />

1% 10 0.5 9.5<br />

2% 10 1.0 9<br />

3% 10 1.5 8.5<br />

4% 10 2 8<br />

5% 10 2.5 7.5<br />

6% 10 3 7<br />

7% 10 3.5 6.5<br />

8% 10 4 6<br />

9% 10 4.5 5.5<br />

10% 10 5 5<br />

The plates were allowed to set at room temperature for 30 min and then placed in 37 o C<br />

incubator to remove excess moisture. The plates <strong>of</strong> varying <strong>honey</strong> concentration were<br />

then inoculated with 1 µl <strong>of</strong> each test organism (typically 1x10 5 cfu/ml) in triplicate<br />

using multipoint inoculator. The plates were placed in an incubator at 37 o C for 24 h to<br />

allow for growth.<br />

2.3.1.1.5 Plates reading (MIC determination):<br />

After overnight incubation, the growth w<strong>as</strong> recorded for each dilution. Positive growth<br />

w<strong>as</strong> confluent growth within the inoculated area and negative growth w<strong>as</strong> recorded<br />

when no colonies where present. The MIC w<strong>as</strong> deduced <strong>as</strong> the lowest concentration <strong>of</strong><br />

<strong>honey</strong> that inhibited growth, and the average for all <strong>as</strong>says on each test organism w<strong>as</strong><br />

calculated.


2.3.1.2 Broth dilution method:<br />

2.3.1.2.1 Honey selection:<br />

Four <strong>honey</strong>s were selected according to their antibacterial activity: Manuka <strong>honey</strong><br />

with non peroxide activity equivalent to 20.0% (w/v) phenol and Omani <strong>honey</strong>s B, C,<br />

G & F with hydrogen peroxide activity equivalent to 22.85, 21.8, 20.1 and 20.8 %<br />

(w/v) phenol respectively.<br />

2.3.1.2.2 Honey dilution:<br />

40% (w/v) stock solutions <strong>of</strong> <strong>honey</strong> were prepared <strong>as</strong> described above and solutions<br />

ranging from 6% to 15% <strong>honey</strong> diluted with iso-sensitest broth (ISB) were prepared <strong>as</strong><br />

shown in (Table 2.12).<br />

Table 2.12: Preparation <strong>of</strong> tubes for MICs.<br />

Honey Conc. (%w/v) Quantity <strong>of</strong> 40%<br />

<strong>honey</strong> (ml)<br />

95<br />

Quantity <strong>of</strong> ISB<br />

(ml)<br />

Final Volume<br />

(ml)<br />

6 0.225 1.275 1.5<br />

7 0.262 1.237 1.5<br />

8 0.3 1.2 1.5<br />

9 0.337 1.163 1.5<br />

10 0.375 1.125 1.5<br />

11 0.412 1.08 1.5<br />

12 0.45 1.05 1.5<br />

13 0.487 1.012 1.5<br />

14 0.525 0.975 1.5<br />

15 0.562 0.937 1.5<br />

2.3.2.1.3 Microtitre plate inoculation:<br />

One 96 well microtitre plate w<strong>as</strong> used for 3 test organisms and each organism w<strong>as</strong><br />

tested in duplicate. 200 µl <strong>of</strong> each <strong>honey</strong> solution w<strong>as</strong> dispensed to wells in row (A)


for <strong>honey</strong> control, rows (B&C) for isolate 1, rows (D&E) for isolate 2 and rows<br />

(F&G) for isolate 3 in [columns 1-10]. 200 µl <strong>of</strong> ISB w<strong>as</strong> dispensed to wells in rows<br />

(A – G) and columns (11&12) only. 1 µl <strong>of</strong> isolates 1, 2 & 3 w<strong>as</strong> added to wells in<br />

rows B&C, D&E and F&G respectively in columns 1-11. After the wells have been<br />

inoculated the plate w<strong>as</strong> incubated at 37°C for 24h. This procedure w<strong>as</strong> repeated for<br />

all other isolates.<br />

2.3.2.1.4 Visual inspection <strong>of</strong> MIC:<br />

Plates were removed from the incubator and inspected visually for growth. Growth<br />

w<strong>as</strong> <strong>as</strong>sessed by turbidity in the wells and comparing them to the negative control (ISB<br />

only). Wells showing turbidity were considered to contain growth and marked <strong>as</strong><br />

positive. Wells showing no turbidity were <strong>as</strong>sessed to have no growth and marked <strong>as</strong><br />

negative.<br />

2.3.2.1.5 Spectrophotometric determination <strong>of</strong> MIC:<br />

The microtitre plates were placed in a plate reader (DYNEX Revelation 4.21) and<br />

optical density w<strong>as</strong> me<strong>as</strong>ured at 405 nm filter for test and 620 nm filter <strong>as</strong> reference.<br />

The printout <strong>of</strong> the reading were obtained and used to confirm the MIC.<br />

2.3.2 Minimum Bactericidal concentration (MBC) method:<br />

Wells that appeared to have no growth (no turbidity) by visual inspection were<br />

streaked onto nutrient agar plates using sterile 10 µl pl<strong>as</strong>tic loops. The plates were<br />

incubated at 37°C for 24 h. After incubation any growth on the plates w<strong>as</strong> marked <strong>as</strong><br />

positive and no growth marked <strong>as</strong> negative. The plates with the lowest concentration<br />

<strong>of</strong> <strong>honey</strong> showing no growth w<strong>as</strong> recorded <strong>as</strong> the MBC.<br />

96


2.4 Time Kill Curve Assay:<br />

2.4.1 Strain selection:<br />

One culture <strong>of</strong> each <strong>of</strong> the six types <strong>of</strong> bacterial strains w<strong>as</strong> selected according to the<br />

highest MIC and the MIC w<strong>as</strong> approximately doubled <strong>as</strong> shown in (Table 2.13) below:<br />

Table 2.13: Cultures and <strong>honey</strong> concentrations used in the time-kill curves<br />

Strain ID MIC (%w/v) Test conc. used<br />

Acinetobacter 15 9 20<br />

E.coli 7 (2139) 13 30<br />

Klebsiella 5 (3042) 13 30<br />

Enterobacteria 9 (5865) 13 30<br />

Citrobacter 2 (3031) 10 20<br />

Serratia 4 (3036) 15 30<br />

Manuka <strong>honey</strong> (Comvita) UMF 18 equivalent to 18 % (w/v) phenol w<strong>as</strong> used in this<br />

<strong>as</strong>say.<br />

2.4.2 Time-kill curve:<br />

An overnight broth culture <strong>of</strong> each test organism in 10 ml ISB w<strong>as</strong> prepared by<br />

inoculating a colony from a pure culture and incubating at 37 o C for 18 h. After<br />

incubation each culture w<strong>as</strong> diluted with ISB to an optical density <strong>of</strong> 0.5 at 550 nm. 50<br />

ml <strong>of</strong> sterile ISB w<strong>as</strong> added to conical fl<strong>as</strong>k labelled <strong>as</strong> control for each isolate. A 50<br />

ml <strong>of</strong> appropriate <strong>honey</strong> solution (approximately 2xMIC) w<strong>as</strong> prepared in another<br />

conical fl<strong>as</strong>k and labelled <strong>as</strong> test for each isolate. Then 5 ml from the diluted overnight<br />

culture w<strong>as</strong> transferred into each fl<strong>as</strong>k. The mixture w<strong>as</strong> then incubated at 37°C in<br />

shaking water bath. One ml from each fl<strong>as</strong>k were removed into a microcuvette and<br />

absorbance w<strong>as</strong> recorded at 550 nm every 30 mins for the first 2 h and every 1 hr until<br />

4 h had p<strong>as</strong>sed. Finally, a graph <strong>of</strong> optical density versus time w<strong>as</strong> plotted to allow the<br />

97


exponential growth ph<strong>as</strong>e to be identified. Also, a total viable count for each collected<br />

specimen w<strong>as</strong> determined <strong>as</strong> follows:<br />

20 µl <strong>of</strong> the specimen w<strong>as</strong> diluted in a decimal dilution series from 10 -1 to 10 -7 using<br />

maximum recovery diluent (Oxoid). For each dilution 3 x 20 µl aliquots were plated to<br />

the respectively labelled seven segments <strong>of</strong> a nutrient agar plate. Following this, the<br />

plates were left to dry before incubating for 24 h at 37°C. After incubation the colonies<br />

in different sections were counted and recorded and used to calculate the number <strong>of</strong><br />

CFU/ml at each time point. Time-kill curves <strong>of</strong> total viable count versus time were<br />

plotted.<br />

A T-test w<strong>as</strong> applied to establish the statistical difference between untreated cells and<br />

<strong>honey</strong> treated cells in the selected 6 species.<br />

2.5 Effect <strong>of</strong> <strong>honey</strong> on bacterial structure:<br />

2.5.1 Scanning Electron Microscopy (SEM):<br />

SEM is one <strong>of</strong> the multipurpose equipement that is used for the analysis and<br />

examination <strong>of</strong> ultr<strong>as</strong>tructure features <strong>of</strong> solid object. It allows the observer to see<br />

entire specimen and larger part <strong>of</strong> an object (Williams and Carter 2009).<br />

2.5.1.1 Bacterial Selection for SEM<br />

The same test organisms <strong>as</strong> selected for time-kill <strong>as</strong>says were used for morphological<br />

studies.<br />

98


2.5.1.2 Preparation <strong>of</strong> cells in the exponential ph<strong>as</strong>e <strong>of</strong> growth:<br />

1 ml <strong>of</strong> an overnight culture w<strong>as</strong> inoculated into each <strong>of</strong> two fl<strong>as</strong>ks containing 50 ml <strong>of</strong><br />

ISB. The fl<strong>as</strong>ks were labelled <strong>as</strong> either control (no <strong>honey</strong>) or test (with <strong>honey</strong>). The<br />

culture w<strong>as</strong> incubated in a 37 o C shaking water bath for 3 h when it w<strong>as</strong> estimated that<br />

cells would be in the exponential ph<strong>as</strong>e <strong>of</strong> growth. Honey solution (Table 2.13) w<strong>as</strong><br />

then <strong>as</strong>eptically added into the fl<strong>as</strong>k labelled <strong>as</strong> test and the fl<strong>as</strong>ks continued to be<br />

incubated with shaking at 37 o C. The point at which the <strong>honey</strong> w<strong>as</strong> added to the test<br />

fl<strong>as</strong>ks w<strong>as</strong> recorded <strong>as</strong> time 0. Absorbance w<strong>as</strong> me<strong>as</strong>ured every 30 mins till 4 h had<br />

p<strong>as</strong>sed (0, 30, 60, 90, 120, 150 and 180 mins at 550 nm) and 4 ml w<strong>as</strong> removed at each<br />

time point and distributed equally into 4 sterile eppendorff tubes. The tubes were<br />

centrifuged at 13,000 rpm (19.89 G) for 2 mins the supernatant w<strong>as</strong> decanted and the<br />

pellet re-suspended in 250 µl <strong>of</strong> PBS. The contents from all tubes were combined into<br />

1 ml tube, which w<strong>as</strong> then centrifuged. The resulting pellet w<strong>as</strong> re-suspended in 1 ml<br />

<strong>of</strong> PBS and centrifuged again. After removing the supernatant, the pellet w<strong>as</strong> fixed<br />

with 750 µl <strong>of</strong> 3% glutaraldehyde for 1 h at 4 o C. After fixation the cells were w<strong>as</strong>hed<br />

twice (2 x 2 mins) with 1 ml PBS. W<strong>as</strong>hed cells were suspended in PBS were then<br />

stored on a shaker overnight at 4 o C. At this point samples were transferred to Cardiff<br />

<strong>University</strong> for electron microscopy.<br />

2.5.1.3 Preparation <strong>of</strong> cells for scanning electron microscopy:<br />

All samples were centrifuged at 13,000 rpm (19.89 G) for 2 mins. The resulting pellet<br />

w<strong>as</strong> re-suspended in 0.5 ml <strong>of</strong> 1% osmium tetroxide in PBS for 1 h. After the post<br />

fixation, the cells were spun down <strong>as</strong> above and the osmium tetroxide solution w<strong>as</strong><br />

decanted. The cells in pellet were dehydrated in a graded series <strong>of</strong> ethanol<br />

concentrations <strong>as</strong> follows:<br />

99


50%, 70%, 80% and 90% for 5 mins, 100% for 10 mins (twice). The samples were<br />

added to small pl<strong>as</strong>tic containers (labelled by cutting an identification number into it)<br />

that were fitted with 0.22 µm filter membrane attached to blotting paper to trap cells<br />

for critical point drying. A small volume <strong>of</strong> ethanol w<strong>as</strong> added to the containers taking<br />

care not to let the sample completely dry. The dehydrated samples were dried for 16 h<br />

under a high pressure (100 bars) in a critical point dryer (CPD 030; Balzers,<br />

Lerchtenstein) using CO2. The dried samples were then mounted on metal stubs with<br />

carbon – double sided - sticky tape and coated with gold in sputter-coater (EMscope<br />

AE 1232, Kent, UK). Structural observations <strong>of</strong> cells size and shape were performed<br />

using a scanning electron microscopy (JEOL 5600V SEM-UK) operated at 20 kV<br />

accelerating voltage with magnifications ranging from 5000x to 10,000x<br />

magnification. More than 2000 cells for each isolate were observed and examined.<br />

The percentage <strong>of</strong> structural differences between untreated and treated cells w<strong>as</strong><br />

determined using SMile View programme-JEOL (Version 2). The results were<br />

analysed statistically for significance using the Mann-Witney test in Mini Tab<br />

Statistical package (version 15).<br />

2.5.2 Transmission Electron Microscopy (TEM):<br />

TEM allow the observer to see the internal structure <strong>of</strong> an object with high<br />

magnification and greater degree (Williams and Carter 2009).<br />

2.5.2.1 Bacterial Selection for TEM<br />

Two isolates were selected in this method. Acintobacter isolate no. 15 and E. coli<br />

isolate no.7 with 20% (w/v) and 30% (w/v) <strong>of</strong> manuka <strong>honey</strong> respectively.<br />

100


2.5.2.2 Preparation <strong>of</strong> cells for transmission electron microscopy:<br />

Fixed cells with and without <strong>honey</strong> were prepared <strong>as</strong> described for scanning<br />

microscopy in section 2.5.1.2 and 2.5.1.3 up until dehydration w<strong>as</strong> completed. When<br />

ethanol replacement w<strong>as</strong> completed, pellets were rele<strong>as</strong>ed into a fresh 10 ml gl<strong>as</strong>s<br />

container <strong>of</strong> 100% ethanol with a cocktail stick. The pellet w<strong>as</strong> allowed to stand 15<br />

minutes in 100% ethanol and then transferred to fresh 100% ethanol for further 5 min.<br />

The pellet w<strong>as</strong> transferred to 5 ml propylene oxide and left for 15 min. The pellet w<strong>as</strong><br />

transferred to 50:50 propylene oxide/araldite resin (5 g <strong>of</strong> araldite resin and 5 g <strong>of</strong><br />

DDSA were preheated, mixed together and 0.15 g <strong>of</strong> BDMA/propylene oxide about 9<br />

ml per sample and left overnight in the arbitrator. The infiltrated pellet were then<br />

transferred into 5 ml <strong>of</strong> fresh resin (no propylene oxide) and left for 12 hours into the<br />

arbitrator. After incubation, the pallets were transferred into moulds previously filled<br />

with <strong>full</strong> strength resin. The moulds were then incubated in the oven for 3 days at<br />

60°C.<br />

2.5.2.3 Pallet trimming and sectioning:<br />

The samples were trimmed in order to remove the excess resin around the samples and<br />

to expose the pellet <strong>of</strong> cells. Using gl<strong>as</strong>s knives and ultratome III ultra thin sections<br />

were cut and placed onto uncoated 3.00 mm copper grids.<br />

2.5.2.4 Staining <strong>of</strong> thin sections<br />

A paraffin sheet w<strong>as</strong> placed onto either a ceramic tile or Petri dish. A drop <strong>of</strong> 2%<br />

uranyl acetate solution w<strong>as</strong> placed on the paraffin sheet. The grids were floated on the<br />

drop by placing the thin sections side faced down ward. The grids were covered and<br />

stained in a similar manner with Reynolds lead citrate for 5 minutes, then, w<strong>as</strong>hed<br />

twice in distilled water. The grids were dried with filter paper. 12 blocks containing<br />

101


Acinetobacter test & control were prepared (3 blocks at each time point). The same<br />

w<strong>as</strong> done also for E.coli. At le<strong>as</strong>t 8 grids from each sample were stained and<br />

examined. Eleven TEM micrographs were taken for each sample at each time point.<br />

Ten grids were captured by TEM under 16,000 x magnification and one under 32,000<br />

x magnification using (TEM, Philips-PW60100, 10EM 208). A total <strong>of</strong> 400 cells for<br />

each sample were observed and examined for structural differences between <strong>honey</strong><br />

treated cells and untreated cells, and the presence or absence <strong>of</strong> septa w<strong>as</strong> recorded.<br />

2.6 Effect <strong>of</strong> <strong>honey</strong> on bacterial proteins:<br />

The effect <strong>of</strong> exposure to 20% (w/v) manuka <strong>honey</strong> on proteins w<strong>as</strong> tested in selected<br />

test organisms using 2-D gel electrophoresis.<br />

2.6.1 Two Dimensional Gel Electrophoresis:<br />

2.6.1.1 Buffers preparation:<br />

The following re<strong>agent</strong>s were prepared (Table 2.14)<br />

102


Table 2.14: Re<strong>agent</strong>s and buffers used in 2-D Electrophoresis<br />

Re-hydration buffer Per 10 ml<br />

Urea 8 M 4.8 g<br />

CHAPS 2% w/v 0.2 g<br />

DDT 50 mM 0.08 g<br />

Biolyte ampholytes 0.2% w/v 0.05 ml<br />

Equilibration buffer 1 Per 10 ml<br />

Urea 6 M 3.6 g<br />

Tris-HCl, pH 8.8, 0.375M 0.592 g<br />

SDS 2% 0.2 g<br />

Glycerol 20% 2 ml<br />

DTT 2% w/v 0.2 g<br />

Equilibration buffer 2 Per 10 ml<br />

Urea 6 M 3.6 g<br />

Tris-HCl, pH 8.8, 0.375M 0.592 g<br />

SDS 2% 0.2 g<br />

Glycerol 20% 2 ml<br />

Iodoacetamide 2.5% w/v 0.252 g<br />

Overlay Agarose<br />

0.04 g Bromophenol blue 100 ml water<br />

1:20 <strong>of</strong> BPB in MOPS Add 0.12 ml <strong>of</strong> agarose<br />

MOPS running buffer Per 1000 ml<br />

MOPS 10.45 g<br />

Tris b<strong>as</strong>e 6.06 g<br />

SDS 1.00 g<br />

EDTA 0.30 g<br />

NaCl Per 100 ml<br />

20% 20 g<br />

103


2.6.1.2 Cell preparation:<br />

10 ml <strong>of</strong> TSB (Oxoid) w<strong>as</strong> inoculated with 2-4 pure colonies <strong>of</strong> Acinetobacter and<br />

incubated at 37 o C for 18 h. 2 ml <strong>of</strong> overnight culture w<strong>as</strong> added to 50 ml <strong>of</strong> ISB in a<br />

500 ml fl<strong>as</strong>k and 2 ml w<strong>as</strong> used to inoculate a second fl<strong>as</strong>k containing 50 ml ISB with<br />

20% (w/v) manuka <strong>honey</strong>. The cultures were incubated at 37 o C with shaking (100 rpm<br />

for 24 h). After incubation, either 50 ml <strong>of</strong> TSB or 50 ml containing 20% manuka<br />

<strong>honey</strong> w<strong>as</strong> added to each respective culture and incubated for a further 3 h with<br />

shaking (100 rpm) to ensure that the cells were in the exponential ph<strong>as</strong>e <strong>of</strong> growth.<br />

Cells were harvested by centrifugation in a Sorvell centrifuge (10,000 rpm [15.3 G]<br />

for 6 min) and the cells were re-suspended in 10 ml deionised water, the OD <strong>of</strong> the<br />

resulting supernatant w<strong>as</strong> recorded at 550 nm using water <strong>as</strong> a blank. The proteins<br />

were extracted by sonicating (Jencons, Sonics VCX 500) with 500 watts power, 20<br />

kHz frequency and 30% amplitude for 2.5 min. In every 30 seconds each sample w<strong>as</strong><br />

tacked <strong>of</strong>f to Eppendorff tube and kept on ice to cool and reduce air bubbles. The cells<br />

were spun down in a micr<strong>of</strong>uge on top speed (13,000 G for 4 mins). Then, the<br />

supernatant w<strong>as</strong> removed into sterile 1.5 ml Eppendorf tubes and stored at -20 o C. A<br />

protein determination w<strong>as</strong> performed for each extract <strong>as</strong> described above.<br />

2.6.1.3 Protein determination <strong>of</strong> Acinetobacter extracts for each extract:<br />

Five µl <strong>of</strong> each sample to be analysed w<strong>as</strong> dispensed into wells <strong>of</strong> a 96 well microtitre<br />

plate, then 25 µl <strong>of</strong> re<strong>agent</strong> A (alkaline copper tartrate) were added followed by 200 µl<br />

<strong>of</strong> re<strong>agent</strong> B (dilute Folin re<strong>agent</strong>). The plate w<strong>as</strong> incubated at room temperature for<br />

15 minutes and then read in a microtiter plate reader (DYNEX Revelation 4.21) at 620<br />

nm. Suitable standard solutions (low standard 3mg/ml) <strong>of</strong> bovine serum albumin<br />

(Sigma) were used and calibration curve obtained, to which a best-fit line w<strong>as</strong> added<br />

and the equation <strong>of</strong> this line w<strong>as</strong> used to obtain the concentration <strong>of</strong> proteins in the<br />

104


samples (Fig 2.3). Each sample w<strong>as</strong> run in triplicate. The results from the protein<br />

<strong>as</strong>say (Table 2.15) were used to calculate the value needed to give 150 µg <strong>of</strong> protein in<br />

each sample for 2-D gel electrophoresis.<br />

Table 2.15: Quantity <strong>of</strong> proteins in Acinetobacter cells with and without 20% <strong>honey</strong><br />

Protein sample <strong>of</strong><br />

Acinetobacter<br />

Protein conc.<br />

(mg/ml)<br />

105<br />

Volume <strong>of</strong><br />

protein<br />

sample µl<br />

Volume <strong>of</strong> re-<br />

hydration buffer<br />

µl<br />

Control cells (no <strong>honey</strong>) 4.55 32 168<br />

Test cells (20% <strong>honey</strong>) 4.2 35.7 164.3<br />

Figure 2.3: Calibration curve <strong>of</strong> protein concentration in Acinetobacter with and<br />

without 20% manuka <strong>honey</strong>.


2.6.1.4 Rehydration and sample application:<br />

The rehydration tray w<strong>as</strong> prepared and protein samples were pipetted <strong>as</strong> a line along<br />

the edge <strong>of</strong> one <strong>of</strong> the rehydration channels, leaving 1 cm free at each end <strong>of</strong> the<br />

channel and ensuring that there were no bubbles. The cover sheet w<strong>as</strong> peeled from the<br />

IPG strip using forceps. The strips were placed gel side down onto samples and ensure<br />

„‟+‟‟ and pH range are clear and legible with no bubbles. The lid w<strong>as</strong> placed back onto<br />

the rehydration tray. Then the samples were kept for 60 min to be <strong>full</strong>y absorbed.<br />

Mineral oil w<strong>as</strong> dripped care<strong>full</strong>y over the pl<strong>as</strong>tic back <strong>of</strong> the IPG strip to <strong>full</strong>y cover<br />

the strip. The rehydration tray w<strong>as</strong> covered with the lid and left for 11-16 h (overnight)<br />

to allow rehydration/loading to be <strong>full</strong>y completed.<br />

2.6.1.5 Rehydration in the PROTEAN ® IEF Focusing Tray:<br />

A paper wick w<strong>as</strong> placed over each <strong>of</strong> the electrodes using forceps, and 10 µl <strong>of</strong><br />

deionised water (18 megohoms-cm) w<strong>as</strong> pipetted on each wick. Using forceps, the<br />

IPG strip w<strong>as</strong> removed from the rehydration channel and held vertically for 10<br />

seconds to drain. The lower end <strong>of</strong> the strip w<strong>as</strong> touched onto blotting paper. The IPG<br />

strip w<strong>as</strong> transferred to the prepared channel ensuring the gel w<strong>as</strong> facing down and the<br />

„‟+‟‟ sign is at the positive end <strong>of</strong> the tray. The IPG strip w<strong>as</strong> covered with 2 ml <strong>of</strong><br />

fresh mineral oil ensuring there are no bubbles. The lid w<strong>as</strong> put on the tray and the tray<br />

w<strong>as</strong> put in an iso-electric focusing cell. The IEF cell w<strong>as</strong> programmed <strong>as</strong> per sheet<br />

provided (35000 volt for 24 h); this w<strong>as</strong> suitable for 11 cm pH 3-10. After completion,<br />

the IPG strip w<strong>as</strong> removed with forceps, and it w<strong>as</strong> held vertically for 10 seconds to<br />

drain, the lower end w<strong>as</strong> touched onto blotting paper to remove the excess <strong>of</strong> mineral<br />

oil. To ensure that all the oil w<strong>as</strong> removed, the IPG strip w<strong>as</strong> placed on dry filter paper<br />

gel side up and plotted gently with wet filter paper. To store the gels at this point, the<br />

IPG strip w<strong>as</strong> transferred to a clean rehydration/equilibration tray (gel side up). The<br />

106


tray w<strong>as</strong> covered and wrapped in saran wrap and placed in a -70 o C freezer. The gel<br />

w<strong>as</strong> defrosted for not more than 15 min before SDS-PAGE separation, <strong>as</strong> proteins will<br />

migrate.<br />

2.6.1.6 Equilibration and SDS-PAGE:<br />

3 ml <strong>of</strong> equilibration buffer 1 (Table 2.14) w<strong>as</strong> transferred to a lane in the rehydration<br />

tray then the IPG strip w<strong>as</strong> slid into the buffer (gel side up), ensuring there were no<br />

bubbles. The lid w<strong>as</strong> placed on the rehydration tray and put on an orbital shaker and<br />

shacked gently for 30 min at room temperature. The used buffer 1 w<strong>as</strong> decanted from<br />

the squared end <strong>of</strong> the tray until the tray w<strong>as</strong> vertical. 3 ml <strong>of</strong> equilibration buffer 2<br />

w<strong>as</strong> added to the strip and placed on an orbital shaker and shaken gently for 30 min at<br />

room temperature. The used buffer 2 w<strong>as</strong> decanted from the squared end <strong>of</strong> the tray<br />

until the tray w<strong>as</strong> vertical. The overlay agarose w<strong>as</strong> made <strong>as</strong> mentioned above and<br />

melted by gentle heating. A me<strong>as</strong>uring cylinder w<strong>as</strong> filled with MOPS buffer. Any<br />

excess water from the SDS page gel w<strong>as</strong> removed by pipetting and blotting gently.<br />

The IPG strip w<strong>as</strong> removed from the rehydration tray and dipped briefly into the<br />

cylinder <strong>of</strong> running buffer. The IPG strip w<strong>as</strong> placed gel facing forwarded onto the<br />

back plate <strong>of</strong> the 2-D gel and pushed down care<strong>full</strong>y with forceps so that it made<br />

contact with the gel. The IPG strip w<strong>as</strong> covered with overlay agarose and allowed<br />

setting for 10 mins. The gel w<strong>as</strong> mounted in the electrophoresis criterion cell, and the<br />

reservoirs were filled with 60 ml MOPS buffer and allowed for 55 min for the run to<br />

complete (200 volts).<br />

2.6.1.7 Staining and gel visualising:<br />

The gel w<strong>as</strong> placed in 100 ml ultrapure water and microwaved on <strong>full</strong> power for 1<br />

min. The gel w<strong>as</strong> shaken in an orbital shaker for 1 min, and then the water w<strong>as</strong><br />

discarded. The l<strong>as</strong>t two steps were repeated twice. 20-30 ml <strong>of</strong> simplyBlue safe stain<br />

107


w<strong>as</strong> added to the gel and microwaved on high power for 45 seconds to 1 min. The gel<br />

w<strong>as</strong> placed on an orbital shaker and shaken gently for 5-10 mins before discarding the<br />

stain. 100 ml <strong>of</strong> ultrapure water w<strong>as</strong> added to the gel and placed on an orbital shaker<br />

for 10 min, the water w<strong>as</strong> discarded after use. 20 ml <strong>of</strong> 20% NaCl w<strong>as</strong> added and the<br />

gel w<strong>as</strong> placed back onto the orbital shaker for 5-10 mins. After staining, the gel w<strong>as</strong><br />

visualised using the UVP AutoChemi, gel system and analysed using PDQuest B<strong>as</strong>ic<br />

8.0 s<strong>of</strong>tware.<br />

2.7 Statistical analysis <strong>of</strong> the data:<br />

In time-kill <strong>as</strong>say the statistical difference in cell count <strong>of</strong> two variables (<strong>honey</strong><br />

treatment and non <strong>honey</strong> treatment) in term <strong>of</strong> two factors (time and isolates) w<strong>as</strong><br />

applied using T-test.<br />

In SEM the percentage <strong>of</strong> structural differences (length <strong>of</strong> cells) between untreated and<br />

treated cells w<strong>as</strong> determined using SMile View programme-JEOL (Version 2). The<br />

results were analysed statistically for significance using the Mann-Witney test in Mini<br />

Tab Statistical package (version 15).<br />

108


Chapter 3<br />

Results<br />

109


3.1 Confirmations <strong>of</strong> the identity and antibiotics sensitivities<br />

<strong>of</strong> test organisms:<br />

Before the effects <strong>of</strong> <strong>honey</strong> on test organisms were investigated, the identity <strong>of</strong> each<br />

isolate w<strong>as</strong> checked using BBL kits, and antibiotic susceptibilities were confirmed by<br />

antibiotic disc sensitivity tests. Results for Acinetobacter, Escherichia coli, Klebsiella,<br />

Citrobacter, Enterobacter and Serratia are presented in Tables 3.1, 3.2, 3.2, 3.4, 3.5<br />

and 3.6 respectively.<br />

Identification and antibiotics sensitivities <strong>of</strong> thirty multi-drug resistant isolates <strong>of</strong><br />

Acinetobacter baumannii and Acinetobacter baumannii/calcoaceticus complex<br />

provided from <strong>University</strong> <strong>of</strong> Wales Hospital (Table 3.1a & 3.1b)<br />

Identification and antibiotics sensitivity tests for the fifty six clinical isolates <strong>of</strong><br />

extended spectrum beta-lactam<strong>as</strong>es (ESBL) <strong>of</strong> Enterobacteriaceae with resistant to<br />

third generation cephalosporins (3 rd G cephalosporin) that were provided from<br />

<strong>University</strong> <strong>of</strong> Wales Hospital did confirm that all cultures conformed to their <strong>as</strong>signed<br />

labels (Table 3.2 to 3.6)<br />

110


Table 3.1a: Identification and antibiotics sensitivities <strong>of</strong> 15 MDR Acinetobacter<br />

isolates<br />

Identification <strong>of</strong> isolates Resistance Pattern<br />

1. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

2. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

111<br />

CAZ, CTX, CE, CXM, CIP, AMP<br />

CAZ, CTX, CXM, FOX, ERT, TAZ,AZT,<br />

AMP<br />

3. Acinetobacter baumannii CAZ, CTX, CXM, FOX, ERT,AZT, AMP<br />

4. Acinetobacter baumannii CAZ, CTX, AK, CN, FOX, CXM, , AMP<br />

5. Acinetobacter baumannii CAZ, CTX, FOX, CXM, ERT, AMP<br />

6. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

7. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

8. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

9. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

10. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

CAZ, CTX, FOX, CN, TAZ, ERT, CIP<br />

CAZ, CTX, CN, FOX, TAZ, ERT, CIP,<br />

AMP<br />

CAZ, CTX, FOX, CXM, ERT, AZT, AMP<br />

CAZ, CTX, FOX, TAZ, ERT, AZT, AMP<br />

CAZ, CTX, CN, FOX, TAZ,ERT, CIP, AZT<br />

11. Acinetobacter baumannii CAZ, CTX, CN, FOX, TAZ, ERT, CIP, SXT<br />

12. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

CAZ, CTX, CN, FOX, TAZ, ERT, CIP, SXT<br />

13. Acinetobacter baumannii CAZ, CTX, CN, FOX, TAZ, ERT, CIP, SXT<br />

14. Acinetobacter baumannii CAZ, CTX, CN, FOX, TAZ, ERT, CIP, AZT<br />

15. Acinetobacter baumannii CAZ, CTX, FOX, TAZ, ERT, CIP, AZT, CN


Table 3.1b: Identification and antibiotics sensitivities <strong>of</strong> 15 MDR Acinetobacter<br />

isolates<br />

Identification <strong>of</strong> isolates Resistance Pattern<br />

16. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

17. Acinetobacter baumannii<br />

/calcoaceticus complex<br />

CAZ, CTX, CXM, FOX, TAZ, ERT, CIP, SXT, CN<br />

CAZ, CTX, CN, FOX, TAZ, ERT, CIP, SXT<br />

18. Acinetobacter baumannii CAZ, CTX, FOX, TAZ, ERT, CN,CIP, SXT, AZT<br />

19. Acinetobacter baumannii CAZ, CTX, FOX, CXM, ERT, AZT, AMP<br />

20. Acinetobacter baumannii CAZ, CTX, FOX, CXM, AZT, ERT, AMP, SXT<br />

21. Acinetobacter baumannii CAZ, CTX, FOX, CXM, AZT, ERT, AMP, SXT<br />

22. Acinetobacter baumannii CAZ, CTX, CRO, CE, CXM, AK, CN,CIP, AMP<br />

23. Acinetobacter baumannii CAZ, CTX, CRO, CE, CXM, AK, CN,CIP, AMP<br />

24. Acinetobacter baumannii CAZ, CTX, CRO, CE, CXM, AK, CN,CIP, AMP<br />

25. Acinetobacter baumannii CAZ, CTX, CRO, CE, CXM, AK, CN,CIP, AMP<br />

26. Acinetobacter baumannii CAZ, CTX, CRO, CE, CXM, AK, CN,CIP, AMP<br />

27. Acinetobacter baumannii CAZ, CTX, CRO, CE, CXM, AK, CN,CIP, AMP<br />

28. Acinetobacter baumannii CAZ, CTX, CRO, CE, CXM, AK, CN,CIP, AMP<br />

29. Acinetobacter baumannii CAZ, CTX, CRO, CE, CXM, AK, CN,CIP, AMP<br />

30. Acinetobacter baumannii CAZ, CTX, CRO, CE, CXM, AK, CN,CIP, AMP<br />

cefotaxime (CTX 30µg), ceftazidime (CAZ 30µg), cefoxitin (FOX 30µg), cefuroxime<br />

(CXM 30µg), gentamicin (CN 10µg), cipr<strong>of</strong>loxacin (CIP 5µg), ampicillin (AMP<br />

10µg), ertapenem (ERT 10µg), aztreonam (AZT 10µg), tazobactam (TAZ 110µg),<br />

septrin (SXT 25µg), amikacin (AK 30µg). All isolates were sensitive to imipenem<br />

(IMP 10µg), meropenem (MEM 10µg) & colistin (CT10µg).<br />

112


Table 3.2: Confirmation <strong>of</strong> identity and antibiotics sensitivity including ESBL tests<br />

for 10 E.coli isolates.<br />

Isolate<br />

No.<br />

Identification <strong>of</strong><br />

isolates<br />

Resistance Pattern ESBL test ESBL/<br />

AmpC * test<br />

CAZ CTX CPM TZP<br />

1. Escherichia coli R R R S ESBL (+) -<br />

2. Escherichia coli R R R S ESBL (+) -<br />

3. Escherichia coli R R S S ESBL (+) -<br />

4. Escherichia coli R R R R ESBL (+) -<br />

5. Escherichia coli R R R R ESBL (+) ESBL<br />

6. Escherichia coli R R S S ESBL (+) AmpC<br />

7. Escherichia coli R R S R ESBL (+) ESBL<br />

8. Escherichia coli R R S R ESBL (+) -<br />

9. Escherichia coli R R S S ESBL (+) ESBL<br />

10. Escherichia coli R R S S ESBL (+) ESBL<br />

R- Resistant, S - Sensitive<br />

All cephalosporin tested (CAZ, CTX & CPM) should be reported resistant if ESBL<br />

test positive.<br />

113


Table 3.3: Confirmation <strong>of</strong> identity and antibiotics sensitivity including ESBL tests<br />

for 11 Klebsiella isolates<br />

Isolate<br />

No.<br />

Identification <strong>of</strong><br />

isolates<br />

Resistance Pattern ESBL<br />

test<br />

CAZ CTX CPM TZP<br />

1. Klebsiella pneumoniae R R S S ESBL (+) -<br />

2. Klebsiella pneumoniae R R S S ESBL (+) -<br />

114<br />

ESBL<br />

/<br />

Amp<br />

C *<br />

test<br />

3. Klebsiella pneumoniae R R R R ESBL (+) ESBL<br />

4. Klebsiella pneumoniae R R R R ESBL (+) -<br />

5. Klebsiella oxytoca R R R R ESBL (+) -<br />

6. Klebsiella pneumoniae R R S S ESBL (+) -<br />

7. Klebsiella pneumoniae R R S S ESBL (+) ESBL<br />

8. Klebsiella oxytoca R R S R ESBL (+) -<br />

9. Klebsiella pneumoniae R R S R ESBL (+) ESBL<br />

10. Klebsiella oxytoca R R S R ESBL (+) -<br />

11. Klebsiella pneumoniae R R R S ESBL (+) -<br />

R- Resistant, S - Sensitive<br />

All cephalosporin tested (CAZ, CTX & CPM) should be reported resistant if ESBL<br />

test positive.


Table 3.4: Confirmation <strong>of</strong> identity and antibiotics sensitivity including ESBL tests<br />

for 12 Citrobacter isolates.<br />

Identification <strong>of</strong><br />

isolates<br />

Resistance Pattern ESBL<br />

CAZ CTX CPM TZP<br />

test<br />

115<br />

ESBL/<br />

AmpC *<br />

test<br />

1. Citrobacter farmeri R R S R ESBL (+) ESBL<br />

2. Citrobacter farmeri R R S S ESBL (-) AmpC<br />

3. Citrobacter farmeri R S S S ESBL (-) AmpC<br />

4. Citrobacter farmeri R R S S ESBL (-) AmpC<br />

5. Citrobacter farmeri R R S R ESBL (-) AmpC<br />

6. Citrobacter farmeri R R S R ESBL (-) AmpC<br />

7. Citrobacter farmeri R R S R ESBL (-) AmpC<br />

8. Citrobacter farmeri R R S S ESBL (-) AmpC<br />

9. Citrobacter farmeri R R S S ESBL (-) AmpC<br />

10. Citrobacter farmeri R R S R ESBL (-) AmpC<br />

11. Citrobacter farmeri R R S S ESBL (-) AmpC<br />

12. Citrobacter farmeri R R S R ESBL (-) AmpC<br />

R- Resistant, S - Sensitive<br />

All cephalosporin tested (CAZ, CTX & CPM) should be reported resistant if ESBL<br />

test positive.


Table 3.5: Confirmation <strong>of</strong> identity and antibiotics sensitivity including ESBL tests<br />

for 15 Enterobacter isolates.<br />

Identification <strong>of</strong><br />

isolates<br />

Resistance Pattern ESBL<br />

test<br />

CAZ CTX CPM TZP<br />

116<br />

ESBL/<br />

AmpC *<br />

test<br />

1. Enterobacter cloacae R R S R ESBL (+) -<br />

2. Enterobacter cloacae R R S R ESBL (+) ESBL<br />

3. Enterobacter cloacae R R S R ESBL (+) -<br />

4. Enterobacter cloacae R R S R ESBL (+) -<br />

5. Enterobacter hormaechi R R S R ESBL (+) -<br />

6. Enterobacter cloacae R R S R ESBL (+) -<br />

7. Enterobacter cloacae R R S R ESBL (+) ESBL<br />

&<br />

AmpC<br />

8. Enterobacter aerogenes R R S R ESBL (+) -<br />

9. Enterobacter cloacae R R S R ESBL (+) AmpC<br />

10. Enterobacter cloacae R R S R ESBL (-) AmpC<br />

11. Enterobacter cloacae R R S R ESBL (-) AmpC<br />

12. Enterobacter cloacae R R S R ESBL (-) AmpC<br />

13. Enterobacter cloacae R R S R ESBL (+) AmpC<br />

14. Enterobacter cloacae R R S R ESBL (+) AmpC<br />

15. Enterobacter cloacae R R S R ESBL (+) AmpC<br />

R- Resistant, S – Sensitive<br />

All cephalosporin tested (CAZ, CTX & CPM) should be reported resistant if ESBL test<br />

positive.


Table 3.6: Confirmation <strong>of</strong> identity and antibiotics sensitivity including ESBL tests<br />

for 8 Serratia isolates.<br />

Identification <strong>of</strong><br />

isolates<br />

Resistance Pattern ESBL<br />

test<br />

CAZ CTX CPM TZP<br />

117<br />

ESBL/<br />

AmpC * test<br />

1. Serratia plymuthica R R S R ESBL (+) ESBL<br />

2. Serratia marcescens R R S R ESBL (-) AmpC<br />

3. Serratia marcescens R R S R ESBL (-) AmpC<br />

4. Serratia marcescens R R S R ESBL (-) AmpC<br />

5. Serratia marcescens R R S R ESBL (-) AmpC<br />

6. Serratia marcescens R R S R ESBL (-) AmpC<br />

7. Serratia marcescens R R S R ESBL (+) AmpC<br />

8. Serratia marcescens R R S R ESBL (-) AmpC<br />

R- Resistant, S – Sensitive<br />

All cephalosporin tested (CAZ, CTX & CPM) should be reported resistant if ESBL test<br />

positive.<br />

ESBL/AmpC * confirmation test w<strong>as</strong> used for the negative ESBL double-disk test.<br />

cefotaxime (CTX 30µg), ceftazidime (CAZ 30µg), cefepem (FEP 30µg).<br />

All isolates (MDR and ESBLs) were sensitive to imipenem (IMP 10µg) and<br />

meropenem (MEM 10µg). The identity <strong>of</strong> these isolates w<strong>as</strong> confirmed using API 20E<br />

identification kit.


3.2 Characterization <strong>of</strong> <strong>honey</strong> samples:<br />

3.2.1 Determination <strong>of</strong> antibacterial activity:<br />

The antibacterial activity <strong>of</strong> <strong>honey</strong> samples w<strong>as</strong> determined by the method <strong>of</strong> Allen et<br />

al (1991). Essentially in this bio<strong>as</strong>say a reference strain <strong>of</strong> Staphylococcus aureus<br />

(NCTC 6571) w<strong>as</strong> seeded into agar, and the extent <strong>of</strong> inhibition induced by <strong>honey</strong><br />

samples were compared to that induced by phenol. An example <strong>of</strong> an incubated plate<br />

is given in Fig 3.1.<br />

Figure 3.1: A typical <strong>honey</strong> bio<strong>as</strong>say plate.<br />

118<br />

Zones <strong>of</strong> inhibition


The diameter <strong>of</strong> the zones <strong>of</strong> inhibition <strong>of</strong> the phenol standards were me<strong>as</strong>ured<br />

(horizontal and vertical) and plotted against the phenol concentration. A linear<br />

relationship between <strong>honey</strong> concentration and the diameter <strong>of</strong> the zone <strong>of</strong> inhibition<br />

w<strong>as</strong> determined and the equation (Fig 3.2) w<strong>as</strong> used to calculate the potency relating<br />

to phenol <strong>of</strong> the <strong>honey</strong> samples. Three bio<strong>as</strong>say plates were used in total, with the<br />

manuka <strong>honey</strong> sample included on each plate for consistency. Each Omani <strong>honey</strong><br />

sample w<strong>as</strong> tested in quadruplicate on a plate. Good agreement w<strong>as</strong> found between the<br />

three bio<strong>as</strong>say plates (R 2 =0.9482, 0.9657 & 0.9743 respectively)<br />

Honey samples were diluted with and without catal<strong>as</strong>e to determine whether the zones<br />

<strong>of</strong> inhibition were due to the generation <strong>of</strong> hydrogen peroxide or not. Hence, total<br />

activity w<strong>as</strong> determined when the <strong>honey</strong> sample w<strong>as</strong> diluted in deionised water, and<br />

non-peroxide activity w<strong>as</strong> calculated when the <strong>honey</strong> w<strong>as</strong> diluted with catal<strong>as</strong>e (Table<br />

3.7).<br />

119


Figure 3.2: A typical calibration curve <strong>of</strong> the bio<strong>as</strong>say.<br />

Mean Diameter (mm)<br />

The data obtained from this method w<strong>as</strong> repeated twice<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Phenol Standard Curve<br />

120<br />

y = 2.1643x + 5.3857<br />

R² = 0.9719<br />

0 1 2 3 4 5 6 7 8<br />

% (W/V) Phenol


Table 3.7: Antibacterial activity <strong>of</strong> <strong>honey</strong> samples<br />

Type <strong>of</strong> <strong>honey</strong> Total activity phenol<br />

equivalent %w/v<br />

121<br />

Non- peroxide activity<br />

phenol equivalent %<br />

w/v<br />

Manuka <strong>honey</strong>-(plate1) 20.4 20.6<br />

Manuka <strong>honey</strong>-(plate2) 22.1 20.2<br />

Manuka <strong>honey</strong>-(plate3) 21.3 19.3<br />

Mean ± SD(n) 21.3± 0.9 (3)<br />

OH-B (plate1) 21.5<br />

OH-B (plate2) 24.2<br />

Mean ± SD(n)<br />

22.9± 1.9 (2)<br />

OH-C 21.8<br />

20.0± 0.8 (3)<br />

3.7<br />


w<strong>as</strong> tested on 2 plates. Omani <strong>honey</strong>s B & C demonstrated the highest total activity<br />

<strong>of</strong> (22.9% & 21.8% (w/v) phenol equivalent) respectively. Omani <strong>honey</strong>s (F & G)<br />

have almost same activity. There w<strong>as</strong> no activity detected in Omani <strong>honey</strong> I.<br />

3.2.2 Chemical & physical analysis <strong>of</strong> <strong>honey</strong> samples<br />

As well <strong>as</strong> antibacterial activity, <strong>honey</strong>s were analysed for their chemical and<br />

physical characteristics. All Omani <strong>honey</strong> samples were tested for their pH, water,<br />

sugar content, protein content, HMF level, colour, pollen and anti-oxidant activity<br />

these were compred to reference manuka <strong>honey</strong> (Tables 3.8, 3.9 & 3.10). pH w<strong>as</strong><br />

tested in triplicate using 50% (w/v) <strong>honey</strong> solutions and the mean w<strong>as</strong> recorded. The<br />

pH w<strong>as</strong> ranged between 3.5 to 6.27 with mean <strong>of</strong> 4.76 ±0.93. This indicates that all<br />

<strong>honey</strong>s fall within acidic range that able to inhibits the growth <strong>of</strong> most micro-<br />

organisms. Sugar and water content were tested using refractometer. Mean sugar w<strong>as</strong><br />

81%±2.86 with range between 75% and 85%. While mean water content w<strong>as</strong> 15.8%<br />

± 1.9 with range between 12.5% and 19.5%. Again low water and high sugar make<br />

the survival <strong>of</strong> bacteria are imposible. HMF test w<strong>as</strong> used to determine the quality<br />

and history <strong>of</strong> <strong>honey</strong> or if it w<strong>as</strong> subjected to heat treatment while processing (Sanz<br />

et al., 2003). An elevated level <strong>of</strong> HMF w<strong>as</strong> expected in the Omani <strong>honey</strong>s due to<br />

the hot climate in Oman; nevertheless HMF level w<strong>as</strong> not unduly elevated (Table<br />

3.8). The mean range w<strong>as</strong> 11.1±11.9 with a range between 2.9 and 38.5 mg/kg.<br />

Conversely the mean protein level w<strong>as</strong> quite high (4.7±1.5 mg/g) (Table 3.8)<br />

compared to values previously established (White & Rudyj 1978). The colour <strong>of</strong><br />

each <strong>honey</strong> sample w<strong>as</strong> primarily visually inspected and then the optical density <strong>of</strong><br />

50% (w/v) from each sample w<strong>as</strong> read at 560 nm and worked out according to the<br />

Townsend‟s cl<strong>as</strong>sification (Townsend, 1969). The results were completely matched<br />

122


with visual estimation. The colour <strong>of</strong> Omani <strong>honey</strong>s w<strong>as</strong> ranged between extra light<br />

amber and dark amber (Table 3.8). The darker <strong>honey</strong> the highest antibacterial<br />

activity presents (Molan 1992). All parameters indicated that all <strong>honey</strong>s fell within<br />

the ranges normally expected for pH, sugar, water content, protein and HMF from<br />

National Honey Broad (NHB) (Table 3.8).<br />

Table 3.8: Chemical & physical analysis <strong>of</strong> different types <strong>of</strong> Omani <strong>honey</strong><br />

compared to manuka <strong>honey</strong><br />

Honey<br />

Sample<br />

pH Water<br />

content<br />

%(w/v)<br />

Sugar<br />

content<br />

% (w/v)<br />

123<br />

Protein<br />

(mg/g)<br />

HMF<br />

(mg/kg)<br />

Colour<br />

OH-B 4.6 15.1 82 5.8 5.7 Amber<br />

OH-C 4.3 16 81 5.2 3.5 Amber<br />

OH-D 4.7 15.1 82.5 2.6 7.1 Light amber<br />

OH-E 3.9 15.3 82 6.2 2.9 Dark amber<br />

OH-F 4.6 12.5 85 6.3 10.4 Dark amber<br />

OH-G 6.2 17 80.5 2.6 38.5 Extra light<br />

amber<br />

OH-H 3.5 15.8 80 5.8 4.2 Dark amber<br />

OH-I 5.9 19.5 75 3.5 16.7 Extra light<br />

amber<br />

Range 3.5-6.2 12.5-<br />

19.5<br />

Mean ±<br />

SD (n)<br />

Manuka<br />

<strong>honey</strong><br />

4.7±<br />

0.9(8)<br />

15.8±<br />

1.9(8)<br />

75-85 2.6-6.3 2.9-38.5 Extra light<br />

amber to<br />

Dark amber<br />

81.0±<br />

2.9(8)<br />

4.7±<br />

1.6(8)<br />

11.1±<br />

11.9(8)<br />

3.5 20 78 10 3 Light amber<br />

-


3.2.2.1 Pollen analysis<br />

Floral sources reported by beekeepers did not completely match to confirmed<br />

identities provided by the National Pollen And Research Unit (NPARU) at Worcester<br />

university (Table 3.9). Images from each <strong>honey</strong> samples were taken and the grains<br />

were cl<strong>as</strong>sified according to the predominant pollen (›45% pollen grains) (Fig 3.3)<br />

Table 3.9: Represent the identification <strong>of</strong> flora sources by pollen analysis<br />

Honey<br />

Sample<br />

Beekeepers<br />

Identification<br />

National Pollen And Research Unit (NPARU)<br />

124<br />

Identification<br />

OH-B Multi-flora Graminae (the gr<strong>as</strong>ses), Acacia (thorn trees, part <strong>of</strong><br />

mimosoideae family) Asteraceae (daisy/sunflower<br />

family), Balsam (scented trees and shrubs), Tilia (lime)<br />

OH-C Honey dew Calendula arvensis (marigold), Graminae, Mimosa<br />

(subfamily <strong>of</strong> legume family), Myrtacae Eucalpyus,<br />

rosaceae (rose) Graminae w<strong>as</strong> very high<br />

OH-D Honey dew little pollen w<strong>as</strong> found on the slide<br />

OH-E Honey dew Myrtacae eucalyptus, Asteraceae inc. Bellis (daisy) and<br />

Ambrosia (ragweed) Fabaceae legume family) – specific<br />

to this slide Br<strong>as</strong>sica (cabbage /mustard /rape)<br />

OH-F Citrus High proportions <strong>of</strong> Br<strong>as</strong>sica types, Mimosa and Allium<br />

OH-G Honey dew little pollen w<strong>as</strong> found on the slide<br />

OH-H Acacia High proportion <strong>of</strong> Acacia, Br<strong>as</strong>sica and Ambrosia<br />

OH-I Honey dew little pollen w<strong>as</strong> found on the slide


Figure 3.3: Image <strong>of</strong> pollen present in Omani <strong>honey</strong> samples at 100x magnification.<br />

125<br />

OH-B<br />

Lime<br />

Graminae<br />

Acacia<br />

OH-C<br />

Marigold<br />

Graminae<br />

Rose<br />

OH-D<br />

Not<br />

Known<br />

OH-E<br />

Eucalyptus<br />

Daisy<br />

Br<strong>as</strong>sica<br />

OH-F<br />

Br<strong>as</strong>sica<br />

Mimosa<br />

Allium<br />

OH-H<br />

Acacia


3.2.2.2 Antioxidant activity <strong>as</strong>say:<br />

All <strong>honey</strong> samples were analysed for antioxidant activity which included total<br />

phenolic content and anti- radical activity (Table 3.10). The indication <strong>of</strong> linearity <strong>of</strong><br />

the calibration curve (R 2 = 0.997) w<strong>as</strong> evident between 0 and 250 µg/ml <strong>of</strong> gallic acid<br />

(Fig 2.2). From that equation total phenolic content w<strong>as</strong> calculated for each <strong>honey</strong><br />

sample equivalent to gallic acid. A positive correlation between the antioxidant and<br />

phenol content w<strong>as</strong> found (Table 3.10) which suggests that a higher amount <strong>of</strong><br />

phenolic content present in <strong>honey</strong> can deactivate free radicals and thus provide<br />

protection against dise<strong>as</strong>es. Also differences in phenolic content between <strong>honey</strong>s<br />

were observed marked with a mean <strong>of</strong> 89.7±64.8. OH-H which contain high<br />

proportaion <strong>of</strong> acacia pollen and exhibit dark colour had the highest phenolic content<br />

approaching 244.6 mg gallic acid /Kg <strong>of</strong> <strong>honey</strong> and approximately same antioxidant<br />

activity <strong>as</strong> manuka <strong>honey</strong>. The le<strong>as</strong>t phenolic content and antioxidant activity w<strong>as</strong><br />

observed in OH-I with 46.1eg/kg and 59.5% respectively. This is because the<br />

antioxidant activity <strong>of</strong> <strong>honey</strong> varies and depends mainly on floral source, climate and<br />

environment conditions.<br />

Table 3.10: Amount <strong>of</strong> free radical and phenolic content in each <strong>honey</strong> samples<br />

Honey sample Anti- radical activity % Phenol content (eq/kg)<br />

OH-B 67.3 73.0<br />

OH-C 68.1 80.7<br />

OH-D 61.8 56.9<br />

OH-E 64.7 63.0<br />

OH-F 71.7 99.2<br />

OH-G 62.6 53.8<br />

OH-H 61.4 244.6<br />

OH-I 59.5 46.1<br />

Mean ±SD (n) 64.6±4.1 89.7±64.8<br />

Manuka <strong>honey</strong> 61.1 65.4<br />

126


3.3 Determination <strong>of</strong> antibacterial activity <strong>of</strong> <strong>honey</strong> samples<br />

against test cultures:<br />

3.3.1 Minimum Inhibitory Concentration (MIC) and Minimum<br />

Bactericidal Concentration (MBC) <strong>of</strong> manuka <strong>honey</strong>:<br />

Using sterile manuka MIC and MBC w<strong>as</strong> determined for all <strong>of</strong> the clinical isolates.<br />

Initially MICs were performed by agar incorporation (Table 3.11), but the method<br />

w<strong>as</strong> found to require large amounts <strong>of</strong> <strong>honey</strong>, it w<strong>as</strong> time consuming and MBCs<br />

could not be determined. A broth dilution method w<strong>as</strong> therefore adopted.<br />

3.3.1.1 Agar incorporation method:<br />

In this <strong>as</strong>say, two <strong>honey</strong>s were selected according to initial bio<strong>as</strong>say results (Table<br />

3.7). These were manuka <strong>honey</strong> and Omani <strong>honey</strong>-B. However, because Omani<br />

<strong>honey</strong> did not give clear results and required too large an amount <strong>of</strong> <strong>honey</strong> to reach<br />

the correct MIC, only manuka <strong>honey</strong> w<strong>as</strong> tested here against 10 Acinetobacter<br />

clinical isolates (Table 3.11).<br />

127


Table 3.11: Susceptibility <strong>of</strong> Acinetobacter isolates against manuka <strong>honey</strong><br />

Isolates MIC % (w/v) Mean<br />

MIC%<br />

(w/v)<br />

SD *(n)<br />

E.coli NCTC 10418 5 5 5 5 0<br />

K. oxytoca NCTC 8167 9 9 9 9 0<br />

Acinetobacter 1 7 7 7 7 0<br />

Acinetobacter 2 7 8 8 7.6 0.57<br />

Acinetobacter 3 7 7 7 7 0<br />

Acinetobacter 4 6 6 6 6 0<br />

Acinetobacter 5 8 8 8 8 0<br />

Acinetobacter 6 7 7 8 7.3 0.57<br />

Acinetobacter 7 7 7 7 7 0<br />

Acinetobacter 8 7 7 7 7 0<br />

Acinetobacter 9 7 7 8 7.3 0.57<br />

Acinetobacter 10 7 7 9 7.6 1.15<br />

Mean <strong>of</strong> Means MIC 7.18 0.28<br />

*(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> done in triplicate<br />

The MICs <strong>of</strong> 10 Acinetobacter isolates ranged between 6% (w/v) and 8% (w/v)<br />

manuka <strong>honey</strong>. This indicates a medium variation <strong>of</strong> 2% (w/v) and standard<br />

deviations showed good reproducibility. Clinical isolate four had the lowest MIC at<br />

6% (w/v), where<strong>as</strong> the highest MIC w<strong>as</strong> for clinical isolate five at 8% (w/v). The<br />

mode MIC w<strong>as</strong> 7% (w/v) with four isolates inhibited at this concentration. The<br />

overall mean average <strong>of</strong> MIC w<strong>as</strong> 7.18 ± 0.28 % (w/v) manuka <strong>honey</strong>.<br />

128


3.3.1.2 Broth dilution method:<br />

The minimum inhibitory concentration (MIC) and Minimum Bactericidal<br />

Concentration (MBC) for all MDR & ESBL clinical isolates against manuka can be<br />

seen in Tables 3.12 to 3.16. A microtitre plate <strong>as</strong>say w<strong>as</strong> preferred to the agar<br />

incorporation method because <strong>of</strong> the small quantity <strong>of</strong> <strong>honey</strong> used and the ability to<br />

perform replicate <strong>as</strong>says in the same plate. Besides that, MBC values could be e<strong>as</strong>ily<br />

found by sub-culturing from non turbid wells onto nutrient agar plates. Also,<br />

examining MIC/MBC ratios obtained by broth dilutions can be used <strong>as</strong> an indicator<br />

<strong>of</strong> mode <strong>of</strong> inhibition (bacteriostatic or bactericidal). The <strong>antimicrobial</strong> <strong>agent</strong><br />

considered to have a bactericidal action is when MIC/MBC ratio < 4 while the<br />

bacteriostatic action is when this ratio become >4 (Levison 2004). From all<br />

MIC/MBC observations (Table 3.12 to 3.16), manuka <strong>honey</strong> w<strong>as</strong> considered to have<br />

a bactericidal mode <strong>of</strong> action.<br />

The mean MIC and MBC and standard deviation for 30 MDR stains <strong>of</strong> Acinetobacter<br />

data derived from triplicate experiments were illustrated (Table 3.12)<br />

129


Table 3.12: Sensitivity <strong>of</strong> 30 Acinetobacter isolates treated with manuka <strong>honey</strong> by<br />

broth dilution method<br />

Isolates<br />

No.<br />

1.<br />

2.<br />

3.<br />

4.<br />

5.<br />

6.<br />

7.<br />

8.<br />

9.<br />

10.<br />

11.<br />

12.<br />

13.<br />

14.<br />

15. *<br />

Mean MIC<br />

± SD (n)<br />

Mean<br />

MBC ± SD<br />

(n)<br />

Mean MIC<br />

%(w/v)<br />

±SD(3)<br />

7 ±1<br />

7.3 ±1.5<br />

7 ±1<br />

6 ±0<br />

8 ±1<br />

7.5 ±1.5<br />

7 ±1<br />

7 ±0<br />

8 ±1<br />

8 ±0<br />

7 ±1<br />

8 ±1<br />

7 ±0<br />

6 ±1<br />

9 ±1.7<br />

Mean MBC<br />

%(w/v)<br />

±SD (3)<br />

7 ±1.7<br />

8 ±1.7<br />

8 ±1.7<br />

10 ±1<br />

10 ±0<br />

9 ±1<br />

9 ±1<br />

10 ±0<br />

9 ±0<br />

11 ±0<br />

10 ±1<br />

10 ±0<br />

9 ±1.7<br />

7 ±1<br />

11 ±1<br />

7.17 ± 0.7 (30)<br />

9.2 ± 1.3 (30)<br />

(*)The le<strong>as</strong>t susceptible strain to manuka <strong>honey</strong><br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> done in triplicate<br />

130<br />

Isolates<br />

No.<br />

16.<br />

17.<br />

18.<br />

19.<br />

20.<br />

21.<br />

22.<br />

23.<br />

24.<br />

25.<br />

26.<br />

27.<br />

28.<br />

29.<br />

30.<br />

Mean MIC<br />

%(w/v)<br />

± SD(3)<br />

6 ±0<br />

6 ±0<br />

8 ±1<br />

7 ±2.6<br />

8 ±2<br />

7 ±1.7<br />

7 ±1<br />

7 ±1<br />

7 ±1<br />

7 ±0<br />

7 ±1.7<br />

6 ±1<br />

7 ±0<br />

8 ±1<br />

8 ±0<br />

Mean MBC<br />

%(w/v)<br />

±SD (3)<br />

7 ±1<br />

7 ±0<br />

9 ±1<br />

9 ±1.7<br />

10 ±1.7<br />

10 ±2<br />

7 ±1<br />

9 ±0<br />

8 ±1<br />

10 ±0<br />

11 ±0<br />

10 ±1<br />

10 ±0<br />

10 ±1<br />

11 ±0


The MIC range <strong>of</strong> 30 Acinetobacter isolates w<strong>as</strong> between 6% (w/v) and 9% (w/v)<br />

manuka <strong>honey</strong> and the MBC range w<strong>as</strong> between 7% (w/v) and 11% (w/v) (Table<br />

3.12). Considering that MICs are normally performed by doubling dilutions, these<br />

differences are not large. Five clinical isolates (4, 14, 16, 17 & 27) have the lowest<br />

MIC at 6% (w/v), where<strong>as</strong> the lowest MBC at 7% (w/v) w<strong>as</strong> also found in five<br />

clinical isolates (1, 14, 16, 17 & 22). Hence isolates 14, 16 and 17 were the most<br />

susceptible cultures to manuka <strong>honey</strong>. However, the highest MIC and, MBC w<strong>as</strong><br />

observed in clinical isolate fifteen (15*) at 9% (w/v) and 11% (w/v) respectively<br />

(Table 3.12). The difference between MIC and MBC w<strong>as</strong> < 4 for all isolates which<br />

means Acinetobacter exhibited bactericidal mode <strong>of</strong> inhibition with manuka <strong>honey</strong>.<br />

The mode MIC w<strong>as</strong> 7% (w/v) with almost half <strong>of</strong> isolates inhibited at this<br />

concentration and the mode MBC w<strong>as</strong> 10% (w/v) with eleven isolates killed at this<br />

concentration. The overall mean average <strong>of</strong> MIC and MBC were 7.17±0.7 (%w/v)<br />

and 9.2± 1.3 (%w/v) respectively (Table 3.12). This finding correlates with the<br />

study published by Blair et al., (2009) who reported the MIC <strong>of</strong> Acinetobacter with<br />

8.1±1.5 (%w/v) using Medi<strong>honey</strong>. In addition similar potency against Acinetobacter<br />

(6-8 %v/v) h<strong>as</strong> been reported by George & Cutting 2007.<br />

The inhibition <strong>of</strong> 10 ESBLs Klebsiella isolates were also determined with manuka<br />

<strong>honey</strong> and the mean MIC, MBC values were derived from duplicate expirements<br />

with standard deviation (SD) obtained (Table 3.13)<br />

131


Table 3.13: Sensitivity <strong>of</strong> 10 Klebsiella isolates to manuka <strong>honey</strong> tested by the broth<br />

dilution method<br />

Klebsiella<br />

Isolate<br />

Kleb-1<br />

Kleb-2<br />

Kleb-3<br />

Kleb-4<br />

Kleb-5*<br />

Kleb-6<br />

Mean<br />

MIC<br />

%(w/v)<br />

±SD(2)<br />

12 ±0<br />

8 ±1.4<br />

12 ±1.4<br />

12 ±0<br />

13 ±0<br />

12 ±0<br />

Mean<br />

MBC<br />

%(w/v)<br />

±SD(2)<br />

12 ±0<br />

10 ±0<br />

13 ±1.4<br />

13 ±1.4<br />

16 ±0<br />

13 ±0<br />

(*) The le<strong>as</strong>t susceptible isolate<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> repeated twice.<br />

The MICs <strong>of</strong> 10 Klebsiella isolates were found to range between 8% & 13% (w/v)<br />

manuka <strong>honey</strong> and between 10% & 16% (w/v) for MBC (Table 3.13). Although<br />

these values extended over a slightly wider range than the values obtained with<br />

Acinetobacter isolates, the variation w<strong>as</strong> not large. Clinical isolate two demonstrated<br />

the lowest MIC where 8% (w/v) manuka <strong>honey</strong> were required to inhibit this<br />

bacterium and the highest MIC and MBC values were found in clinical isolate five at<br />

13% (w/v) and 16% (w/v), respectively in the Klebsiella cohort. The mode MIC w<strong>as</strong><br />

12% (w/v) manuka <strong>honey</strong> with eight isolates inhibited at this concentration and the<br />

mode MBC w<strong>as</strong> 13% (w/v) with four isolates killed at this concentration. The<br />

MIC/MBC ratio w<strong>as</strong> less than 4 for all Klebsiella isolates tested which proved the<br />

bactericidal mode <strong>of</strong> manuka <strong>honey</strong> on this species. The overall mean average <strong>of</strong><br />

MIC and MBC were 11.7±1.3 & 12.9±1.7 (%w/v) respectively which also fairly<br />

correlate with Blair et al., (2009) finding with MIC <strong>of</strong> Klebsiella 13±2.4 (%w/v).<br />

132<br />

Klebsiella<br />

Isolate<br />

Kleb-7<br />

Kleb-8<br />

Kleb-9<br />

Kleb-10<br />

Mean ± SD<br />

(n)<br />

Mean<br />

MIC<br />

%(w/v)<br />

±SD(2)<br />

12 ±1.4<br />

12 ±2.8<br />

12 ±0<br />

12 ±0<br />

11.7± 1.3<br />

(10)<br />

Mean<br />

MBC<br />

%(w/v)<br />

±SD(2)<br />

15 ±1.4<br />

12 ±2.8<br />

13 ±0<br />

12 ±1.4<br />

12.9± 1.7<br />

(10)


Mean MIC and MBC (%w/v) values <strong>of</strong> manuka <strong>honey</strong> were determined for eight<br />

ESBLs strains <strong>of</strong> Serratia and E.coli and the standard deviations (SD) were<br />

calculated from duplicate expirements (Table 3.14)<br />

Table 3.14: Sensitivity <strong>of</strong> 8 Serratia and 8 E.coli isolates to manuka <strong>honey</strong> using a<br />

broth dilution method<br />

SERRATIA<br />

ISOLATES<br />

Serratia-1<br />

Serratia -2<br />

Serratia -3<br />

Serratia -4*<br />

Serratia -5<br />

Serratia -6<br />

Serratia -7<br />

Serratia -8<br />

Mean ± SD<br />

(n)<br />

Mean MIC<br />

%(w/v)<br />

±SD(2)<br />

10 ±0<br />

15 ±0<br />

13 ±1.4<br />

15 ±1.4<br />

15 ±0<br />

14 ±0<br />

13 ±0<br />

14 ±0<br />

13.3 ± 1.7<br />

(8)<br />

Mean MBC<br />

%(w/v)<br />

±SD(2)<br />

12 ±0<br />

18 ±0<br />

22 ±1.4<br />

20 ±0<br />

16 ±0<br />

16 ±0<br />

14 ±0<br />

18 ±1.4<br />

17 ± 3.2<br />

(8)<br />

(*) Le<strong>as</strong>t susceptible isolates<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> repeated twice<br />

The MIC values <strong>of</strong> 8 Serratia isolates ranged between 10% & 15% (w/v) and MBCs<br />

were between 12% & 22% (w/v) for manuka <strong>honey</strong>. Although there were large<br />

variations in the MIC and MBC ranges noted, manuka <strong>honey</strong> still demonstrated<br />

bactericidal action (Table 3.14). Concentrations <strong>of</strong> 13.3 ± 1.7 % (w/v) were required<br />

to inhibit test isolate. Serratia w<strong>as</strong> considered to be the le<strong>as</strong>t susceptible isolates<br />

against manuka <strong>honey</strong> among the other species tested.<br />

Susceptibility <strong>of</strong> the E. coli isolates tested w<strong>as</strong> similar to that <strong>of</strong> Klebsiella with<br />

MICs against manuka <strong>honey</strong> varying between 8% & 13% (w/v) (Table 3.14). The<br />

bactericidal action <strong>of</strong> <strong>honey</strong> also observed on E.coli where the MIC/MBC ratio w<strong>as</strong><br />

133<br />

E.COLI<br />

ISOLATES<br />

E.coli-1<br />

E.coli-2<br />

E.coli-3<br />

E.coli -4<br />

E.coli -5<br />

E.coli -6<br />

E.coli -7*<br />

E.coli -8<br />

Mean ± SD<br />

(n)<br />

Mean MIC<br />

%(w/v)<br />

±SD(2)<br />

10 ±1.4<br />

10 ±0<br />

10 ±0<br />

10 ±0<br />

10 ±0<br />

12 ±0<br />

13 ±0<br />

8 ±0<br />

10.4 ± 1.5<br />

(8)<br />

Mean MBC<br />

%(w/v)<br />

±SD(2)<br />

12 ±1.4<br />

10 ±1.4<br />

10 ±0<br />

10 ±0<br />

12 ±0<br />

12 ±0<br />

13 ±0<br />

8 ±0<br />

10.9 ± 1.6<br />

(8)


very close (2 or less). It indicates that E.coli w<strong>as</strong> inhibited and killed at the same<br />

concentration.<br />

There are several reports documenting the sensitivity <strong>of</strong> different <strong>honey</strong>s including<br />

manuka <strong>honey</strong> against E.coli (Blair et al., 2009; George & Cutting 2007; Tan et al.,<br />

2009; Sherlock et al., 2010). Some <strong>of</strong> them have an almost similar MIC value that<br />

w<strong>as</strong> determined in this <strong>project</strong> with 10 % (w/v) (Lusby et al., 2005). Unlike E.coli,<br />

limited studies documented the sensitivity <strong>of</strong> <strong>honey</strong> against Serratia.<br />

134


Table 3.15: Susceptibility <strong>of</strong> 15 Enterobacter isolates to manuka <strong>honey</strong> determined<br />

by broth dilution method and the SD were determined from the duplicate<br />

expirements.<br />

ISOLATES<br />

Enterobacter-1<br />

Enterobacter-2<br />

Enterobacter-3<br />

Enterobacter-4<br />

Enterobacter-5<br />

Enterobacter-6<br />

Enterobacter-7<br />

Enterobacter-8<br />

Mean<br />

MIC<br />

%(w/v)<br />

±SD(2)<br />

9 ±0<br />

9 ±0<br />

9 ±0<br />

13 ±0<br />

11 ±0<br />

9 ±0<br />

8 ±0<br />

8 ±0<br />

Mean<br />

MBC<br />

%(w/v)<br />

±SD(2)<br />

15 ±1.4<br />

13 ±0<br />

13 ±0<br />

14 ±0<br />

14 ±0<br />

11 ±1.4<br />

12 ±0<br />

11 ±0<br />

(*) le<strong>as</strong>t susceptible isolate<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> repeated twice<br />

The MICs <strong>of</strong> 15 Enterobacter isolates ranged between 8% & 13% (w/v) manuka<br />

<strong>honey</strong> and MBCs were between 11% & 14% (w/v) (Table 3.15). The mode MIC &<br />

MBC w<strong>as</strong> 9% & 14% respectively. The sensitivity <strong>of</strong> Enterobacter w<strong>as</strong> similar to<br />

that <strong>of</strong> E.coli. The closest MIC, MBC ranges indicates the bactericidal mode <strong>of</strong><br />

manuka <strong>honey</strong>. These results were corresponded with a previous report (Lusby et al.,<br />

2005), where the inhibitory concentration <strong>of</strong> Enterobacter w<strong>as</strong> 10% (w/v). However,<br />

this contr<strong>as</strong>t with Tan et al., (2009) finding where manuka <strong>honey</strong> also used at<br />

concentration <strong>of</strong> 20% (w/v) compared to our finding with 10.1± 1.7 (%w/v).<br />

135<br />

ISOLATES<br />

Enterobacter-9*<br />

Enterobacter-10<br />

Enterobacter-11<br />

Enterobacter-12<br />

Enterobacter-13<br />

Enterobacter-14<br />

Enterobacter-15<br />

Mean ± SD<br />

(n)<br />

Mean<br />

MIC<br />

%(w/v)<br />

±SD(2)<br />

13 ±0<br />

11 ±0<br />

11 ±0<br />

8 ±1.4<br />

11 ±0<br />

12 ±0<br />

10 ±0<br />

10.1± 1.7<br />

(15)<br />

Mean<br />

MBC<br />

%(w/v)<br />

±SD(2)<br />

14 ±0<br />

14 ±0<br />

14 ±0<br />

12 ±0<br />

14 ±0<br />

13 ±0<br />

13 ±0<br />

13.1± 1.2<br />

(15)


Table 3.16: Susceptibility <strong>of</strong> 12 Citrobacter isolates to manuka <strong>honey</strong> using broth<br />

dilution method and the SD were determined from the duplicate expirements.<br />

ISOLATES<br />

Citrobacter 1<br />

Citrobacter 2<br />

Citrobacter 3<br />

Citrobacter 4<br />

Citrobacter 5*<br />

Citrobacter 6<br />

Citrobacter 7<br />

Mean<br />

MIC<br />

%(w/v)<br />

±SD(2)<br />

9 ±0<br />

10 ±1.4<br />

10 ±0<br />

10 ±0<br />

11 ±0<br />

10 ±0<br />

9 ±1.4<br />

Mean<br />

MBC<br />

%(w/v)<br />

±SD(2)<br />

12 ±0<br />

15 ±0<br />

14 ±0<br />

15 ±0<br />

15 ±0<br />

14 ±0<br />

11 ±0<br />

(*) le<strong>as</strong>t <strong>honey</strong>-susceptible isolate.<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> repeated twice<br />

The inhibition ranges <strong>of</strong> 12 Citrobacter isolates between 8% & 11% (w/v) for MIC<br />

and between 11% & 15% (w/v) for MBC. This indicates a medium variation <strong>of</strong> 3%<br />

& 4% (w/v) for MIC and MBC respectively. The overall average MIC <strong>of</strong><br />

Citrobacter w<strong>as</strong> similar to that reported by Lusby et al., 2005 with 10 % (w/v) using<br />

manuka <strong>honey</strong> and Blair et al., 2009 with 9.1±3 % (w/v) using medi<strong>honey</strong>.<br />

136<br />

ISOLATES<br />

Citrobacter 8<br />

Citrobacter 9<br />

Citrobacter 10<br />

Citrobacter 11<br />

Citrobacter12<br />

Mean ± SD<br />

(n)<br />

Mean<br />

MIC<br />

%(w/v)<br />

±SD(2)<br />

9 ±0<br />

10 ±0<br />

10 ±0<br />

8 ±1.4<br />

10 ±0<br />

9.7± 0.8<br />

(12)<br />

Mean<br />

MBC<br />

%(w/v)<br />

±SD(2)<br />

11 ±1.4<br />

13 ±0<br />

15 ±0<br />

11 ±0<br />

14 ±0<br />

13.3± 1.7<br />

(12)


3.3.2 Sensitivity <strong>of</strong> MDR and ESBLs to Omani <strong>honey</strong>:<br />

The sensitivity <strong>of</strong> all <strong>of</strong> the clinical isolates w<strong>as</strong> determined against the four Omani<br />

<strong>honey</strong> samples with the highest total antibacterial activity detected (Table 3.7) using<br />

the broth dilution method. MICs and MBCs % (w/v) w<strong>as</strong> performed at once for all<br />

85 MDR/ESBLs isolates, mean and SD w<strong>as</strong> calculated (Tables 3.17 to 3.22). Similar<br />

to manuka <strong>honey</strong>, it w<strong>as</strong> deduced that Omani <strong>honey</strong> had a bactericidal mode <strong>of</strong><br />

action against test isolates because it w<strong>as</strong> found that OH-B, OH-C and OH-F<br />

exhibited relatively similar values for MIC and MBC for each species.<br />

(The highlighted box indicates the lowest MIC mean± SD (highest susceptibility)<br />

observed <strong>of</strong> that isolate among the other type <strong>of</strong> <strong>honey</strong>).<br />

Table 3.17: Susceptibility <strong>of</strong> 30 MDR Acinetobacter isolates against 4 types <strong>of</strong><br />

Omani <strong>honey</strong> using broth dilution method.<br />

Isolate<br />

No.<br />

MIC<br />

(%w/v)<br />

OH-B OH-C OH-G OH-F<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

137<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

1. 18 18 14 14 16 16 14 14<br />

2. 14 14 16 18 18 20 18 18<br />

3. 20 20 18 18 20 20 14 14<br />

4. 41 18 18 20 20 22 16 16<br />

5. 41 18 18 20 16 18 20 20<br />

6. 41 16 18 18 22 24 14 14<br />

7. 41 16 20 20 20 22 14 14<br />

8. 18 20 16 22 20 22 16 16<br />

9. 20 20 18 18 24 24 18 22<br />

10. 20 20 18 18 18 18 18 18<br />

11. 41 18 20 20 20 22 18 18<br />

12. 41 16 18 22 20 22 20 20


13. 41 18 20 20 16 16 14 14<br />

14. 41 18 16 16 24 24 20 20<br />

15. 41 20 16 16 20 22 18 18<br />

16. 41 20 20 20 22 24 18 18<br />

17. 41 20 20 20 18 22 20 20<br />

18. 41 20 18 20 18 22 18 18<br />

19. 41 20 20 20 20 22 18 18<br />

20. 41 18 20 20 22 >24 18 18<br />

21. 20 20 20 20 24 >24 18 18<br />

22. 16 18 18 22 24 >24 18 20<br />

23. 14 14 18 22 20 22 20 20<br />

24. 18 20 20 20 22 24 18 20<br />

25. 18 20 18 18 24 24 20 20<br />

26. 18 18 18 20 24 >24 18 18<br />

27. 18 18 16 16 22 22 18 18<br />

28. 18 18 18 20 20 20 18 18<br />

29. 18 18 16 20 20 22 18 18<br />

30. 18 20 20 20 22 22 18 18<br />

Mean<br />

± SD<br />

(30)<br />

16.6±<br />

2<br />

18.4±<br />

1.7<br />

18.1±<br />

1.6<br />

19.2±<br />

1.9<br />

138<br />

20.5±<br />

2.4<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> done once<br />

21.8±<br />

2.3<br />

17.6±<br />

1.9<br />

17.8±<br />

2.1


Table 3.18: Susceptibility <strong>of</strong> 11 Klebsiella isolates against 4 types <strong>of</strong> Omani <strong>honey</strong><br />

using broth dilution method.<br />

Isolate<br />

No.<br />

Mean<br />

± SD<br />

(11)<br />

MIC<br />

(%w/v)<br />

OH-B OH-C OH-G OH-F<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

139<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

1. 12 14 14 14 20 24 14 18<br />

2. 18 18 18 20 22 26 18 20<br />

3. 20 20 20 20 24 28 20 24<br />

4. 20 22 20 20 26 28 20 22<br />

5. 20 22 20 20 24 26 18 22<br />

6. 16 16 18 18 22 24 18 20<br />

7. 20 24 20 22 24 28 20 22<br />

8. 16 20 18 18 20 24 16 18<br />

9. 14 16 18 20 20 22 18 20<br />

10. 14 16 18 18 20 24 16 20<br />

11. 16 16 18 18 22 24 18 22<br />

12. 20 22 20 22 24 26 20 22<br />

17.2±3 18.8±<br />

3.2<br />

18.5±<br />

1.7<br />

19± 2 22± 2 25±<br />

1.9<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> done once<br />

18±<br />

1.9<br />

20.8±<br />

1.8


Table 3.19: Susceptibility <strong>of</strong> 10 E.coli isolates against 4 types <strong>of</strong> Omani <strong>honey</strong> using<br />

broth dilution method.<br />

Isolate<br />

No.<br />

Mean ±<br />

SD (10)<br />

MIC<br />

(%w/v)<br />

OH-B OH-C OH-G OH-F<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

140<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

1. 18 20 20 20 22 24 20 20<br />

2. 20 20 20 20 24 24 22 22<br />

3. 20 22 20 20 26 26 22 24<br />

4. 16 18 18 20 20 22 18 18<br />

5. 20 22 20 20 26 28 22 22<br />

6. 20 20 20 20 24 24 18 20<br />

7. 18 18 18 18 22 24 18 20<br />

8. 18 20 18 22 22 24 18 20<br />

9. 20 20 20 20 26 28 20 22<br />

10. 18 18 20 20 24 26 18 20<br />

18.8±<br />

1.3<br />

19.8±<br />

1.4<br />

19.4±1 20±<br />

0.9<br />

23.6±2 25±<br />

1.9<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> done once<br />

19.6±<br />

1.8<br />

20.8±<br />

1.6


Table 3.20: Susceptibility <strong>of</strong> 15 Enterobacter isolates against 4 types <strong>of</strong> Omani<br />

<strong>honey</strong> using broth dilution method.<br />

Isolate<br />

No.<br />

MIC<br />

(%w/v)<br />

OH-B1 OH-C OH-G OH-F<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

141<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

1. 22 22 18 24 22 26 18 22<br />

2. 20 20 20 22 26 30 20 24<br />

3. 22 >24 20 22 30 >30 20 24<br />

4. 22 24 20 22 26 28 20 24<br />

5. 22 >24 20 20 26 30 20 22<br />

6. 22 22 18 20 24 24 18 22<br />

7. 22 22 20 22 26 >30 18 20<br />

8. 24 24 22 22 30 30 24 24<br />

9. >24 >24 24 24 30 >30 22 24<br />

10. 22 24 22 24 30 >30 22 28<br />

11. 22 >24 20 24 26 30 20 22<br />

12. 22 22 22 22 30 >30 20 20<br />

13. 22 >24 20 22 22 24 22 24<br />

14. 22 >24 18 22 24 24 18 18<br />

15. 20 20 18 22 24 24 18 20<br />

Mean<br />

±SD<br />

(15)<br />

21.6±<br />

0.9<br />

22.9±<br />

1.5<br />

20.1±<br />

1.7<br />

22.2±<br />

1.3<br />

26.4±<br />

2.9<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> done once<br />

28±<br />

2.7<br />

20±<br />

1.8<br />

22.5±<br />

2.4


Table 3.21: Susceptibility <strong>of</strong> 12 Citrobacter isolates against 4 types <strong>of</strong> Omani <strong>honey</strong><br />

using broth dilution method.<br />

Isolate<br />

No.<br />

MIC<br />

(%w/v)<br />

OH-B OH-C OH-G OH-F<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

142<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

1. 22 >24 20 22 28 >28 18 18<br />

2. 24 24 20 22 >28 >28 22 24<br />

3. 18 22 16 18 28 >28 16 18<br />

4. 16 16 14 16 28 >28 16 16<br />

5. 20 24 16 18 >28 >28 16 16<br />

6. 18 18 16 18 >28 >28 16 18<br />

7. 16 18 14 18 28 >28 14 16<br />

8. 18 18 16 20 26 >28 14 18<br />

9. 16 16 14 16 28 >28 14 16<br />

10. 18 18 16 18 22 24 14 16<br />

11. 16 16 14 16 28 >28 16 18<br />

12. 16 16 14 18 24 26 18 20<br />

Mean<br />

± SD<br />

(12)<br />

18.1±<br />

2.6<br />

18.5 ±<br />

3.2<br />

15.8±<br />

2.1<br />

18.3<br />

±2<br />

26 ±<br />

2.5<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> done once<br />

27.5±<br />

1.4<br />

16.1±<br />

2.3<br />

17.8±<br />

2.3


Table 3.22: Susceptibility <strong>of</strong> 8 Serratia isolates against 4 types <strong>of</strong> Omani <strong>honey</strong><br />

using broth dilution method.<br />

Isolate<br />

No.<br />

MIC<br />

(%w/v)<br />

OH-B OH-C OH-G OH-F<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

143<br />

MIC<br />

(%w/v)<br />

MBC<br />

(%w/v)<br />

MIC<br />

(%w/v) MBC<br />

(%w/v)<br />

1. 24 >24 18 20 >28 >28 18 20<br />

2. 20 22 20 24 >28 >28 20 24<br />

3. 22 24 20 24 >28 >28 20 24<br />

4. 20 22 20 26 >28 >28 14 26<br />

5. 20 22 20 24 >28 >28 14 28<br />

6. 20 24 22 24 >28 >28 14 28<br />

7. 18 20 20 22 >28 >28 20 24<br />

8. 18 18 22 26 >28 >28 16 16<br />

Mean ±<br />

SD (8)<br />

20.2±<br />

1.9<br />

21.7±<br />

2.1<br />

20.2±<br />

1.2<br />

23.7±<br />

1.9<br />

(n) number <strong>of</strong> isolates tested, this expirement w<strong>as</strong> done once<br />

>28 >28 17±<br />

2.8<br />

23.7±<br />

4<br />

According to the above tables Acinetobacter and Citrobacter were the most sensitive<br />

(low MIC recorded) isolates to Omani <strong>honey</strong>s and to manuka <strong>honey</strong> <strong>as</strong> well (lowest<br />

MIC recorded). Acinetobacter were inhibited with Omani <strong>honey</strong>s B with MIC 16.6<br />

(%w/v) (Table 3.17). The remaining two Omani <strong>honey</strong>s C & F with MIC 15.8 &<br />

16.6 (%w/v) respectively were more inhibitory to Citrobacter (Table 3.21). Serratia<br />

were less susceptible to most <strong>honey</strong>s tested with MIC ranged between 17 - >28<br />

(%w/v) (Table 3.22).<br />

Thus it w<strong>as</strong> difficult to state which is the most effective <strong>honey</strong> because each species<br />

responded in a different way. However, OH-G had the highest MIC/MBC ratio<br />

among the other types <strong>of</strong> <strong>honey</strong>. But OH-B had lowest MIC values reported for<br />

Acinetobacter, E.coli and Klebsiella so can be considered to be the most effective


(Table 3.17-3.19), where<strong>as</strong> OH-C and OH-F were more effective against<br />

Citrobacter, Enterobacter and Serratia (Table 3.20-3.22).<br />

The susceptibility <strong>of</strong> isolates tested w<strong>as</strong> recorded against manuka <strong>honey</strong> and 4 types<br />

<strong>of</strong> Omani <strong>honey</strong> tested according to above tables were:<br />

Acinetobacter > Citrobacter > Klesiella > E.coli > Enterobacter > Serratia<br />

144


3.4 Time Kill Curves<br />

One representative isolate from each <strong>of</strong> the different cohorts <strong>of</strong> clinical test species<br />

(the le<strong>as</strong>t susceptible one) w<strong>as</strong> selected to investigate the kinetics <strong>of</strong> inhibition with<br />

manuka <strong>honey</strong>. Bactericidal concentrations <strong>of</strong> manuka <strong>honey</strong> were required in each<br />

experiment for each isolate. Approximately doubled MIC values <strong>of</strong> <strong>honey</strong><br />

concentration were therefore utilised (Table 3.23).<br />

Table 3.23: Cultures and <strong>honey</strong> concentrations used in the time-kill curves <strong>as</strong>say<br />

Test organism Strain No. MIC (%w/v) Test conc. used<br />

145<br />

2x MIC (%w/v)<br />

Acinetobacter 15 9 20<br />

E.coli 7 13 30<br />

Klebsiella 5 13 30<br />

Enterobacteria 9 13 30<br />

Citrobacter 2 10 20<br />

Serratia 4 15 30<br />

3.4.1 Inhibition <strong>of</strong> test organisms by manuka <strong>honey</strong> monitored by<br />

optical density:<br />

Initially the optical density <strong>of</strong> each isolate incubated in isosensitest broth for 24 hours<br />

with and without a bactericidal concentration <strong>of</strong> manuka <strong>honey</strong> w<strong>as</strong> plotted against<br />

time (Fig 3.4 to 3.9). As shown in Figures 3.4 to 3.9, no incre<strong>as</strong>e in optical density<br />

observed over the 24 h exposure to <strong>honey</strong> thus w<strong>as</strong> interpreted <strong>as</strong> complete inhibition<br />

<strong>of</strong> growth <strong>of</strong> these six isolates at test concentration <strong>of</strong> manuka <strong>honey</strong> used. A steady<br />

incre<strong>as</strong>e in optical density w<strong>as</strong> observed during 24 h for all six isolates without the<br />

addition <strong>of</strong> <strong>honey</strong> w<strong>as</strong> expected.


Optical density at 550 nm<br />

Figure 3.4: The effect <strong>of</strong> manuka <strong>honey</strong> (%w/v) on the growth <strong>of</strong> Acinetobacter<br />

2.2<br />

2<br />

1.8<br />

1.6<br />

1.4<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

0 200 400 600 800 1000 1200 1400 1600<br />

Figure 3.5: The effect <strong>of</strong> manuka <strong>honey</strong> (%w/v) on the growth <strong>of</strong> E.coli<br />

Optical density at 550nm<br />

2<br />

1.8<br />

1.6<br />

1.4<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

The growth curve <strong>of</strong> Acinetobacter<br />

Time (minutes)<br />

Acinetobacter without <strong>honey</strong> Acinetobacter with 20% (w/v) manuka <strong>honey</strong><br />

The growth curve <strong>of</strong> E.coli<br />

0 200 400 600 800 1000 1200 1400 1600<br />

Time (minutes)<br />

E.coli without <strong>honey</strong> E.coli with 30% (w/v) manuka <strong>honey</strong><br />

146


Figure 3.6: The effect <strong>of</strong> manuka <strong>honey</strong> (%w/v) on the growth <strong>of</strong> Klebsiella<br />

Optical density at 550nm<br />

2<br />

1.8<br />

1.6<br />

1.4<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

0 200 400 600 800 1000 1200 1400 1600<br />

Figure 3.7: The effect <strong>of</strong> manuka <strong>honey</strong> (%w/v) on the growth <strong>of</strong> Citrobacter<br />

Optical density at 550 nm<br />

1.8<br />

1.6<br />

1.4<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

The growth curve <strong>of</strong> Klebsiella<br />

Time (minutes)<br />

Klebsiella without <strong>honey</strong> Klebsiella with 30% manuka <strong>honey</strong><br />

The growth curve <strong>of</strong> Citrobacter<br />

0 200 400 600 800 1000 1200 1400 1600<br />

Time (minutes)<br />

Citrobacter without <strong>honey</strong> Citrobacter with 20% (w/v) manuka <strong>honey</strong><br />

147


Figure 3.8: The effect <strong>of</strong> manuka <strong>honey</strong> (%w/v) on the growth <strong>of</strong> Enterobacter<br />

Optical density at 550 nm<br />

3.5<br />

3<br />

2.5<br />

2<br />

1.5<br />

1<br />

0.5<br />

0<br />

0 200 400 600 800 1000 1200 1400 1600<br />

Figure 3.9: The effect <strong>of</strong> manuka <strong>honey</strong> (%w/v) on the growth <strong>of</strong> Serratia<br />

Optical density at 550 nm<br />

2.2<br />

2<br />

1.8<br />

1.6<br />

1.4<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

The growth curve <strong>of</strong> Enterobacter<br />

Time (minutes)<br />

Enterobacter without <strong>honey</strong> Enterobacter with 30% (w/v) manuka <strong>honey</strong><br />

The growth curve <strong>of</strong> Serratia<br />

0 200 400 600 800 1000 1200 1400 1600<br />

Time (minutes)<br />

Serratia without <strong>honey</strong> Serratia with 30% (w/v) manuka <strong>honey</strong><br />

148


3.4.2 Inhibition <strong>of</strong> test organisms by manuka <strong>honey</strong> monitored by<br />

total viable count<br />

Time-kill curves were repeated for each representative test organism <strong>as</strong> above,<br />

except that viability w<strong>as</strong> monitored by total viable count (TVC) using the surface<br />

drop method (Miles & Misra 1938). For total culturable counts decimal dilutions<br />

from 10 -1 to 10 -7 were determined. The lowest dilution that gives a re<strong>as</strong>onable count<br />

w<strong>as</strong> selected and calculated the colony forming units (CFU) to determine the actual<br />

viable cells in 1 ml. The mean data (Log10 CFU/ml) against time were plotted for<br />

each isolate on the graph. A linear regression analysis for each set <strong>of</strong> data w<strong>as</strong><br />

performed. Time-kill curves were obtained for Acinetobacter (Fig 3.10), E.coli (Fig<br />

3.11), Klebsiella (Fig 3.12), Enterobacter (Fig 3.13), Citrobacter (Fig 3.14) and<br />

Serratia (Fig 3.15). Each graph illustrated the difference in viability between the test<br />

(with <strong>honey</strong> treatment) and control (without <strong>honey</strong> treatment). The activity <strong>of</strong><br />

manuka <strong>honey</strong> against all strains demonstrated by maximal bacterial killing and<br />

surviving bacteria were counted at 0, 30, 60, 90, 120, 180, 240 and 300 minutes<br />

incubated at 37 o C. From figures bellow it w<strong>as</strong> observed that manuka <strong>honey</strong> at 2 times<br />

the MIC (Table 3.23) w<strong>as</strong> bactericidal for all strains tested up to 5 h. Log reduction<br />

(LD) were calculated for each isolate up to 5 h exposure to manuka <strong>honey</strong> (Table<br />

3.24). T-test w<strong>as</strong> applied to establish the statistical difference between untreated cells<br />

and <strong>honey</strong> treated cells in the selected 6 species (Table 3.25).<br />

149


3.4.2.1 Acinetobacter<br />

Acinetobacter cultivated without <strong>honey</strong> demonstrated a typical growth curve and<br />

reached the stationary ph<strong>as</strong>e after 5 h incubation with broth only (Fig 3.10).<br />

However, the test organism incubated with 2x MIC demonstrated rapid loss <strong>of</strong><br />

viability. Therefore, 20% manuka <strong>honey</strong> resulted in 1.44-log10 reduction in CFU/ml<br />

compared to the starting inoculum (≈99% killing <strong>of</strong> Acinetobacter) at 5 h incubation<br />

(Table 3.24). Although <strong>honey</strong> treated Acinetobacter cells demonstrated the slowest<br />

killing rate among the selected isolates, there w<strong>as</strong> a marked difference in viable cell<br />

count between Acinetobacter with and without <strong>honey</strong> (Table 3.25).<br />

150


This data w<strong>as</strong> done in triplicate<br />

Number <strong>of</strong> bacterial cells Log 10 CFU/ml<br />

Figure 3.10: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> Acinetobacter.<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Time -to-kill curve <strong>of</strong> Acinetobacter<br />

0 200 400 600 800 1000 1200 1400 1600<br />

Time/Minutes<br />

Acinetobacter without <strong>honey</strong> Acinetobacter with 20% (w/v) manuka <strong>honey</strong><br />

151


3.4.2.2 E.coli<br />

The time-kill curve clearly shows an incre<strong>as</strong>e in number <strong>of</strong> E.coli cells without<br />

<strong>honey</strong> treatment (Fig 3.11). However in <strong>honey</strong> treated cells a reduction in the number<br />

<strong>of</strong> E.coli survivors were observed over time by 1.97-log10 reduction in CFU/ml at 5 h<br />

incubation with 30% (w/v) manuka <strong>honey</strong> (Table 3.24). In other words, 30%<br />

manuka <strong>honey</strong> w<strong>as</strong> expected to be capable <strong>of</strong> almost killing 99% (equivalent 2-log10<br />

reduction) <strong>of</strong> the population <strong>of</strong> E.coli within 5 h and to yield an undetectable<br />

bacterial count (


This data w<strong>as</strong> done in triplicate<br />

Figure 3.11: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> E.coli.<br />

Number <strong>of</strong> bacterial cells log10 CFU/ml<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Time -to-kill curve <strong>of</strong> E.coli<br />

0 200 400 600 800 1000 1200 1400 1600<br />

153<br />

Time (minutes)<br />

E.coli without <strong>honey</strong> E.coli with 30% (w/v) manuka <strong>honey</strong>


3.4.2.3 Klebsiella<br />

Klebsiella cultivated without <strong>honey</strong> demonstrated a typical growth curve <strong>as</strong> the<br />

isolates presented above (Fig 3.12). However, the test organism incubated with<br />

2xMIC manuka <strong>honey</strong> demonstrated a marked decre<strong>as</strong>e with 5 x10 5 cfu/ml after 24<br />

hours exposure. Hence 30% manuka <strong>honey</strong> achieved a 1.7-log10 reduction (≈99%<br />

killing) in Klebsiella population at 5 h. The cells continued to decre<strong>as</strong>e till it reached<br />

< 500 CFU in 1 ml at 24 h (Fig 3.12). Addition <strong>of</strong> 30% manuka <strong>honey</strong> therefore<br />

caused significant difference in the viability <strong>of</strong> Klebsiella population (Table 3.25).<br />

154


This data w<strong>as</strong> done in triplicate<br />

Figure 3.12: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> Klebsiella<br />

Number <strong>of</strong> bacterial cells log10 CFU/ml<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Time -to-kill curve <strong>of</strong> Klebsiella<br />

0 200 400 600 800 1000 1200 1400 1600<br />

Time (minutes)<br />

Klebsiella without <strong>honey</strong> Klebsiella with 30% (w/v) manuka <strong>honey</strong><br />

155


3.4.2.4 Citrobacter<br />

Citrobacter incubated with broth alone only showed incre<strong>as</strong>e in cell number for the<br />

first 120 minutes then there were no significant change in colony counts in each time<br />

ponit with incubation beyond 24 h. Viable counts <strong>of</strong> Citrobacter with 20% (w/v)<br />

manuka <strong>honey</strong> showed markedly declined <strong>of</strong> colony forming unit from 1x10 8 cfu/ml<br />

(eight Log CFU) to below 5.5x10 4 cfu/ml (five Log CFU). A time <strong>of</strong> exposure to<br />

<strong>honey</strong> from 4 to 5 h w<strong>as</strong> enough to reduce 99.9% (3-log10 reduction) <strong>of</strong> the<br />

population <strong>of</strong> Citrobacter (Fig 3.13). Citrobacter incubated with 20% (w/v) manuka<br />

<strong>honey</strong> demostrated the greatested bactericidal activity at 5 h incubation with >3-log10<br />

killing unit (Table 3.24). The change in cell count in Citrobacter between non <strong>honey</strong><br />

and <strong>honey</strong> treatment w<strong>as</strong> statistically significant (Table 3.25).<br />

156


This data w<strong>as</strong> done in triplicate<br />

Figure 3.13: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> Citrobacter<br />

Number <strong>of</strong> bacterial cells Log10 CFU/ml<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Time-to-kill curve <strong>of</strong> Citrobacter<br />

0 200 400 600 800 1000 1200 1400 1600<br />

157<br />

Time (minutes)<br />

Citrobacter without <strong>honey</strong> Citrobacter with 20% (w/v) manuka <strong>honey</strong>


3.4.2.5 Enterobacter<br />

The number <strong>of</strong> untreated cells incre<strong>as</strong>ed with time <strong>as</strong> shown below (Fig 3.14). The<br />

total number <strong>of</strong> live Enterobacter cells significantly decre<strong>as</strong>ed when exposed to 30%<br />

manuka <strong>honey</strong> (Table 3.25). These differences were statistically significant (Table<br />

3.25). A slow drop in numbers can be observed over time from 55 x10 6 just below<br />

(eight Log CFU) to 1.5 x10 6 just below (six Log CFU). Manuka <strong>honey</strong> with twice<br />

MIC concentration produced 1.57-log10 decre<strong>as</strong>e in viable counts at 5 h (Fig 3.14).<br />

158


This data w<strong>as</strong> done in triplicate<br />

Figure 3.14: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> Enterobacter<br />

Number <strong>of</strong> bacterial cells Log 10 CFU/ml<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Time -to- kill curve <strong>of</strong> Enterobacter<br />

0 200 400 600 800 1000 1200 1400 1600<br />

159<br />

Time (minutes)<br />

Enterobacter without <strong>honey</strong> Enterobacter with 30% manuka <strong>honey</strong>


3.4.2.6 Serratia<br />

An incre<strong>as</strong>ed number <strong>of</strong> colony forming units <strong>of</strong> Serratia w<strong>as</strong> observed <strong>as</strong> shown by<br />

the trend line in the growth curve without <strong>honey</strong>, while number <strong>of</strong> cells decre<strong>as</strong>ed<br />

with time after exposure to 30% manuka <strong>honey</strong> with 1.87-log10 reduction in CFU/ml<br />

(≈99% killing). There w<strong>as</strong> marked bacterial killing at 5 h incubation with <strong>honey</strong>. The<br />

cells continued to decre<strong>as</strong>e till


Figure 3.15: The effect <strong>of</strong> manuka <strong>honey</strong> on the viability <strong>of</strong> Serratia<br />

Number <strong>of</strong> bacterial cell Log 10 CFU/ml<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

0 200 400 600 800 1000 1200 1400 1600<br />

This data w<strong>as</strong> done in triplicate<br />

Time-to-kill curve <strong>of</strong> Serratia<br />

Time (minutes)<br />

Serratia without <strong>honey</strong> Serratia with 30% (w/v) manuk <strong>honey</strong><br />

161


The Log Reduction (LR) w<strong>as</strong> calculated for each isolate by subtracting the Log CFU<br />

at zero time and the Log CFU at 5 h incubation with 2x respective MICs <strong>of</strong> manuka<br />

<strong>honey</strong>. The higher LR, the f<strong>as</strong>ter w<strong>as</strong> the killing rate (Table 3.24). Acinetobacter w<strong>as</strong><br />

the most susceptible MDR when tested with MIC, but Citrobacter were inhibited at<br />

a f<strong>as</strong>ter rate (Table 3.24).<br />

Table 3.24: Log reduction (LR) for each isolate after 5 h exposure to 2x respective<br />

MICs <strong>of</strong> manuka <strong>honey</strong><br />

Isolate LogCFU/ml + <strong>honey</strong> LR<br />

At 0 h At 5 h<br />

Acinetobacter 7.04 5.6 1.44<br />

E.coli 7.97 6 1.97<br />

Klebsiella 7.39 5.69 1.7<br />

Enterobacter 7.74 6.17 1.57<br />

Citrobacter 8 4.74 3.26<br />

Serratia 7.87 6 1.87<br />

Sensitivity pr<strong>of</strong>ile <strong>of</strong> six tested strains against <strong>honey</strong> at each concentration according<br />

to the table above is:<br />

Citrobacter > E.coli > Serratia > Klebsiella > Enterobacter > Acinetobacter<br />

162


All isolates showed statistically significant differences in the mean <strong>of</strong> cell count<br />

between <strong>honey</strong> treated and untreated cells P


3.5 Effect <strong>of</strong> <strong>honey</strong> on bacterial structure:<br />

Electron microscopy is an essential tool to observe the ultr<strong>as</strong>tructural and physical<br />

changes that occur to cells after adding certain <strong>agent</strong>s. Details <strong>of</strong> surface structures<br />

and bacterial morphology aid an understanding <strong>of</strong> the action <strong>of</strong> various drugs on<br />

bacteria. In this <strong>project</strong> both scanning and transmission electron microscopy w<strong>as</strong><br />

used to determine membrane integrity, morphological changes <strong>of</strong> cells, electron<br />

density <strong>of</strong> the cytopl<strong>as</strong>m and evidence <strong>of</strong> cell division before and after exposure to<br />

<strong>honey</strong>.<br />

3.5.1 Growth Curves:<br />

The exponential cells were collected after 3 h incubation at 37°C in ISB <strong>as</strong> indicated<br />

by the arrows in each <strong>of</strong> the growth curves for Acinetobacter (Fig 3.16), E.coli (Fig<br />

3.17), Klebsiella (Fig 3.18), Enterobacter (Fig 3.19), Citrobacter (Fig 3.20), and<br />

Serratia (Fig 3.21). The exponential ph<strong>as</strong>e w<strong>as</strong> selected because <strong>of</strong> active growth <strong>of</strong><br />

cells w<strong>as</strong> expected to result in many proteins and enzymes being synthesized. These<br />

molecules are considered to be the main target site for various inhibitors and thereby<br />

help in identifying the action <strong>of</strong> an <strong>antimicrobial</strong> <strong>agent</strong>. Biosynthetic pathways in<br />

stationary ph<strong>as</strong>e bacteria are more likely to reflect secondary metabolism, rather than<br />

central pathways. Exponential ph<strong>as</strong>e cultures were treated with and without<br />

approximately twice MIC concentrations <strong>of</strong> <strong>honey</strong> and monitored for 180 minutes.<br />

164


Figure 3.16: Growth curve <strong>of</strong> Acinetobacter in ISB<br />

Optical Density at 550 nm<br />

0.7<br />

0.6<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0<br />

0 100 200 300 400 500 600 700<br />

Figure 3.17: Growth curve <strong>of</strong> E.coli in ISB<br />

Optical Density at 550 nm<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

Growth Curve <strong>of</strong> Acinetobacter<br />

Time (minutes)<br />

Growth Curve <strong>of</strong> E.coli<br />

0 100 200 300 400 500 600 700<br />

Time/minutes<br />

165


Figure 3.18: Growth curve <strong>of</strong> Klebsiella in ISB<br />

Optical Density at 550 nm<br />

2<br />

1.8<br />

1.6<br />

1.4<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

0 100 200 300 400 500 600 700<br />

Figure 3.19: Growth curve <strong>of</strong> Enterobacter in ISB<br />

Optical Density at 550 nm<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

Growth curve for Klebsiella<br />

Time (minutes)<br />

Growth curve for Enterobacter<br />

0 100 200 300 400 500 600 700<br />

Time (minutes)<br />

166


Figure 3.20: Growth curve <strong>of</strong> Citrobacter in ISB<br />

Optical Density at 550 nm<br />

1.6<br />

1.4<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

0 100 200 300 400 500 600 700<br />

Figure 3.21: Growth curve <strong>of</strong> Serratia in ISB<br />

Optical Density at 550 nm<br />

1.8<br />

1.6<br />

1.4<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

Growth curve for Citrobacter<br />

Time (minutes)<br />

Growth curve for Serratia<br />

0 100 200 300 400 500 600 700<br />

Time(minutes)<br />

167


3.5.2 Scanning Electron Microscopy (SEM):<br />

3.5.2.1 SEM <strong>of</strong> Acinetobacter<br />

For Acinetobacter an average <strong>of</strong> ten SEM micrographs with a magnification <strong>of</strong> 5,000<br />

(for cell count) and two micrographs with a magnification <strong>of</strong> 20,000 (for cellular<br />

details) were taken for each time point. In total 30 SEM micrographs were taken for<br />

untreated <strong>honey</strong> samples (control) and 50 micrographs for treated <strong>honey</strong> samples.<br />

However only representative images for 0, 60, 90, 150 and 180 time points are<br />

shown here.<br />

SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter incubated with broth only at<br />

time 0 were observed to have a regular rod shaped cell with a smooth surface for<br />

95% <strong>of</strong> the cells and cell length w<strong>as</strong> 3 µm (Fig. 3.22); normal cell size is between 1.5<br />

to 2.5 µm (Gillespie and Hawkey 2006). However, after 3 h incubation in liquid<br />

media this bacterium became enlarged and appeared <strong>as</strong> long bacilli (Fig. 3.24, 3.26,<br />

3.28) with mean cell size incre<strong>as</strong>ed to 5.5 µm. This bacterium normally tends to be a<br />

bacillus in shape when it incubated for long time in liquid media and/or if collected<br />

in exponential growth ph<strong>as</strong>e (Murray et al., 2007). Septated cells were also observed<br />

with no surface changes and no debris.<br />

In the <strong>honey</strong> treated sample, the majority <strong>of</strong> cells (94%) appeared unchanged for the<br />

first 30 minutes exposure to manuka <strong>honey</strong> (Fig 3.23). From 60 minutes onwards<br />

lots <strong>of</strong> debris w<strong>as</strong> observed, suggesting that some leakage <strong>of</strong> cellular material or lysis<br />

had taken place. Also clumping and cell aggregation incre<strong>as</strong>ed (Fig. 3.25, 3.27 &<br />

3.29). There were significant changes in the cell length <strong>of</strong> Acinetobacter after 1 h<br />

exposed to 20% (w/v) manuka <strong>honey</strong> compared to untreated <strong>honey</strong> (p = 0.0216)<br />

168


(Table 3.26). The length <strong>of</strong> individual cells treated with <strong>honey</strong> appeared to be shorter<br />

than those without <strong>honey</strong>, which in turn resulted in decre<strong>as</strong>ed cellular volume.<br />

In addition an incre<strong>as</strong>ed number <strong>of</strong> septa were seen after 150 minutes incubation<br />

with 20% (w/v) manuka <strong>honey</strong> (Fig 3.29). Formation <strong>of</strong> elongated cells seemed to<br />

be decre<strong>as</strong>ed (P = 0.025) (Table 3.26) which again would lead to decre<strong>as</strong>e in cellular<br />

volume. An interesting effect seen in this stage w<strong>as</strong> that multiple cell division<br />

constriction sites were apparent on some filaments <strong>of</strong> cells, suggesting that cell<br />

separation had not been completed (Fig 3.29 b, c & d). Also more fragments <strong>of</strong><br />

debris were present in <strong>honey</strong>- treated cells than in untreated cells incubated for the<br />

same time (Fig 3.28 b). A significant change occurred after 3 hrs incubation where<br />

marked damage observed in cell surface. It appeared that treated cells entirely<br />

diffused and lysed (Fig 3.31) compared to control cells.<br />

The statistical analysis <strong>of</strong> morphological changes in untreated and treated <strong>honey</strong> cells<br />

are shown in Table (3.26)<br />

169


Figure 3.22: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after 0 minutes at<br />

x5,000 magnification<br />

Figure 3.23: SEM micrograph <strong>of</strong> Acinetobacter cells exposed to 20% (w/v) manuka<br />

<strong>honey</strong> after 0 minutes at x5,000 magnification<br />

170


Figure 3.24: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after 60 minutes at<br />

x5,000 magnification<br />

Figure 3.25: SEM micrograph <strong>of</strong> Acinetobacter cells exposed to 20% (w/v) manuka<br />

<strong>honey</strong> after 60 minutes at x5,000 magnification<br />

171<br />

Debris<br />

Clumps


Figure 3.26: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after 90 minutes at<br />

x5,000 magnification<br />

Figure 3.27: SEM micrograph <strong>of</strong> Acinetobacter cells exposed to 20% (w/v)<br />

manuka <strong>honey</strong> after 90 minutes at x5,000 magnification<br />

Debris<br />

172<br />

Clumps


Figure 3.28: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after 150 minutes<br />

at x5,000 A & x20,000 B magnification respectively<br />

A<br />

B<br />

Single<br />

division<br />

173


Figure 3.29: SEM micrographs <strong>of</strong> Acinetobacter cells exposed to 20% (w/v)<br />

manuka <strong>honey</strong> after 150 minutes at x5,000A, x10,000B, x20,000C & x25,000D<br />

magnification respectively<br />

A<br />

B<br />

Multiple<br />

division<br />

sites<br />

174<br />

Debris


C<br />

D<br />

Cell division<br />

Multiple septa<br />

in each cell<br />

175


Figure 3.30: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter after 180<br />

minutes at x5,000 A & x10,000 B magnification respectively<br />

A<br />

B<br />

176


Figure 3.31: SEM micrographs <strong>of</strong> Acinetobacter cells exposed to 20% (w/v)<br />

manuka <strong>honey</strong> after 180 minutes at x 5,000 (A) & (B) magnification respectively<br />

A<br />

B<br />

Cell lysis<br />

177


3.5.2.2 SEM <strong>of</strong> E.coli<br />

For E.coli an average <strong>of</strong> four SEM micrographs with a magnification <strong>of</strong> 5,000 (for<br />

cell count) and one micrograph with a magnification <strong>of</strong> 20,000 (for cellular details)<br />

were taken for each time point. In total 20 SEM micrographs were taken for<br />

untreated <strong>honey</strong> samples and 30 micrographs for treated <strong>honey</strong> samples. However,<br />

due to small variation in cell morphology between each time point, only<br />

representative images collected at 30 and 180 time points were shown here.<br />

SEM micrographs <strong>of</strong> control cells <strong>of</strong> E.coli incubated with broth only were shown to<br />

have rods with regular structure and normal size after 3 h incubation in liquid media<br />

(Fig 3.32 & 3.34)<br />

In <strong>honey</strong> treated cells there w<strong>as</strong> no change in shape and size at 0 min (not shown) (P<br />

>0.05). There w<strong>as</strong> no significant difference <strong>of</strong> cell length between treated <strong>honey</strong><br />

sample and untreated one at most <strong>of</strong> the time point (P> 0.05) (Table 3.26). However,<br />

deformation <strong>of</strong> cells w<strong>as</strong> observed after 3 hrs incubation with 30% manuka <strong>honey</strong><br />

(P= 0.025) (Fig 3.35, white arrows). Also it w<strong>as</strong> observed that the cells exposed to<br />

<strong>honey</strong> aggregated together by forming extended strand materials (filament) shape<br />

around each cell probably cytopl<strong>as</strong>mic consistuent (Fig 3.33 & 3.35). Septated cells<br />

were not observed in this stage.<br />

178


Figure 3.32: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> E.coli after 30 minutes at x5,000<br />

magnification<br />

Normal cells<br />

Figure 3.33: SEM micrograph <strong>of</strong> E.coli cells exposed to 30% (w/v) manuka <strong>honey</strong><br />

after 30 minutes at x5,000 magnification<br />

179<br />

Cells with<br />

clumps


Figure 3.34 : SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> E.coli after 180 minutes at<br />

x5,000 magnification<br />

Figure 3.35 : SEM micrograph <strong>of</strong> E.coli cells exposed to 30% (w/v) manuka <strong>honey</strong><br />

after 180 minutes at x5,000 magnification<br />

Filament<br />

shapes<br />

180


3.5.2.3 SEM <strong>of</strong> Klebsiella<br />

Klebsiella w<strong>as</strong> treated with 30% manuka <strong>honey</strong> and 40% Omani <strong>honey</strong> (OH-B). An<br />

average <strong>of</strong> eight SEM micrographs with a magnification <strong>of</strong> 5,000 (for cell count) and<br />

two micrographs with a magnification <strong>of</strong> 15,000 and 20,000 (for cellular details)<br />

were collected for analysis <strong>of</strong> both types <strong>of</strong> <strong>honey</strong> at each time point. In total 20<br />

SEM micrographs were taken for untreated <strong>honey</strong> samples and 60 micrographs for<br />

treated <strong>honey</strong> samples. However, only 30 and 180 time points were shown here.<br />

SEM micrographs <strong>of</strong> Klebsiella incubated with broth alone (no <strong>honey</strong>) were found to<br />

have a conventional rod shape after 3 h incubation in liquid media with approximate<br />

size <strong>of</strong> 0.6 to 6 µm long (Fig 3.36 & 3.37). In SEM micrographs <strong>of</strong> <strong>honey</strong> treated<br />

cells cell size and shape did not appear to be significantly different to untreated cells<br />

after 30 min (Fig 3.38 & 3.40) for both <strong>honey</strong>s (P = > 0.05). However, cell size<br />

significantly incre<strong>as</strong>ed between 1 to 3 hours exposure for both <strong>honey</strong>s (P = < 0.05)<br />

(Table 3.26). In addition rough cell surfaces were observed at the same time points<br />

(Fig 3.39 b,c,d manuka <strong>honey</strong>) and (Fig 3.41 b,c,d Omani <strong>honey</strong>). There few septa<br />

present with no debris observed.<br />

181


Figure 3.36: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Klebsiella after 30 minutes at<br />

x5,000 (A) & 15,000 (B) magnification respectively<br />

A<br />

B<br />

Normal cells<br />

182


Figure 3.37: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Klebsiella after 180 minutes at<br />

x5,000 (A) & 15,000 (B) magnification respectively<br />

A<br />

B<br />

Normal cells<br />

183


Figure 3.38 : SEM micrographs <strong>of</strong> Klebsiella cells exposed to 30% manuka <strong>honey</strong><br />

after 30 minutes at x5,000 (A) & 15,000 (B) magnification respectively<br />

A<br />

B<br />

184


Figure 3.39: SEM micrographs <strong>of</strong> Klebsiella cells exposed to 30% (w/v) manuka<br />

<strong>honey</strong> after 180 minutes at x5,000A, x15,000B,C & x20,000D magnification<br />

A<br />

B<br />

Rough<br />

surfaces<br />

185


C<br />

D<br />

Rough surface<br />

186


Figure 3.40: SEM micrographs <strong>of</strong> Klebsiella cells exposed to 40% (w/v) Omani<br />

<strong>honey</strong> after 30 minutes at x5,000 A & x15,000 B magnification respectively<br />

A<br />

B<br />

187


Figure 3.41 : SEM micrographs <strong>of</strong> Klebsiella cells exposed to 40% (w/v) Omani<br />

<strong>honey</strong> after 180 minutes at x5,000 A & x 20,000 B,C & D magnification respectively<br />

A<br />

B<br />

Rough<br />

surface<br />

188


C<br />

D<br />

189


3.5.2.4 SEM <strong>of</strong> Enterobacter<br />

Similarly for Enterobacter an average <strong>of</strong> four SEM micrographs with a<br />

magnification <strong>of</strong> 5,000 (to do cell counts or cell sizes) and one micrograph with a<br />

magnification <strong>of</strong> 20,000 (for examination <strong>of</strong> cellular surface details) were taken for<br />

each time point. In total 20 SEM micrographs were taken for untreated <strong>honey</strong><br />

samples and 30 micrographs for treated <strong>honey</strong> samples. However, only 30 and 180<br />

time points were shown here.<br />

Again, regular bacilli shaped cells with no surface irregularities were observed in<br />

untreated sample (Fig. 3.42). After exposure to 30% (w/v) manuka <strong>honey</strong> lots <strong>of</strong><br />

mesh like material between cells w<strong>as</strong> obvious <strong>as</strong> a result <strong>of</strong> cells aggregation (Fig.<br />

3.43 & 3.45). There were no significant difference in cell size between untreated and<br />

treated <strong>honey</strong> samples at any time point (P = > 0.05) (Table 3.26).<br />

190


Figure 3.42: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Enterobacter after 30 minutes<br />

at x5,000 A & x15,000 B magnification respectively<br />

A<br />

B<br />

Normal cells<br />

191


Figure 3.43: SEM micrographs <strong>of</strong> Enterobacter cells exposed to 30% (w/v)<br />

manuka <strong>honey</strong> after 30 minutes at x5,000 A & B magnification respectively<br />

A<br />

B<br />

Cells with clumps<br />

192


Figure 3.44: SEM micrographs <strong>of</strong> untreated Enterobacter cells after 180 minutes at<br />

x5,000 A & x20,000 B magnification respectively<br />

A<br />

B<br />

Normal cells<br />

Filter membrane<br />

193


Figure 3.45: SEM micrographs <strong>of</strong> Enterobacter cells exposed to 30% (w/v) manuka<br />

<strong>honey</strong> after 180 minutes at x5,000 (A & B) magnification respectively<br />

A<br />

B<br />

Mesh like structure<br />

194


3.5.2.5 SEM <strong>of</strong> Citrobacter<br />

For Citrobacter an average <strong>of</strong> four SEM micrographs with a magnification <strong>of</strong> 5,000<br />

(for cell count) and one micrograph with a magnification <strong>of</strong> 20,000 (for cellular<br />

details) were taken at each time point. In total 20 SEM micrographs were taken for<br />

untreated <strong>honey</strong> samples and 30 micrographs for treated <strong>honey</strong> samples. However,<br />

only 30 and 180 time points are shown here.<br />

Regular bacilli shaped cells with approximate size 2 to 6 µm long with smooth<br />

surfaces were observed in untreated cells (Fig. 3.46). After exposure to 20% (w/v)<br />

manuka <strong>honey</strong>, the surface <strong>of</strong> bacterial cells seemed to be rougher, maybe <strong>as</strong> a result<br />

<strong>of</strong> outer membrane damage (Fig. 3.47 & 3.49). Also, at higher magnification black<br />

are<strong>as</strong> were present on the surface <strong>of</strong> some cells (Fig. 3.49 b) which w<strong>as</strong> suggestive <strong>of</strong><br />

pore formation. There were significant difference in cell size between untreated and<br />

treated <strong>honey</strong> samples at 30, 90 and 180 minutes time point (P ≤ 0.05) (Table 3.26).<br />

195


Figure 3.46: SEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Citrobacter after 30 minutes at<br />

x5,000 A & x15,000 B magnification respectively<br />

B<br />

A<br />

196<br />

Normal cells


Figure 3.47: SEM micrographs <strong>of</strong> Citrobacter cells exposed to 20% (w/v) manuka<br />

<strong>honey</strong> after 30 minutes at x10,000 A & x15,000 B magnification respectively<br />

A<br />

B<br />

197


Figure 3.48: SEM micrographs <strong>of</strong> untreated Citrobacter cells after 180 minutes at<br />

x5,000 (A & B) magnification respectively<br />

A<br />

B<br />

198


Figure 3.49: SEM micrographs <strong>of</strong> Citrobacter cells exposed to 20% (w/v) manuka<br />

<strong>honey</strong> after 180 minutes at x5,000 A & x15,000 B magnification respectively<br />

A<br />

B<br />

Filter membrane<br />

Pore<br />

formation<br />

199


3.5.2.6 SEM <strong>of</strong> Serratia<br />

For Serratia an average <strong>of</strong> four SEM micrographs with a magnification <strong>of</strong> 5,000 (for<br />

cell count) and one micrograph with a magnification <strong>of</strong> 20,000 (for cellular details)<br />

were taken at each time point. In total 20 SEM micrographs were taken for untreated<br />

<strong>honey</strong> samples and 30 micrographs for treated <strong>honey</strong> samples. However, only 30 and<br />

180 time points are shown here.<br />

Regular bacilli shaped cells with approximate size 0.9 to 2 µm long with smooth<br />

surfaces were observed in untreated sample (Fig. 3.50). After exposure to 30% (w/v)<br />

manuka <strong>honey</strong> for 30 minutes cells surfaces were noticeably uneven and cell debris<br />

seemed to be present, suggesting that some cells had lysed. Blebs were also seen on<br />

some <strong>honey</strong> treated cells (Fig 3.51). After 180 minutes exposure to 30 % (w/v)<br />

manuka <strong>honey</strong>, elongated cells were significantly present (p = 0.0002) with cells<br />

aggregation (Table 3.26). Extracellular material w<strong>as</strong> observed, suggesting that<br />

bi<strong>of</strong>ilm formation had been initiated (Fig. 3.53). Septated cells were not observed at<br />

this stage.<br />

200


Figure 3.50: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Serratia after 30 minutes at<br />

x5,000 magnification<br />

Figure 3.51: SEM micrograph <strong>of</strong> Serratia cells exposed to 30% (w/v) manuka<br />

<strong>honey</strong> for 30 minutes at x 5,000 magnification<br />

Filter membrane<br />

201


Figure 3.52: SEM micrograph <strong>of</strong> untreated cells <strong>of</strong> Serratia after 180 minutes at<br />

x5,000 magnification<br />

Figure 3.53: SEM micrograph <strong>of</strong> Serratia cells exposed to 30% (w/v) manuka<br />

<strong>honey</strong> after 180 minutes at x5,000 magnification<br />

202


Table 3.26: Comparison <strong>of</strong> changes in cell length (μm) <strong>of</strong> isolates observed in scanning electron microscopy between untreated and <strong>honey</strong><br />

treated cells, respectively, after 240 min. (P value


3.5.2 Transmission Electron Microscopy (TEM):<br />

3.5.2.1 TEM for Acinetobacter:<br />

TEM micrographs <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter revealed regularly shaped<br />

cocco-bacilli with entire margins after 60 minutes incubation in ISB (blue arrows)<br />

with a few elongated cells were also seen (Fig 3.54). After 3 hrs cells did not appear<br />

to have major change in cell shape or size (Fig 3.56). However, distinct<br />

morphological changes had occurred in <strong>honey</strong> treated cells after 60 minutes<br />

incubation with 20% (w/v) manuka <strong>honey</strong> in ISB. These changes took place in the<br />

cell membrane, which w<strong>as</strong> observed to be incomplete and indistinct in some cells<br />

(Fig 3.55, red arrows) compared to the well defined cell membrane seen in untreated<br />

cells (Fig 3.54, blue arrows). Moreover, some cells presented with black deposit<br />

possibly mineral and some with empty vacuoles inside some cells (Fig. 3.55). The<br />

presence <strong>of</strong> cellular debris indicated lysis <strong>of</strong> cells (Fig. 3.55 & 3.57). The number <strong>of</strong><br />

cells with septa w<strong>as</strong> incre<strong>as</strong>ed following treatment with 20% (w/v) manuka <strong>honey</strong><br />

(Fig 3.57 black arrows).<br />

204


Figure 3.54: Transmission micrographs <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter<br />

after 1 h incubation with isosensitest broth (ISB) at 16,000x magnification (A, B, C)<br />

Cocco-bacilli shape<br />

B<br />

A<br />

C<br />

205<br />

Define cell wall<br />

300 nm x16,000<br />

300 nm x16,000<br />

300 nm x16,000


Figure 3.55: Transmission micrographs <strong>of</strong> Acinetobacter incubated with isosensitest<br />

borth (ISB) containing 20% (w/v) manuka <strong>honey</strong> for 1 h at 16,000x magnification.<br />

A<br />

Black deposit<br />

B<br />

C<br />

Hole<br />

s<br />

206<br />

Debri<br />

s<br />

300 nm x16,000<br />

300nm x16,000<br />

300nm x16,000


Figure 3.56: Transmission micrographs <strong>of</strong> untreated cells <strong>of</strong> Acinetobacter<br />

incubated with isosensitest broth (ISB) after 3 h at 16,000x(A,B) & 32,000x(C)<br />

B<br />

C<br />

A<br />

207<br />

300nm x16,000<br />

300nm x16,000<br />

300nm x32,000


Figure 3.57: Transmission micrographs <strong>of</strong> Acinetobacter incubated with isosensitest<br />

broth (ISB) containing 20% (w/v) manuka <strong>honey</strong> after 3 h at 16,000x magnification<br />

A<br />

B<br />

C<br />

Septa<br />

208<br />

300nm x16,000<br />

Debris<br />

300nm x16,000<br />

300nm x32,000


3.5.2.2. TEM for E.coli:<br />

TEM images <strong>of</strong> untreated cells <strong>of</strong> E.coli did not appear to show obviously damaged<br />

or abnormal shaped and sized cells. Nevertheless empty spaces or holes were<br />

observed in some cells (Fig 3.58 & 3.60).<br />

Honey-treated cells <strong>of</strong> E.coli with 30% (w/v) manuka <strong>honey</strong> demonstrated gaps<br />

between the cell membrane and the cytopl<strong>as</strong>m which seemed to have caused the<br />

membranes to shrink away from the cytopl<strong>as</strong>m (Fig 3.59). Also deformed cell<br />

membrane could be seen in some images.<br />

Very interestingly, due to membrane rupture the intracellular components had leaked<br />

out from the cells after <strong>honey</strong> treatment and formed empty cells with black deposit<br />

inside (Fig 3.59). After 3 hrs incubation with 30% (w/v) manuka <strong>honey</strong> these<br />

samples also demonstrated cells with irregular shapes with relatively less electron<br />

dense material compared to untreated cells (Fig 3.61). Septated cells were not<br />

observed in this stage.<br />

Unfortunately it w<strong>as</strong> not possible to process all <strong>of</strong> the ESBL organisms for structural<br />

changes induced by <strong>honey</strong>.<br />

209


Figure 3.58: Transmission micrographs <strong>of</strong> untreated cells <strong>of</strong> E.coli after 1 h<br />

incubation with isosensitest borth (ISB) at 16,000x magnification<br />

A<br />

B<br />

C<br />

210<br />

Hole<br />

300nm x16,000<br />

300nm x16,000<br />

300nm x16,000


Figure 3.59: Transmission micrographs <strong>of</strong> E.coli incubated with isosensitest borth<br />

(ISB) containing 20% (w/v) manuka <strong>honey</strong> after 1 h at 16,000x magnification<br />

A<br />

B<br />

C<br />

Lysed cell<br />

Deformed cells<br />

Empty cell with deposit<br />

211<br />

Empty cell with deposit<br />

Empty cell with deposit<br />

300nm x16,000<br />

300nm x16,000<br />

300nm x16,000


Figure 3.60: Transmission micrographs <strong>of</strong> untreated cells <strong>of</strong> E.coli after 3 h<br />

incubation with isosensitest broth (ISB) at 16,000x(A) & 32,000x (B,C).<br />

A<br />

B<br />

C<br />

Holes<br />

212<br />

Define cell wall<br />

300nm x16,000<br />

300nm x32,000<br />

300nm x32,000


Figure 3.61: Transmission micrographs <strong>of</strong> E.coli incubated with isosensitest broth<br />

(ISB) containing 20% (w/v) manuka <strong>honey</strong> after 3 h at 16,000x magnification<br />

B<br />

A<br />

C<br />

Irregular cells<br />

Less electron dense<br />

213<br />

300nm x16,000<br />

300nm x16,000<br />

300nm x16,000


3.6 Effect <strong>of</strong> <strong>honey</strong> on bacterial proteins:<br />

3.6.1 Two Dimensional Gel Electrophoresis:<br />

This technique is able to separate proteins using two distinct steps: the isoelectric<br />

focusing (IEF) separates proteins according to their isoelectric point (pI) and SDS-<br />

polyacrylamide gel electrophoresis (SDS-PAGE) separates proteins according to<br />

their molecular weight (MW). Each resulting spot from a 2-D gel can be matched to<br />

a single protein in the sample. Then it can be analysed using PDQuest B<strong>as</strong>ic 8.0<br />

s<strong>of</strong>tware to determine the number and position <strong>of</strong> spots. Besides obtaining protein<br />

the pI and molecular weight identification, this method detects the amount <strong>of</strong> each<br />

protein present. The proteome analysis <strong>of</strong> Acinetobacter using 2-D electrophoresis<br />

w<strong>as</strong> carried out to establish the differences in protein expression between treated and<br />

untreated <strong>honey</strong> cells. This test w<strong>as</strong> performed in Acinetobacter only because the<br />

electron microscopy results <strong>of</strong> this bacterium were shown more physiological<br />

damage. The s<strong>of</strong>tware that w<strong>as</strong> mention previously w<strong>as</strong> able to detect the number <strong>of</strong><br />

spots present in each test gel (<strong>honey</strong> treatment) (Fig 3.63) and compared it to control<br />

gel (no <strong>honey</strong>) (Fig 3.62). This allows for identifying those spots whether up or<br />

down regulated by comparing their expression between these gels.<br />

A total <strong>of</strong> 10 gels (control & test) were run success<strong>full</strong>y according to the method<br />

described above, but despite repeated attempts, unfortunately the effect on protein<br />

expression within this bacterium after exposure to 20% manuka <strong>honey</strong> w<strong>as</strong> not<br />

possible at this stage. This techniques <strong>of</strong>fers promise in investigating the effects <strong>of</strong><br />

<strong>honey</strong> on ESBLs, but further work in optimising the conditions is needed. It h<strong>as</strong><br />

proved valuable in UWIC in investigating the effect <strong>of</strong> manuka <strong>honey</strong> on MRSA<br />

(Jenkins et al., 2011).<br />

214


Figure 3.62: 2-D protein electrophoresis gel <strong>of</strong> Acinetobacter cells without <strong>honey</strong><br />

treatment<br />

Protein spots<br />

Figure 3.63: 2-D protein electrophoresis gel <strong>of</strong> Acinetobacter cells exposed to 20%<br />

(w/v) manuka <strong>honey</strong><br />

215


Chapter 4<br />

Discussion<br />

216


“The therapeutic potential <strong>of</strong> uncontaminated, pure <strong>honey</strong> is grossly underutilized. It<br />

is widely available in most communities and although the mechanism <strong>of</strong> action <strong>of</strong><br />

several <strong>of</strong> its properties remain obscure and needs further investigation, the time h<strong>as</strong><br />

now come for conventional medicine to lift the blinds <strong>of</strong>f this „traditional remedy‟<br />

and give it its due recognition” (Zumla and Lulat 1989).<br />

This study w<strong>as</strong> carried out to investigate the efficacy <strong>of</strong> Omani <strong>honey</strong> and manuka<br />

<strong>honey</strong> in the inhibition <strong>of</strong> Gram negative multi-drug resistant organisms implicated<br />

in wound infections. The antibacterial potency <strong>of</strong> each <strong>honey</strong> w<strong>as</strong> determined by<br />

bio<strong>as</strong>say method, MIC and MBC, time-kill study and electron microscopy. Each<br />

experiment provided some insight into possible inhibitory mechanisms <strong>of</strong> <strong>honey</strong> on<br />

microorganisms.<br />

4.1 Antibacterial activity <strong>of</strong> <strong>honey</strong> samples:<br />

For many years local, natural and untreated <strong>honey</strong> w<strong>as</strong> previously used in different<br />

countries for their antibacterial activity against different types <strong>of</strong> bacteria<br />

(Alandejani et al., 2009; Al-Jabri et al., 2003; Al-Waili, 2004; B<strong>as</strong>son and Grobler<br />

2008; Wadi et al., 1987).<br />

In order to quantify the antibacterial activity <strong>of</strong> a <strong>honey</strong> using the agar well diffusion<br />

method, a bio<strong>as</strong>say in which activity w<strong>as</strong> compared to phenol <strong>as</strong> a reference w<strong>as</strong><br />

developed in New Zealand (Allen et al., 1991). Phenol is a chemical that w<strong>as</strong> used <strong>as</strong><br />

an antiseptic in surgical procedures in the nineteenth century. It h<strong>as</strong> been used <strong>as</strong> a<br />

reference for the evaluation <strong>of</strong> the potency <strong>of</strong> many antiseptics, but it is not usually<br />

used for wound treatment because <strong>of</strong> its hazardous effect in damaging cells and<br />

tissues. It could be therefore argued that it is not a suitable standard to me<strong>as</strong>ure the<br />

217


activity <strong>of</strong> <strong>honey</strong>, because topical treatments for wounds must be non-cytotoxic, <strong>as</strong><br />

well <strong>as</strong> antibacterial. Nevertheless it h<strong>as</strong> been used in several studies.<br />

A New Zealand study <strong>of</strong> 345 <strong>honey</strong> samples found that non-peroxide activity<br />

presented only in 25 <strong>honey</strong> samples which were identified <strong>as</strong> either manuka or<br />

viper‟s bugloss (Allen et al., 1991). The same bio<strong>as</strong>say w<strong>as</strong> used to survey<br />

antibacterial activity in 139 Welsh <strong>honey</strong>s and it w<strong>as</strong> found that not all <strong>honey</strong>s had a<br />

detectable amount <strong>of</strong> total activity, and non-peroxide activity w<strong>as</strong> not detected<br />

(Wheat 2004). Also, a study <strong>of</strong> 30 Portuguese <strong>honey</strong>s showed only 7 <strong>honey</strong>s<br />

possessed non-peroxide activity with 11.5 % (w/v) phenol equivalent (Henriques et<br />

al., 2005). Twenty years from Allen et al., another New Zealand survey on the<br />

antibacterial activity <strong>of</strong> 477 Australian <strong>honey</strong> samples found that 80 samples (16.8%)<br />

exhibited non-peroxide activity (Irish et al., 2011)<br />

From the bio<strong>as</strong>say most Omani <strong>honey</strong> tested in this <strong>project</strong> had a certain amount <strong>of</strong><br />

antibacterial activity (total activity) which ranged between < 2% and 22.8% (w/v)<br />

phenol equivalent where 2% (w/v) is the lowest phenol standard able to cause a zone<br />

<strong>of</strong> inhibition. However, none <strong>of</strong> them demonstrated detectable activity when tested<br />

for non-peroxide activity, ie incubation with catal<strong>as</strong>e. All the antibacterial activity <strong>of</strong><br />

Omani <strong>honey</strong> on dilution w<strong>as</strong> therefore related to the formation <strong>of</strong> H2O2 and they can<br />

be called peroxide <strong>honey</strong>s. Manuka <strong>honey</strong> used in this study possessed an equal<br />

amount <strong>of</strong> antibacterial activity both total activity with 21.3±0.85 and non-peroxide<br />

activity with 21.4± 0.8 (% w/v) phenol equivalent. Manuka <strong>honey</strong> therefore had non-<br />

peroxide type activity and this confirmed the deduction <strong>of</strong> Allen, Molan and Reid<br />

(1991).<br />

218


Non-peroxide <strong>honey</strong> shown to be effective in wound management. According to<br />

many studies on the antibacterial activity <strong>of</strong> different types <strong>of</strong> <strong>honey</strong>, manuka <strong>honey</strong><br />

h<strong>as</strong> been shown to be the most effective <strong>honey</strong> and because anti-bacterial activity on<br />

dilution w<strong>as</strong> entirely due to the non-peroxide components that are found in this<br />

<strong>honey</strong>, this gives manuka <strong>honey</strong> an advantage over peroxide <strong>honey</strong>s (Al Somal et al.,<br />

1994; Molan and Russell 1988; Snow and Manley-Harris 2003).<br />

There are several rating systems recently developed to describe the quality and<br />

effectiveness <strong>of</strong> non-peroxide <strong>honey</strong> <strong>as</strong> an anti-microbial <strong>agent</strong> for wound treatment.<br />

Unique Manuka Factors (UMF) w<strong>as</strong> the first rating system identified to denote the<br />

potency <strong>of</strong> <strong>honey</strong>. Manuka <strong>honey</strong> with UMF activity <strong>of</strong> ten or above (10 + ) (same<br />

potency <strong>of</strong> 10% phenol equivalent) is the most acceptable for therapeutic use.<br />

Methylglyoxal (MGO) and the Molan Gold Standard are new registered trademark<br />

from different organizations to rate the potency <strong>of</strong> manuka <strong>honey</strong><br />

(http://www.molangoldstandard.co.nz/article).<br />

4.2. Chemical & physical analysis <strong>of</strong> <strong>honey</strong> samples<br />

Different types <strong>of</strong> local <strong>honey</strong> from different origins were studied for chemical<br />

compositions and antioxidant activity (Aljadi and Kamaruddin 2004; Al-Mamary et<br />

al., 2002; Beretta et al.,2005; Estevinho et al., 2008). However, the physicochemical<br />

analysis <strong>of</strong> Omani <strong>honey</strong> including the phenolic acid and anti-radical activity w<strong>as</strong> not<br />

well studied. Determination <strong>of</strong> <strong>honey</strong> compositions and its source <strong>of</strong> origin aid in<br />

identifying its biochemical properties. Omani <strong>honey</strong> w<strong>as</strong> traditionally <strong>as</strong>sessed by<br />

sensory tests such <strong>as</strong> t<strong>as</strong>te, colour, aroma and texture which make the detection <strong>of</strong><br />

<strong>honey</strong> quality more difficult. Generally natural <strong>honey</strong> is variable in its composition<br />

mainly in pH value, <strong>as</strong>h, water and sugar content.<br />

219


The selected Omani <strong>honey</strong>s were analysed for their sugar, water, pH, HMF, colour,<br />

protein, antioxidant and pollen. The values were compared to the reference National<br />

Honey Board (NHB) (Table 4.1) and most parameters were found to be within the<br />

NHB range.<br />

Table 4.1: The physicochemical analysis <strong>of</strong> selected <strong>honey</strong>s tested<br />

Honey Sample pH Water<br />

content<br />

%(w/v)<br />

Sugar<br />

content<br />

% (w/v)<br />

220<br />

Protein<br />

(mg/g)<br />

HMF<br />

(mg/kg)<br />

Anti-<br />

radical<br />

activity%<br />

Phenol<br />

content<br />

(eq/kg)<br />

NHB 3.5-5.5 17.2 82.4 0.7 15 N/A N/A<br />

Mean ±<br />

SD (n) <strong>of</strong><br />

Omani <strong>honey</strong><br />

Manuka<br />

<strong>honey</strong><br />

N/A (not available)<br />

4.7±<br />

0.9<br />

15.8±<br />

1.9<br />

81±<br />

2.8<br />

4.7±<br />

1.5<br />

11.1±<br />

11.9<br />

64.6±<br />

4.1<br />

89.7±<br />

64.8<br />

3.5 20 78 0.3 3 61.1 65.4<br />

To date there h<strong>as</strong> been only one research study that w<strong>as</strong> conducted in Sultan Qaboos<br />

<strong>University</strong> in Oman in which two parameters (sugar fractions and protein content) in<br />

51 Omani <strong>honey</strong>s were analysed. These <strong>honey</strong>s collected from Muscat and Al-Batina<br />

regions during summer and winter se<strong>as</strong>ons between 1999- 2002. The mean sugar<br />

w<strong>as</strong> recorded <strong>as</strong> 67.41% and the mean protein w<strong>as</strong> <strong>as</strong> low <strong>as</strong> 2 ± 1 mg/g. The same<br />

study also compared the level <strong>of</strong> these two factors in summer and winter <strong>honey</strong>s, and<br />

in multifloral and unifloral <strong>honey</strong>s. It w<strong>as</strong> found the level <strong>of</strong> sugars in <strong>honey</strong>s<br />

collected in summer w<strong>as</strong> higher than winter <strong>honey</strong> where<strong>as</strong> no difference in sugar<br />

between multifloral and unifloral <strong>honey</strong>s w<strong>as</strong> found. However the protein content<br />

w<strong>as</strong> not affected by either se<strong>as</strong>on or the type <strong>of</strong> flora (Sajwani et al., 2007b). This<br />

study, therefore, h<strong>as</strong> provided detailed information about selected Omani <strong>honey</strong>s.


Sugars are the major content <strong>of</strong> <strong>honey</strong>, however when these sugars (glucose and<br />

fructose) broken down, the HMF rele<strong>as</strong>ed. An incre<strong>as</strong>e level <strong>of</strong> HMF in <strong>honey</strong><br />

samples occur when <strong>honey</strong> is stored for long periods at high temperature. Using<br />

improper storage temperature or breakdown <strong>of</strong> sugars by acid hydrol<strong>as</strong>e, therefore<br />

rele<strong>as</strong>es high amount <strong>of</strong> HMF which could be used an indicator <strong>of</strong> <strong>honey</strong> quality<br />

(Badawy et al., 2004; Sanz et al., 2003).<br />

Honey is known to possess antioxidant activity which is one <strong>of</strong> main therapeutic<br />

benefits <strong>of</strong> <strong>honey</strong> in trapping the free radicals and promoting wound healing (Molan<br />

1992b). At an injury or wound site the activity <strong>of</strong> free radicals is incre<strong>as</strong>ed. Topical<br />

application <strong>of</strong> <strong>honey</strong> to the injury site is expected to mop up the free radicals and so<br />

promote rapid healing (Subrahmanyam et al., 2003; Henriques et al., 2005). Omani<br />

<strong>honey</strong> exhibited high levels <strong>of</strong> anti-radical activity (64.6± 4.1%) compared to tualang<br />

<strong>honey</strong> (41.3±0.78%) (Mohammed et al., 2010).<br />

Phenolic compounds in <strong>honey</strong> have been considered <strong>as</strong> possible indicators <strong>of</strong> the<br />

botanical origin <strong>of</strong> <strong>honey</strong> (Al-Mamary et al., 2002; Yao et al., 2003).<br />

Honey acts a natural antioxidant and it is known that the colour <strong>of</strong> <strong>honey</strong> is related to<br />

antioxidants. Dark <strong>honey</strong> h<strong>as</strong> been determined to contain more phenolic components<br />

and so more antioxidant potency than the lighter ones (Bogdanov et al., 2004;<br />

Estevinho et al., 2008; Pyrzynska & Biesaga 2009). There are more than 500<br />

polyphenolic compounds already known in <strong>honey</strong> where phenolic acid and<br />

flavonoids are most predominant (Anklam 1998). These compounds are identified by<br />

high performance liquid chromatography and electrophoresis (Pyrzynska & Biesaga<br />

2009).<br />

221


The colour <strong>of</strong> <strong>honey</strong> is the most obvious physical parameter that is observed directly<br />

by users. It varies with botanical origins, age, storage condition and the amount <strong>of</strong><br />

pollen present. The colour can vary from water white, amber brown shade to almost<br />

black. There are two common cl<strong>as</strong>sification systems used for colour grading, the<br />

Pfund and Townsend cl<strong>as</strong>sifications. These systems depend on optical <strong>as</strong>sessment to<br />

obtain the intensity <strong>of</strong> colour (Bogdanov et al., 2004). A new method h<strong>as</strong> been<br />

introduced using reflectance spectroscopy for colour cl<strong>as</strong>sification (Terrab et al.,<br />

2002a)<br />

The colour <strong>of</strong> each Omani <strong>honey</strong> w<strong>as</strong> estimated initially by visual <strong>as</strong>sessment, then it<br />

w<strong>as</strong> tested by determining the optical density <strong>of</strong> 50% (w/v) <strong>honey</strong> at 560 nm and the<br />

colour w<strong>as</strong> cl<strong>as</strong>sified according to Townsend system (Townsend 1969). The results<br />

obtained from visual <strong>as</strong>sessment completely matched the Townsend system. The<br />

colour <strong>of</strong> most <strong>honey</strong> samples ranged from extra light amber to dark amber even<br />

black. This means that Omani <strong>honey</strong>s exhibited a dark colour, which is also visually<br />

obvious in all <strong>honey</strong>s sold in Oman. Elevated level <strong>of</strong> anti-radical activity with<br />

phenolic content and antioxidant in Omani <strong>honey</strong>s highlight the important <strong>of</strong> these<br />

compounds in human health to fight infections and in nutrition <strong>as</strong> a deitery<br />

supplement <strong>of</strong> vitamins.<br />

Eight <strong>honey</strong> samples collected for this <strong>project</strong> were produced in four different<br />

regions <strong>of</strong> Oman. An estimated flora source for each <strong>honey</strong> sample w<strong>as</strong> provided by<br />

the beekeepers. Pollen grains were analysed and <strong>full</strong> identification <strong>of</strong> flora source<br />

w<strong>as</strong> kindly provided from National Pollen And Research Unit (NPARU) at<br />

Worcester <strong>University</strong> (Table 3.9). Five <strong>honey</strong> samples showed that nectar sources<br />

came from mixed flora sources (multi-flora). These are Graminae (the gr<strong>as</strong>ses),<br />

222


Acacia (thorn trees), Myrtacae eucalyptus and Br<strong>as</strong>sica types. However, the<br />

remaining three <strong>honey</strong>s were not identified because <strong>of</strong> little pollen present in the<br />

slides and may have been <strong>honey</strong>dew <strong>honey</strong>s. This demonstrated that the beekeepers<br />

were not always correct in their <strong>as</strong>sumptions about floral origin <strong>of</strong> <strong>honey</strong> due to lack<br />

materials that could used in identifying pollen.<br />

The quality <strong>of</strong> <strong>honey</strong> is b<strong>as</strong>ed on their flora source which is determined by pollen<br />

analysis, and chemical composition. The latter is more accurate and specific in<br />

analysis <strong>of</strong> various components that present in <strong>honey</strong> and it involves identifying the<br />

flora origin <strong>of</strong> <strong>honey</strong>. The price <strong>of</strong> <strong>honey</strong> is depended on its quality.<br />

4.3 Effect <strong>of</strong> <strong>honey</strong> samples against test cultures:<br />

Several reports have been published regarding the <strong>antimicrobial</strong> activity <strong>of</strong> different<br />

types <strong>of</strong> <strong>honey</strong> against variety <strong>of</strong> organisms including Staphylococcus (MRSA &<br />

MSSA), Pseudomon<strong>as</strong> aeruginosa, Streptococcus and anaerobes (Cooper et al.,<br />

1999; Maedaa et al., 2008; Mullai and Menon 2005, 2007) but little w<strong>as</strong> documented<br />

for Gram negative Enterobacteriaceae especially MDR and ESBL producing<br />

organisms before this study. Here the antibacterial activity <strong>of</strong> five types <strong>of</strong> selected<br />

<strong>honey</strong> w<strong>as</strong> tested against six species <strong>of</strong> MDR organisms that usually cause wound<br />

infections. These were Acinetobacter, E.coli, Klebsiella, Citrobacter, Enterobacter<br />

and Serratia. Eighty seven isolates in total were tested for MIC and MBC against<br />

these five types <strong>of</strong> <strong>honey</strong> including manuka <strong>honey</strong>.<br />

MIC is a me<strong>as</strong>urement <strong>of</strong> the quantity <strong>of</strong> <strong>honey</strong> required for bacterial inhibition. To<br />

determine the MIC <strong>of</strong> <strong>honey</strong> samples against selected bacteria two methods were<br />

223


used: agar incorporation method and broth dilution method using 96 well microtiter<br />

plates. The results in latter method are determined by visual inspection <strong>of</strong> turbidity<br />

and confirmed by using spectrophotometric <strong>as</strong>say. Initially agar incorporation w<strong>as</strong><br />

used for 10 Acinetobacter isolates against manuka <strong>honey</strong>. The mean MIC results<br />

showed 7.18 (%w/v) compared to 7.17 (%w/v) using broth dilution methods.<br />

The nature <strong>of</strong> agar incorporation method is b<strong>as</strong>ic but it h<strong>as</strong> some limitations in the<br />

results <strong>as</strong> it does not report MBC values and limits the number <strong>of</strong> test replicates.<br />

Also it is time consuming, required large amount <strong>of</strong> samples and extensive plate<br />

preparation w<strong>as</strong> also needed. There were no differences in the results between two<br />

techniques though the broth dilution technique presents many advantages including:<br />

smaller volume <strong>of</strong> samples used, rapid and cheaper, allows more replicates in the<br />

same plate, produce large amount <strong>of</strong> statistical data and determines the MBC value<br />

(Patton et al., 2006). Recently, a microtiter plate <strong>as</strong>say h<strong>as</strong> been widely used in<br />

antibiotic testing and bioactivity (C<strong>as</strong>ey et al., 2004; Kuda et al., 2004). Thus<br />

because <strong>of</strong> these advantages, the broth dilution method w<strong>as</strong> used for all other isolates<br />

instead.<br />

The bactericidal activity <strong>of</strong> any <strong>antimicrobial</strong> <strong>agent</strong> is usually more desirable than<br />

<strong>agent</strong>s with bacteriostatic activity. The <strong>antimicrobial</strong> <strong>agent</strong> considered to have a<br />

bactericidal action is when MIC/MBC ratio < 4 while the bacteriostatic action is<br />

when this ratio become >4 (Levison 2004). From the MIC and MBC results it w<strong>as</strong><br />

observed that all <strong>honey</strong>s exhibit bactericidal mode <strong>of</strong> action against all isolates tested<br />

in this <strong>project</strong>. Different <strong>honey</strong> reacts differently on each isolate and at different<br />

concentrations. It is therefore difficult to determine which <strong>honey</strong> h<strong>as</strong> a more potent<br />

activity than others. However, manuka <strong>honey</strong> remained the most effective against<br />

224


major wound pathogens, consistently giving lower MIC values for all <strong>of</strong> the test<br />

organisms.<br />

According to the summarized charts below (Fig. 4.1- 4.6) Acinetobacter isolates<br />

were more sensitive (low MIC recorded) to 3 types <strong>of</strong> <strong>honey</strong> (manuka <strong>honey</strong>, and<br />

Omani <strong>honey</strong>s B & G) with MICs <strong>of</strong> 7.17, 16.6 and 20.5 (%w/v) respectively. The<br />

remaining two Omani <strong>honey</strong>s (C & F) with MICs <strong>of</strong> 15.8 and 16.6 (%w/v)<br />

respectively were more active against Citrobacter. Serratia cultures were less<br />

susceptible to most <strong>of</strong> the <strong>honey</strong>s tested with MICs ranging between 13.3-28<br />

(%w/v).<br />

As previously mentioned the <strong>antimicrobial</strong> activity <strong>of</strong> <strong>honey</strong> against MDR Gram<br />

negative bacilli w<strong>as</strong> not well documented in the medical literature. However, in some<br />

circumstances the MIC values determined from this study were comparable to the<br />

MICs reported earlier (Table 4.2). However, comparisons were limited by variations<br />

in methodology and the <strong>honey</strong>s utilised.<br />

In Oman <strong>honey</strong> h<strong>as</strong> been used for thousand <strong>of</strong> years in the treatment <strong>of</strong> many<br />

dise<strong>as</strong>es. In this study only small number <strong>of</strong> <strong>honey</strong> samples were tested and this does<br />

not account for all <strong>of</strong> the types present in Oman. Few studies <strong>of</strong> Omani <strong>honey</strong> were<br />

reported on their antibacterial activity. It w<strong>as</strong> documented that Omani <strong>honey</strong> h<strong>as</strong> a<br />

broad spectrum activity against Staphylococcus aureus, E. coli and P. aeruginosa<br />

(Al-Jabri et al., 2003, 2005b). In addition the combination between <strong>honey</strong> and<br />

antibiotics w<strong>as</strong> first initiated by Al-Jabri et al., (2005a) who found that Omani <strong>honey</strong><br />

had a synergistic effect with aminoglycosides. However, analysis <strong>of</strong> Omani <strong>honey</strong>s<br />

needs to be more extended by collecting larger number <strong>of</strong> samples, and determing<br />

the effect <strong>of</strong> <strong>honey</strong> on other <strong>antimicrobial</strong> groups.<br />

225


Table 4.2: Comparison between previous studies and current study on MIC <strong>of</strong> different <strong>honey</strong>s including manuka <strong>honey</strong> against six bacteria<br />

species.<br />

Acinetobacter<br />

MIC % (w/v) previous studies MIC % (w/v) this study Comments<br />

Manuka<br />

<strong>honey</strong><br />

Other<br />

<strong>honey</strong><br />

Authors Manuka<br />

<strong>honey</strong><br />

226<br />

Omani <strong>honey</strong> (OH)<br />

OH-B OH-C OH-D OH-F<br />

N/A 8.1±1.5 (Blair et al., 2009) 7.17± 0.7 16.6± 2 18.1± 1.6 20.5± 2.4 17.6± 1.9 Omani <strong>honey</strong> h<strong>as</strong><br />

higher MIC than<br />

12.5 11.25 (Tan et al., 2009)<br />

medi<strong>honey</strong>, tualang<br />

and some Australian<br />

N/A 6-8 (George & Cutting 2007)<br />

<strong>honey</strong><br />

*<br />

E.coli N/A 7.5±0.8 (Blair et al., 2009) 10.4± 1.5 18.8±<br />

1.3<br />

17.5 22.5 (Tan et al., 2009)<br />

10 20 (Lusby et al., 2005)<br />

12.5 12.5 (Sherlock et al., 2010) *<br />

N/A 6-8 (George & Cutting 2007) *<br />

19.4±1 23.6±2 19.6±1.8 OH h<strong>as</strong> lower MIC<br />

than tualang and<br />

some Australian<br />

<strong>honey</strong><br />

Klebsiella N/A 13±2.4 (Blair et al., 2009) 11.7±1 .3 17.2±3 18.5±1.7 22± 2 18± 1.9 OH h<strong>as</strong> lower MIC<br />

than some Australian<br />


MIC % (w/v) Previous studies MIC % (w/v) this study Comments<br />

Manuka<br />

<strong>honey</strong><br />

Other<br />

<strong>honey</strong><br />

Authors Manuka<br />

<strong>honey</strong><br />

227<br />

Omani <strong>honey</strong> (OH)<br />

OH-B OH-C OH-D OH-F<br />

Citrobacter N/A 9.1±3 (Blair et al., 2009) 9.7± 0.8 18.1±2.6 15.8±2.1 26 ± 2.5 16.1± 2.3 OH h<strong>as</strong> lower MIC<br />

than some Australian<br />

10 20 (Lusby et al., 2005)<br />

<strong>honey</strong><br />

Enterobacter N/A 11.7±1.8 (Blair et al., 2009) 10.1± 1.7 21.6±0.9 20.1±1.7 26.4±2.9 20±1.8 OH h<strong>as</strong> same activity<br />

<strong>as</strong> Australian <strong>honey</strong><br />

20 25 (Tan et al., 2009)<br />

and lower MIC than<br />

tualang <strong>honey</strong> but<br />

10 20 (Lusby et al., 2005)<br />

higher than medi<strong>honey</strong><br />

N/A 6 (George & Cutting<br />

2007) *<br />

Serratia N/A 14.8±0.5 (Blair et al., 2009) 13.3± 1.7 20.2± 1.9 20.2± 1.2 >28 17± 2.8 OH h<strong>as</strong> higher MIC<br />

than medi<strong>honey</strong><br />

0 0 (Lusby et al., 2005)<br />

N/A (not available)<br />

Blair et al., 2009 used medi<strong>honey</strong> (Australia) by agar incorporation method (%w/v), Tan et al., 2009 used local tualang <strong>honey</strong> (Malaysian) by broth dilution<br />

method (%w/v), Lusby et al., 2005 used some Australian <strong>honey</strong> by agar incorporation (%w/v), Sherlock et al., 2010 used manuka and ulmo <strong>honey</strong> by<br />

microtitire plate (% v/v). George & Cutting 2007 used medi<strong>honey</strong> (Australia) by agar incorporation method (%v/v).


Figure 4.1: Mean MIC and MBC (%w/v) for 30 Acinetobacter strain against 5 types<br />

<strong>of</strong> <strong>honey</strong><br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

7.17<br />

11<br />

18.4<br />

16.6<br />

19.2<br />

18.1<br />

Figure 4.2: Mean MIC and MBC (%w/v) for 12 Klebsiella strain against 5 types <strong>of</strong><br />

<strong>honey</strong><br />

% bacterial inhibition<br />

% bacterial inhibition<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Susceptibility <strong>of</strong> 30 Acinetobacter strains against<br />

5 types <strong>of</strong> <strong>honey</strong><br />

228<br />

21.8<br />

20.5<br />

17.6 17.8<br />

MH-A OH-B OH-C OH-G OH-F<br />

12.9<br />

11.7<br />

Honey types<br />

Mean MIC (W/V%) Mean MBC (W/V%)<br />

Susceptibility <strong>of</strong>12 Klebsiella strains against 5<br />

types <strong>of</strong> <strong>honey</strong><br />

18.8<br />

17.2<br />

18.5<br />

19.2<br />

MH- A OH-B OH-C OH-G OH-F<br />

Honey types<br />

22<br />

Mean MIC (W/V%) Mean MBC (W/V%)<br />

25<br />

18<br />

20.8


Figure 4.3: Mean MIC and MBC (%w/v) for 10 E.coli strain against 5 types <strong>of</strong><br />

<strong>honey</strong><br />

% bacterial inhibition<br />

10.4 10.9<br />

19.8<br />

18.8<br />

19.4<br />

Figure 4.4: Mean MIC and MBC (%w/v) for 12 Citrobacter strain against 5 types <strong>of</strong><br />

<strong>honey</strong><br />

% bacterial inhibition<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Susceptibility <strong>of</strong> 10 E.coli strains against 5 types<br />

<strong>of</strong> <strong>honey</strong><br />

229<br />

20<br />

23.6<br />

25<br />

20.8<br />

19.6<br />

MH- A OH-B OH-C OH-G OH-F<br />

9.7<br />

13.3<br />

Honey type<br />

Mean MIC (W/V%) Mean MBC (W/V%)<br />

Susceptibility <strong>of</strong> 12 Citrobacter strains<br />

against 5 types <strong>of</strong> <strong>honey</strong><br />

18.1 18.5 18.3<br />

15.8<br />

26<br />

27.5<br />

17.8<br />

16.1<br />

MH- A OH-B OH-C OH-G OH-F<br />

Honey type<br />

Mean MIC (W/V%) Mean MBC (W/V%)


Figure 4.5 Mean MIC and MBC (%w/v) for 15 Enterobacter strain against 5 types<br />

<strong>of</strong> <strong>honey</strong><br />

10.1<br />

13.1<br />

22.9<br />

21.6<br />

22.2<br />

20.1<br />

Figure 4.6: Mean MIC and MBC (%w/v) for 8 Serratia strain against 5 types <strong>of</strong><br />

<strong>honey</strong><br />

% bacterial inhibition<br />

% bacterial inhibition<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Susceptibility <strong>of</strong> 15 Enterobacter strains against<br />

5 types <strong>of</strong> <strong>honey</strong><br />

13.3<br />

230<br />

26.4<br />

MH- A OH-B OH-C OH-G OH-F<br />

17<br />

Honey type<br />

Mean MIC (W/V%) Mean MBC (W/V%)<br />

Susceptibility <strong>of</strong> 8 Serratia strains against 5<br />

types <strong>of</strong> <strong>honey</strong><br />

23.75<br />

21.7<br />

20.2 20.2<br />

MH- A OH-B OH-C OH-G OH-F<br />

Honey type<br />

28<br />

28<br />

28<br />

Mean MIC (W/V%) Mean MBC (W/V%)<br />

20<br />

17<br />

22.5<br />

23.7


4.4 Inhibition <strong>of</strong> test organisms by manuka <strong>honey</strong><br />

using time kill curve <strong>as</strong>say<br />

Time-kill curve studies are used to me<strong>as</strong>ure the killing rate <strong>of</strong> organism by<br />

antibiotics using time and concentration. Total cell count is define <strong>as</strong> the total<br />

number <strong>of</strong> both dead and living cells in the a sample, where<strong>as</strong> total viable count<br />

(TVC) is define <strong>as</strong> the number <strong>of</strong> living cells (Singleton 2004).<br />

A T test analysis w<strong>as</strong> applied to establish the statistical difference in cell count<br />

between <strong>honey</strong> treated cells and untreated cells in the selected species under study.<br />

P values <strong>of</strong> less than 0.05 were considered to be statistically significant.<br />

A bactericidal activity is defined <strong>as</strong> in vitro activity <strong>of</strong> 3 log reduction in the cfu/ml<br />

or 99.9% killing over a specific period <strong>of</strong> time (Shriv<strong>as</strong>tava et al., 2009). Killing<br />

me<strong>as</strong>urement w<strong>as</strong> made in this study by the actual decre<strong>as</strong>e in viable counts at 5 h<br />

for each species.<br />

To maintain and minimize the impact <strong>of</strong> time- kill variables on test results several<br />

factors should be considered when performing time-kill studies. These variations<br />

affect the results <strong>of</strong> the <strong>as</strong>say and/or interpretation <strong>of</strong> the results. These factors are:<br />

first, the initial or starting inoculum <strong>of</strong> (10 4 -10 6 cfu/ml) should be applied. Second,<br />

the samples should be incubated at 37°C in shaking water bath. Third, the <strong>as</strong>say<br />

should be continued up to 24 h (Klepser et al., 1998). In this study all these<br />

conditions were applied in the time-kill <strong>as</strong>says and there were significant differences<br />

between treated and untreated <strong>honey</strong> samples (P


Addition <strong>of</strong> <strong>honey</strong> to an exponential ph<strong>as</strong>e culture leads to an obvious reduction for<br />

bactericidal population over a period <strong>of</strong> time (Fig. 3.10 to 3.15). E.coli and Serratia<br />

showed reductions in population <strong>of</strong> 1.97 and 1.87 log10 cfu/ml respectively (99%<br />

growth inhibition) within 5 h. However, Citrobacter w<strong>as</strong> inhibited f<strong>as</strong>ter because it<br />

exhibited the highest reduction in population than other species tested within 5 h<br />

with > 99.9% killing rate (it exceeded 3 log10 cfu/ml reduction). The initial<br />

concentration <strong>of</strong> Acinetobacter and Enterobacter were 7 and 7.74 log10 cfu/ml and<br />

were reduced to 5.6 and 6.17 log10 cfu/ml respectively within 5 h exposure to<br />

manuka <strong>honey</strong>. These isolates were considered to be the le<strong>as</strong>t susceptible species<br />

among those tested here (Table 3.24). This study h<strong>as</strong> therefore shown that manuka<br />

<strong>honey</strong> exhibited bactericidal activity within 5 hours.<br />

4.5 Effect <strong>of</strong> <strong>honey</strong> on bacterial structure:<br />

Shapes <strong>of</strong> various bacteria can be observed by light microscopy, including cocci,<br />

rods, spiral or cubes. The development <strong>of</strong> electron microscopes provides new<br />

insights into bacterial ultr<strong>as</strong>tructural studies and bacterial organisation. Scanning<br />

electron microscopy (SEM) provides a three-dimensional view <strong>of</strong> cellular structures<br />

and information about their surface topography. Transmission electron microscopy<br />

(TEM) examines the external and internal bacterial structures (Hobot 2002). Details<br />

<strong>of</strong> surface structures and bacterial morphology aid to study the action <strong>of</strong> various<br />

drugs on bacteria.<br />

The mechanism <strong>of</strong> <strong>antimicrobial</strong> effects <strong>of</strong> <strong>honey</strong> is not yet <strong>full</strong>y understood. The<br />

effects <strong>of</strong> <strong>honey</strong> on bacteria could be complicated because <strong>of</strong> the complexity <strong>of</strong><br />

232


<strong>honey</strong> chemistry. Nevertheless, observation <strong>of</strong> the bacterial structures and<br />

morphological variation present valuable knowledge <strong>of</strong> complete understanding <strong>of</strong><br />

<strong>antimicrobial</strong> action <strong>of</strong> <strong>honey</strong> on bacteria.<br />

From the EM images <strong>of</strong> the selected isolates all species undergo morphological<br />

changes after exposure to <strong>honey</strong>, however the nature <strong>of</strong> changes seen w<strong>as</strong> different in<br />

each species. Primary research on the action <strong>of</strong> manuka <strong>honey</strong> on Gram positive and<br />

negative bacteria using electron microscopy sequentially identifying the mode <strong>of</strong><br />

action and the target site w<strong>as</strong> initiated by research team at UWIC. Manuka <strong>honey</strong><br />

with Staphylococcus aureus affected the cell division process by the production <strong>of</strong><br />

septated cells which were unable to complete cell division (Henriques et al., 2009).<br />

However, cell destruction and lysis were observed in Pseudomon<strong>as</strong> aeruginosa<br />

which affected the structure <strong>of</strong> the cell wall (Henriques et al., 2009, 2010). It w<strong>as</strong><br />

therefore documented that both species responded differently to certain<br />

concentration <strong>of</strong> manuka <strong>honey</strong>.<br />

Typical prokaryotic bacterial structure consists <strong>of</strong> a cell wall, cytopl<strong>as</strong>mic<br />

membrane, ribosomes, inclusions, chromosome, pl<strong>as</strong>mids, pili or fimbrie, and<br />

possibly a capsule. Each structure h<strong>as</strong> a specific function in regulating nutrition and<br />

protection processes (Hobot 2002). Due to previous observations on Pseudomon<strong>as</strong><br />

aeruginosa which demonstrated that the cell surface w<strong>as</strong> markedly changed by<br />

exposure to <strong>honey</strong> (Henriques et al., 2010), it seemed logical to concentrate on the<br />

outer layer <strong>of</strong> the Gram negative bacteria studied here, especially the cell wall and<br />

cytopl<strong>as</strong>mic membrane.<br />

The cell wall <strong>of</strong> Gram negative bacteria is important for maintaining the bacterial<br />

shape. It allows for the p<strong>as</strong>sage <strong>of</strong> the macromolecules from the outside environment<br />

233


to the cytopl<strong>as</strong>mic membrane to the cytopl<strong>as</strong>m. If the cell wall is ruptured or<br />

damaged, the cytopl<strong>as</strong>mic contents may be lost and eventually cause lysis <strong>of</strong> bacteria<br />

(Sussman 2002). The cell wall consists <strong>of</strong> an outer membrane layer and inner<br />

peptidoglycan layer. The important component <strong>of</strong> the outer membrane is a group <strong>of</strong><br />

proteins referred to <strong>as</strong> porins that permit the p<strong>as</strong>sage <strong>of</strong> small macromolecules, such<br />

<strong>as</strong> glucose or maltose to the cell interior. These porins can open or close their pores<br />

under cellular control. The peptidoglycan (murein) is responsible for maintaining<br />

the cell shape and confers rigidity. It also prevents the osmotic pressure <strong>of</strong> the<br />

cytopl<strong>as</strong>m from bursting the cell and is involved in cell division (Konig et al., 2010).<br />

The structure <strong>of</strong> bacterial cells exposed to inhibitory concentrations <strong>of</strong> manuka<br />

<strong>honey</strong> (2x MIC values) w<strong>as</strong> compared to untreated cells using scanning electron<br />

microscopy. The cells were collected in exponential growth ph<strong>as</strong>e where the cell<br />

growth are f<strong>as</strong>t and most <strong>of</strong> the proteins and enzymes are produced in that ph<strong>as</strong>e<br />

(Madigan et al., 2009) which could be an indication <strong>of</strong> target site <strong>of</strong> the<br />

<strong>antimicrobial</strong> action <strong>of</strong> <strong>honey</strong>.<br />

Images <strong>of</strong> treated Acinetobacter suggested incre<strong>as</strong>ed cellular debris which is an<br />

indication <strong>of</strong> cell lyses. However, the most remarkable change w<strong>as</strong> incre<strong>as</strong>ed<br />

formation <strong>of</strong> constrictions suggesting septa in the individual cell (four septa in each<br />

cell) (Fig 3.29). Instead <strong>of</strong> cells dividing by binary fission they seemed to undergo<br />

multiple fission without cell separation, which normally only occurs in eukaryotic<br />

cells. It could be the cells attempt to adapt to the high exposure <strong>of</strong> osmotic<br />

concentration <strong>as</strong> a kind <strong>of</strong> stress response. However, after 1 h exposure to <strong>honey</strong><br />

onward the cells become significantly shorter compared to untreated cells which led<br />

to a decre<strong>as</strong>e in cellular volume (p = 0.0216).<br />

234


In TEM, the cell membrane w<strong>as</strong> interrupted and septated cells were also observed<br />

with <strong>honey</strong>. The presence <strong>of</strong> dark are<strong>as</strong> in some cells that could be mineral deposit<br />

also evidences cells abnormalities (Fig 3.55). In hypertonic solution (in this c<strong>as</strong>e<br />

<strong>honey</strong>) water molecules move from low concentration (inside bacteria) to high<br />

concentration and this results in decre<strong>as</strong>ed internal pressure and shrinkage <strong>of</strong> cells. In<br />

this c<strong>as</strong>e the high sugar concentration which is believed to aid the <strong>antimicrobial</strong><br />

effect <strong>of</strong> <strong>honey</strong> causes noticeable membrane damage. In addition the minimum pH<br />

limit for most organisms to be viable is pH 5. pH value below this level may inhibit<br />

the metabolism <strong>of</strong> most enzymes, alter fatty acids that responsible for cell wall<br />

synthesis and protein denaturation may also occur which consequently leads to cell<br />

damage (Booth 1985; Cotter and Hill 2003). This could reduce the size <strong>of</strong> bacterial<br />

cells and lead to decre<strong>as</strong>e cell volume. This may have been the re<strong>as</strong>on why a<br />

significant decre<strong>as</strong>e in cells size <strong>of</strong> Acinetobacter from 1 to 3 h exposed to 20%<br />

(w/v) manuka <strong>honey</strong> compared to untreated <strong>honey</strong> (Table 3. 26) w<strong>as</strong> noted.<br />

In normal division the size and volume <strong>of</strong> cells are incre<strong>as</strong>ed before cell separate to<br />

form new daughter cells but the opposite appears to happen here. Acinetobacter cells<br />

in cultures exposed to <strong>honey</strong> were shortened and filamented, suggesting that<br />

incomplete cell division had occurred. The total number <strong>of</strong> culturable cells decre<strong>as</strong>ed<br />

immediately after exposure to <strong>honey</strong>. This w<strong>as</strong> also confirmed by the data from time<br />

to kill curves experiments (Fig 3.10).<br />

In SEM images <strong>of</strong> E.coli treated cells, aggregation <strong>of</strong> cells is seen with strands <strong>of</strong><br />

material extending from <strong>honey</strong> treated cells (Fig 3.35). The most noticeable change<br />

w<strong>as</strong> seen in TEM images where the intracellular components leaked out from the<br />

<strong>honey</strong> treated cells (cell lysed) due to loss <strong>of</strong> membrane integrity and formed empty<br />

235


cells (Fig 3.59). Some treated cells also demonstrated irregular shapes with relatively<br />

less electron dense material compared to untreated cells and the cytopl<strong>as</strong>mic material<br />

had contracted away from the cell boundaries. All <strong>of</strong> these changes support the<br />

suggestion <strong>of</strong> an effect <strong>of</strong> <strong>honey</strong> on bacterial cell membrane <strong>as</strong> a target site in E.coli.<br />

Lysis <strong>of</strong> cells due to alteration in membrane integrity prevents bacterial growth and<br />

multiplication and consequently inhibits normal cells division (Cotter and Hill 2003).<br />

Also, by supporting the results from time kill study, the total number <strong>of</strong> cells were<br />

decre<strong>as</strong>ed <strong>as</strong> shown by growth curve (Fig 3.5) and number <strong>of</strong> culturable cells were<br />

also decre<strong>as</strong>ed (Fig 3.11). Septated cells were not observed in either SEM or TEM<br />

images. However, it does not mean that it w<strong>as</strong> not present, it w<strong>as</strong> possible that it<br />

might have been failed to be spotted through the ultrathin sectioning or due to<br />

improper orientation <strong>of</strong> the section which reduce the chance <strong>of</strong> septum location.<br />

However, cells <strong>of</strong> Klebsiella treated with manuka and Omani <strong>honey</strong> had a noticeably<br />

rougher cell surface, compared to untreated cells (Fig 3.36). In addition, changes in<br />

cells size significantly incre<strong>as</strong>ed for both types <strong>of</strong> <strong>honey</strong>s (p= < 0.05). This suggests<br />

that outer cell surfaces might have been affected by <strong>honey</strong> (Fig 3.39b,c,d, & Fig<br />

3.41b,c,d)<br />

Using SEM with Citrobacter exposed to 20% manuka <strong>honey</strong> revealed a significant<br />

change in cell size between untreated and treated <strong>honey</strong> samples at 30, 90 and 180<br />

minutes time point (p = ≤ 0.05). In addition, the surface <strong>of</strong> bacterial cells becomes<br />

rougher, perhaps <strong>as</strong> a result <strong>of</strong> outer membrane damage (Fig 3.47). The possibility <strong>of</strong><br />

formation <strong>of</strong> pore w<strong>as</strong> observed by black are<strong>as</strong> in the cell surface (Fig 3.49b). This<br />

indicates that <strong>honey</strong> could enter the cells though that pore and cause cell destruction.<br />

The total number <strong>of</strong> culturable cells showed marked decre<strong>as</strong>e in colony forming unit<br />

236


with more than (3-log10 reduction). This means 99.9% <strong>of</strong> the Citrobacter population<br />

were killed. Citrobacter therefore gave the greatest bactericidal response compared<br />

to the other species tested (Table 3.24).<br />

SEM images <strong>of</strong> Enterobacter showed incre<strong>as</strong>ed formation <strong>of</strong> a net like structure in<br />

<strong>honey</strong> treated cells after 30 minutes <strong>of</strong> exposure compared to the background <strong>of</strong> the<br />

untreated cells (Fig 3.45). This could be formed from lysed cells or from leaked<br />

cellular products. Thus comparable to expectation from time- kill curve (Fig 3.14),<br />

the total number <strong>of</strong> culturable cell count decre<strong>as</strong>ed immediately with time after<br />

exposure to <strong>honey</strong>. There were no significant difference in cell size between<br />

untreated and treated <strong>honey</strong> samples at each time point tested (P > 0.05) (Table 3.25)<br />

Serratia treated with 30% manuka <strong>honey</strong> revealed some changes in cell surface and<br />

incre<strong>as</strong>ed incidence <strong>of</strong> cellular debris <strong>as</strong> a result <strong>of</strong> cell lyses (Fig 3.51). Incre<strong>as</strong>ed<br />

elongated cells w<strong>as</strong> significant (P = 0.0002) (Fig 3.53). No septa were observed in<br />

this stage.<br />

237


Table 4.3: Summary <strong>of</strong> the growth inhibition, killing rate and ultra-structure changes in EM for six species selected after exposure to 2x MIC<br />

(%w/v) <strong>of</strong> manuka <strong>honey</strong>:<br />

Honey<br />

Conc.<br />

2xMIC<br />

Growth<br />

inhibition<br />

by MIC<br />

% (w/v)<br />

Killing<br />

rate by<br />

time-kill *<br />

Site <strong>of</strong> cell change Septa<br />

formation<br />

Acinetobacter 20% 9 6 Shorter cells with<br />

clumps<br />

E.coli 30% 13 2 Extended strand<br />

materials around the<br />

cells<br />

238<br />

SEM TEM<br />

Debris (cell<br />

lysed)<br />

High High loss <strong>of</strong> cell membrane, cellular debris,<br />

septated cells and black deposit<br />

Not seen Not seen Excess loss <strong>of</strong> membrane integrity,<br />

irregular shape, empty cells and less<br />

electron dense<br />

Klebsiella 30% 13 4 Cell size, rough surface Few Not seen Not done<br />

Citrobacter 20% 11 1 Cell size, rough<br />

surface, black deposit<br />

Not seen Not seen Not done<br />

Enterobacter 30% 13 5 No change Few High Not done<br />

Serratia 30% 15 3 Cell surface & size Not seen Moderate Not done<br />

Killing rate by time-kill curve * (1-6) = (1) killing at f<strong>as</strong>ter rate, (6) slowest killing rate


To date there is little research on the effect <strong>of</strong> <strong>honey</strong> on multidrug resistant<br />

Acinetobatcter and Enterobacteriaceae in general. However, no medical research w<strong>as</strong><br />

conducted up to electron microscope level to find out the ultra-structural change <strong>of</strong><br />

these species against <strong>honey</strong> using EM in particular. It is difficult therefore to prove<br />

the mechanism <strong>of</strong> the inhibitory effects <strong>of</strong> <strong>honey</strong> on bacteria using this approach<br />

alone. A proteomic approach is likely to give more useful information on the<br />

proteins that are affected by <strong>honey</strong>, but there w<strong>as</strong> insufficient time to develop the<br />

protocols in this study. However, in manuka <strong>honey</strong> cellular protein extraction <strong>of</strong><br />

Gram positive cocci (Staph aureus) w<strong>as</strong> recently studied using 2-D electrophoresis.<br />

The cells showed down regulation in protein synthesis after exposure to 10% (w/v)<br />

manuka <strong>honey</strong>. The protein spot w<strong>as</strong> identified using MALDI-TOF-MS techniques<br />

<strong>as</strong> universal stress protein A (Jenkins et al., 2011). This will give an expectation on<br />

the different proteomic pr<strong>of</strong>ile <strong>of</strong> <strong>honey</strong> treated Gram negative bacteria. That same<br />

research on the mechanisms <strong>of</strong> action <strong>of</strong> manuka <strong>honey</strong> on MRSA stated that the<br />

manuka <strong>honey</strong> affected the process <strong>of</strong> cell division during septa formation. It w<strong>as</strong><br />

recognised that decre<strong>as</strong>ed level <strong>of</strong> FtsZ enzyme, which is involved in cell division,<br />

leads to disruption <strong>of</strong> the cell cycle after <strong>honey</strong> treatment and caused cell death<br />

(Jenkins et al., 2011).<br />

Nonetheless, microarrays would also give valuable information on how gene<br />

expression in Gram negative bacteria is affected by <strong>honey</strong>.<br />

Some researchers suggested that the <strong>antimicrobial</strong> activity w<strong>as</strong> due to the presence <strong>of</strong><br />

high amount <strong>of</strong> tetracycline derivatives, peroxid<strong>as</strong>es, phenols, amyl<strong>as</strong>es, <strong>as</strong>corbic<br />

acid and fatty acid (Nzeako and Hamdi 2000). Nevertheless, this pilot <strong>project</strong> h<strong>as</strong><br />

clearly shown that even the most antibiotic-resistance bacteria succumb to relatively<br />

239


low concentrations <strong>of</strong> <strong>honey</strong> in the laboratory and clinical studies are urgently<br />

needed. The most fe<strong>as</strong>ible suggestion on the action <strong>of</strong> <strong>honey</strong> on Gram negative<br />

bacteria is the alteration in cell wall or cell membrane integrity. The observation <strong>of</strong><br />

cell lysis suggested that there were significant changes in cell wall and pl<strong>as</strong>ma<br />

membrane integrity (possibly by incre<strong>as</strong>ed formation <strong>of</strong> pores) allowing <strong>honey</strong> to<br />

enter cells and cellular contents to escape (Table 4.3). Then the action <strong>of</strong> antioxidant,<br />

phenolic acid, MGO and other factors may be expected to contribute to the process<br />

<strong>of</strong> cell destruction, but the extent <strong>of</strong> each influence is <strong>as</strong> yet unknown. Another line<br />

<strong>of</strong> evidence to support this suggestion is that <strong>honey</strong> can act upon the structure <strong>of</strong> a<br />

biological membrane to disrupt it enough to allow the diffusion <strong>of</strong> protons into a<br />

liposome or bacterial cell (Grovell 2006). Very recently, Kwakman and his<br />

colleagues who identified the bee defensin-1 for the first time studied the<br />

<strong>antimicrobial</strong> activity <strong>of</strong> two important <strong>honey</strong>s (manuka and Revamil <strong>honey</strong>s)<br />

against different species. Also, the quantity <strong>of</strong> bee defensin-1, MGO and H2O2 were<br />

analysed for both <strong>honey</strong>s. It w<strong>as</strong> found Revamil killed bacteria rapidly because <strong>of</strong><br />

unique presence <strong>of</strong> bee defensin-1 and H2O2 which create ion channels in lipid<br />

bilayers and incre<strong>as</strong>e permeability <strong>of</strong> outer and inner biological membranes. Where<br />

<strong>as</strong> manuka <strong>honey</strong> retained its activity even with higher dilution, the presence <strong>of</strong> high<br />

sugar, low pH, high concentration <strong>of</strong> MGO (44 fold higher than Revamil) and other<br />

cationic and non- cationic bactericidal factors aid in the bactericidal activity <strong>of</strong><br />

manuka <strong>honey</strong> (Kwakman et al., 2011).<br />

Defensins are small, cysteine rich broad spectrum antibacterial peptides found in<br />

human neutrophils. Their <strong>antimicrobial</strong> activity on Gram negative and Gram positive<br />

bacteria, Mycobacteria, fungi and enveloped virus w<strong>as</strong> documented in-vitro (Kagan<br />

et al., 1994).<br />

240


The effect <strong>of</strong> defensin peptide w<strong>as</strong> first reported morphologically on Staph. aureus<br />

using TEM and caused major changes in the cytopl<strong>as</strong>mic membrane (Shimoda et al.,<br />

1995). It is known to incre<strong>as</strong>e permeability <strong>of</strong> the cell wall in Gram negative<br />

bacteria, causing cytopl<strong>as</strong>mic contents to leak out. Its effect on E.coli causes damage<br />

<strong>of</strong> outer and inner membranes by rapid permeabilization <strong>of</strong> the pl<strong>as</strong>ma membrane<br />

leading to cell death (Lichtenstein 1991). Defensin w<strong>as</strong> therefore proven to cause<br />

destruction in the biological membrane. However its effect on human tumour cells<br />

revealed that the primary target is the pl<strong>as</strong>ma membrane. Generally, antibacterial<br />

peptides affect several target sites on bacteria such <strong>as</strong> binding to intracellular<br />

compounds (DNA, RNA and proteins) or by bacterial enzymes inhibition (Patrzykat<br />

et al., 2002). The evidence that the effect <strong>of</strong> bee defensin-1 is similar to human<br />

defensin will provide a good indication in the mechanism <strong>of</strong> action <strong>of</strong> <strong>honey</strong><br />

containing that peptide on bacteria.<br />

Therefore, <strong>honey</strong> contains compounds that inhibit bacteria to develop resistance.<br />

Until now no report demonstrated that <strong>honey</strong> can develop resistance even in<br />

excessive use (Blair et al., 2009; Cooper et al., 2010)<br />

The statistics <strong>of</strong> war wound infection in wounded soldiers during military operations<br />

(2003-2005) in Iraq had incre<strong>as</strong>ed compared to previous operation in Korea,<br />

Vietnam and the Persian Gulf. This h<strong>as</strong> caused incre<strong>as</strong>e prevalence <strong>of</strong> MDR<br />

Acinetobacter calcoaceticus-baumannii complex isolated from wound and<br />

subsequently caused bacteraemia (Murray et al., 2008). Most investigations by<br />

medical military research suggested that this is a nosocomial infection but their<br />

source w<strong>as</strong> not known. Despite careful procedure in the selection <strong>of</strong> antibiotic<br />

therapy in such outbreaks there will be limited options <strong>of</strong> drug therapy available to<br />

241


treat this infection (Davis et al., 2005). In addition global incre<strong>as</strong>e in antibiotic<br />

resistant organisms causes major problems in controlling the emergence <strong>of</strong> resistance<br />

(Hawkey and Jones 2009). Currently an incre<strong>as</strong>e <strong>of</strong> aggressive situations worldwide<br />

i.e war in Libya and earthquake in Japan may cause incre<strong>as</strong>ed prevalence <strong>of</strong> resistant<br />

strain <strong>of</strong> Acinetobacter. This highlights the necessirly to develop alternative<br />

treatments to eliminate such infections. As <strong>honey</strong> h<strong>as</strong> several effective properties in<br />

wound healing including anti-inflammatory properties, providing moist environment<br />

around the wound, non toxic, reduce exudates and oedema, painless, debriding <strong>agent</strong><br />

and e<strong>as</strong>e <strong>of</strong> application (Molan 2006). In addition rapid wound healing due to <strong>honey</strong><br />

treatment reduces hospital stays especially in chronic wound patients and reduces the<br />

time required for doctors and nurses until the wound heals. This results in a decre<strong>as</strong>e<br />

in financial cost <strong>of</strong> the health care system. Thus it could be used <strong>as</strong> a perfect<br />

treatment.<br />

The main conclusion obtained from this study is that the <strong>antimicrobial</strong> effect <strong>of</strong><br />

<strong>honey</strong> is most probably due to several factors controlling its activity. Thus the<br />

mechanisms <strong>of</strong> action <strong>of</strong> <strong>honey</strong> could have various target sites such <strong>as</strong> proteins,<br />

enzymes and DNA.<br />

242


4.6 Further investigations:<br />

There is much evidence <strong>of</strong> antibacterial activity <strong>of</strong> using <strong>honey</strong> in the topical<br />

treatment <strong>of</strong> infected wounds, however further <strong>as</strong>sumptions needs to be given to its<br />

systemic application and therapeutic properties in order to improve the use <strong>of</strong> this<br />

product in clinical and systemic infections especially with incre<strong>as</strong>e prevalence <strong>of</strong><br />

multi drug resistant bacteria.<br />

This pilot study <strong>of</strong> the Omani <strong>honey</strong>s needs more comprehensive research with<br />

different types <strong>of</strong> <strong>honey</strong>s collected from different parts in Oman in order to establish<br />

whether all <strong>honey</strong>s possess both antibacterial and healing properties. This could<br />

improve the identification <strong>of</strong> the most appropriate type <strong>of</strong> <strong>honey</strong> to be used for<br />

wound care in Oman particularly after isolation <strong>of</strong> highly resistant NDM gene <strong>of</strong><br />

Klebsiella pneumonia for the first time in Oman (Poirel et al., 2010). Also using an<br />

advanced method to identify and study the nature <strong>of</strong> other components in Omani<br />

<strong>honey</strong> could aid its <strong>antimicrobial</strong> effect such <strong>as</strong>, MGO, glucose oxid<strong>as</strong>e enzyme,<br />

antioxidant and bee defensin-1.<br />

Further study on the idenfication <strong>of</strong> bee defensin-1 in other <strong>honey</strong> samples to<br />

determine its effect on Gram postive and negative bacteria and to prove its<br />

simillarity effect <strong>as</strong> human defensin is needed. Omani <strong>honey</strong>s could be tested for bee<br />

defensin.<br />

Further study on the action <strong>of</strong> different types <strong>of</strong> <strong>honey</strong> to identify the intracellular<br />

morphological changes using electron microscopy is required. Investigation <strong>of</strong> the<br />

molecules that leaked out from the cells after exposure to <strong>honey</strong> such <strong>as</strong> DNA,<br />

pot<strong>as</strong>sium ions or other cell organell using leakage study. Furthermore, extensive<br />

243


study on the alteration <strong>of</strong> Gram negative bacterial cell wall structure after <strong>honey</strong><br />

treatment by invistigating the synthesis and degradation <strong>of</strong> murien and/or expression<br />

<strong>of</strong> murein hydrol<strong>as</strong>e and peptidoglycan hydrol<strong>as</strong>e enzymes is needed. These<br />

enzymes are involved in cell cycle by controlling the breakdown <strong>of</strong> cell wall<br />

components before cytokinesis and cell separation occurs. However, in Gram<br />

positive (MRSA) these enzymes showed reduced activity after <strong>honey</strong> treatment<br />

(Jenkins et al., 2011). By investigating these effects, a better understanding <strong>of</strong> the<br />

effect <strong>of</strong> manuka <strong>honey</strong> on cell cycle <strong>of</strong> Gram negative bacterial will be achieved.<br />

Large scale analysis on proteome expression using 2-D electrophoresis and kinetics<br />

<strong>of</strong> RNA using radioactive isotopes in different organisms, provides important<br />

information about the mode <strong>of</strong> action. Another method using MALDI-TOF- MS<br />

techniques for the identity <strong>of</strong> protein after the detetcion <strong>of</strong> down regulated or<br />

upregulated protiens in the proteome analysis in 2-D electrophoresis. Microarray<br />

methods could be used to explain the reproducibility <strong>of</strong> gene expression on <strong>honey</strong><br />

treated cells compared to non <strong>honey</strong> treated cells.<br />

To determine whether <strong>honey</strong> h<strong>as</strong> simillar activity in vivo, further clinical trials on<br />

wounds infected with MDR Acinetobacter or other ESBLs and treated with different<br />

types <strong>of</strong> <strong>honey</strong> must be conducted. With the current difficulties in treating ESBL<br />

infections in wounds, <strong>honey</strong> does seem to <strong>of</strong>fer real potential in eradicating these<br />

pathogens.<br />

244


Chapter 5<br />

References<br />

245


� Adams, C. J., Boult, C. H., Deadman, B. J., Farr, J. M., Grainger, M. N. C.,<br />

Manley-Harris M., Snow, M. J. (2008) Isolation by HPLC and<br />

characterisation <strong>of</strong> the bioactive fraction <strong>of</strong> New Zealand manuka<br />

(Leptospermum scoparium) <strong>honey</strong>, Carbohydrate Research, 343(4), pp, 651-<br />

659.<br />

� Ahmed, A. K. J., Hoekstra, M. J., Hage, J. J. and Karim, R. B. (2003) Honey-<br />

medicated dressing: transformation <strong>of</strong> an ancient remedy, Annals <strong>of</strong> Pl<strong>as</strong>tic<br />

Surgery, 50, pp, 143–148.<br />

� Akratanakul, P. (1990) Beekeeping in Asia; Honeybees <strong>of</strong> the Genus Apis,<br />

Food and Agriculture Organization <strong>of</strong> the United Nations, FAO Agricultural<br />

Services Bulletin68/4, Rome, Italy<br />

� Aksoy, D. Y., Tanriover, M. D. and Unal, S. (2007) Antimicrobial resistance,<br />

in Gould, I. M. and Van Deer Meer, J. W. M. (ed.) Antibiotic policies:<br />

fighting resistance, Springer Sciences.<br />

� Alandejani, T., Marsan, J., Ferris, W. and Chen, F. (2009) Effectiveness <strong>of</strong><br />

<strong>honey</strong> on Staphylococcus aureus and Pseudomon<strong>as</strong> aeruginosa bi<strong>of</strong>ilms,<br />

Otolaryngology-Head and Neck Surgery, 139 (1), pp, 107-111.<br />

� Al-Jabri, A. A., Al-Hosni, S. A., Nzeako, B. C., Al-Mahrooqi, Z. H. and<br />

Nsanze, H. (2005a) Antibacterial activity <strong>of</strong> Omani <strong>honey</strong> alone and in<br />

combination with gentamicin, Saudi Medical Journal, 26 (5), pp, 767-771.<br />

� Al-Jabri, A. A., Al-Hosni, S. A., Nzeako, B. C. and Nsanze, H. (2005b)<br />

Antistaphylococcal activity <strong>of</strong> Omani <strong>honey</strong> in combination with bovine<br />

milk, British Journal <strong>of</strong> Biomedical Science, 62 (2), pp, 92-93.<br />

246


� Al-Jabri, A. A., Nzeako, B. C., Al-Mahrooqi, Z. H., Al-Naqdi, A. and<br />

Nsanze, H. (2003) In vitro antibacterial activity <strong>of</strong> Omani and African <strong>honey</strong>,<br />

British Journal <strong>of</strong> Biomedical Science, 60 (1), pp, 1-4<br />

� Al-Jadi, A. M. and Kamaruddin, M.Y. (2004) Evaluation <strong>of</strong> the phenolic<br />

contents and antioxidant capacities <strong>of</strong> two Malaysian floral <strong>honey</strong>s, Food<br />

Chemistry, 85, pp, 513-518.<br />

� Al-Naqdy, A., Al-Jabri, A. A., Al-Mahrooqi, Z. H., Nzeako, B. C. and<br />

Nsanze, H. (2005) Inhibition effect <strong>of</strong> <strong>honey</strong> on the adherence <strong>of</strong> Salmonella<br />

to intestinal epithelial cells in vitro, International Journal <strong>of</strong> Food<br />

Microbiology, 103, pp, 347-351.<br />

� Allen, K. L., Molan, P. C. and Reid, G. M. (1991) A survey <strong>of</strong> the<br />

antibacterial activity <strong>of</strong> some New Zealand <strong>honey</strong>s, Journal <strong>of</strong> Pharmacy<br />

Pharmacology, 43 (12), pp, 817-822.<br />

� Al-Mamary, M., Al-Meeri, A. and Al-Habori M. (2002) Antioxidant<br />

activities and total phenolic <strong>of</strong> different types <strong>of</strong> <strong>honey</strong>, Nutrition Research,<br />

22, pp, 1041-1047.<br />

� Al Somal, N., Coley, K. E., Molan, P. C. and Hancock, B. M (1994)<br />

Susceptibility <strong>of</strong> Helicobacter pylori to the antibacterial activity <strong>of</strong> manuka<br />

<strong>honey</strong>, Journal <strong>of</strong> the Royal Society <strong>of</strong> Medicine, 87, pp, 9-12.<br />

� Al-Taie, H., Pickersgill, J., and Al-Taie, N. (1999) Oman, A comprehensive<br />

Guide to the Sultanate <strong>of</strong> Oman, 2 nd edition, Al-Roya Publishing, Oman.<br />

� Al- Waili, N. S. and Saloom, K. Y. (1999) Effects <strong>of</strong> topical <strong>honey</strong> on post-<br />

operative wound infections due to gram positive and gram negative bacteria<br />

following caesarean sections and hysterectomies, European Journal <strong>of</strong><br />

Medical Research, 4, pp, 126-30.<br />

247


� Al- Waili, N. S. (2004) Investigating the <strong>antimicrobial</strong> activity <strong>of</strong> natural<br />

<strong>honey</strong> and its effects on the pathogenic bacterial infections <strong>of</strong> surgical wound<br />

and conjunctiva, Journal <strong>of</strong> Medicinal Food, 7 (2), pp, 210-222.<br />

� Amabile-Cuev<strong>as</strong>, C. F. (2007) Antimicrobial Resistance in Bacteria, Horizon<br />

Bioscience, UK.<br />

� Anklam, E. (1998). A review <strong>of</strong> the analytical methods to determine the<br />

geographical and botanical origin <strong>of</strong> <strong>honey</strong>, Food Chemistry, pp, 63, 549-<br />

562.<br />

� Atrott, J. and Henle, T. (2009) Methylglyoxal in Manuka Honey -Correlation<br />

with Antibacterial Properties, Czech Journal Food Science, 27, Special Issue,<br />

S163-S165.<br />

� Atrouse, O. M., Oran, S. A. and Al-Abbadi, S. Y. (2004) Chemical analysis<br />

and identification <strong>of</strong> pollen grains from different Jordanian <strong>honey</strong> samples,<br />

International Journal <strong>of</strong> Food Science and Technology, 39 (4): 413-417.<br />

� Babik, J. and Bodnarova, L. (2008) Acinetobacter- serious danger for burn<br />

patients, Acta Chir Pl<strong>as</strong>t, 50 (1), pp, 27-32.<br />

� Bacakoglu, F. and Korkmaz, E. P. (2009) Multidrug-resistant Acinetobacter<br />

baumannii infection in respiratory intensive care unit, Microbiology Bul, 43<br />

(4), pp, 575-585.<br />

� Badawy, O.F., Shafii, S.S., Tharwat, E.E. and Kamal, A.M. (2004)<br />

Antibacterial activity <strong>of</strong> bee <strong>honey</strong> and its therapeutic usefulness against<br />

Escherichia coli O157:H7 and Salmonella typhimurium infection, Rev. sci.<br />

tech. Off. int. Epiz, 23 (3), pp, 1011-1022<br />

248


� Bang, L. M., Buntting, C. and Molan, P.C. (2003) The effect <strong>of</strong> dilution on<br />

the rate <strong>of</strong> hydrogen peroxide production in <strong>honey</strong> and its implications for<br />

wound healing, J Alternative Complement Med, 9 (2), pp, 267-273<br />

� Barchitta, M., Cipresso, R., Giaquinta, L., Romeo, M. A., Denaro, C.,<br />

Pennisi, C. and Agodi, A. (2009) Acquisition and spread <strong>of</strong> Acinetobacter<br />

baumannii and Stenotrophomon<strong>as</strong> maltophilia in intensive care patients,<br />

International Journal <strong>of</strong> Hygiene and Environmental Health, 212 (3), pp,<br />

330-337.<br />

� Bardy, J., Slevin, N. J., Mais, K. L. and Mol<strong>as</strong>siotis, A. A. (2008) Systematic<br />

review <strong>of</strong> <strong>honey</strong> uses and its potential value within oncology care, Journal <strong>of</strong><br />

Clinical Nursing, 17(19), pp, 2604-2623.<br />

� Baron, S. (1996) Baron's Medical Microbiology, (ed), 4 th edition, Galveston,<br />

<strong>University</strong> <strong>of</strong> Tex<strong>as</strong> Medical Branch, ISBN 0-9631172-1-14<br />

� B<strong>as</strong>son, N. J. and Grobler,S. R. (2008) Antimicrobial activity <strong>of</strong> two South<br />

African <strong>honey</strong>s produced from indigenous Leucospermum cordifolium and<br />

Erica species on selected micro-organisms, BMC Complementary and<br />

Alternative Medicine, 8:41, doi:10.1186/1472-6882-8-41.<br />

� Beretta, G., Granata, P., Ferrero, M., Orioli, M. and Facino, R. M. (2005)<br />

Standardization <strong>of</strong> antioxidant properties <strong>of</strong> <strong>honey</strong> by a combination <strong>of</strong><br />

spectrophotometric/fluorimetric <strong>as</strong>says and chemometrics, Anal Chim<br />

Acta,533, pp, 185-191<br />

� Bergogne-Bérézin, E., Friedman, H. and Bendinelli, M. (2008)<br />

Acinetobacter: Biology and Pathogenesis (Infectious Agent and<br />

Pathogenesis), Springer Sciences and Business Media.<br />

249


� Bergman, A., Yanai, J., Weiss, J., Bell, D. and David, M. P. (1983)<br />

Acceleration <strong>of</strong> wound healing by topical application <strong>of</strong> <strong>honey</strong>: An animal<br />

model, Am J Surg, 145, pp, 374-376.<br />

� Bhattacharya, S. (2006) ESBL- from petri dish to the patient, Indian J<br />

Medical Microbiology, 24(1), pp, 20-24.<br />

� Blair, S. E., Cokecetin, N. N., Harry, E. J. and Carter, D. A. (2009) The<br />

unusual antibacterial activity <strong>of</strong> medical-grade Leptospermum <strong>honey</strong>:<br />

antibacterial spectrum, resistance and transcriptome analysis, Eur J Clin<br />

Microbial Infect Dise<strong>as</strong>e, Springer.<br />

� Bl<strong>as</strong>a, M., Candiracci, M., Accorsi, A., Piacentini, M. P., Albertini, M. C.<br />

and Piatti, E. (2006) Raw Millefiori <strong>honey</strong> is packed <strong>full</strong> <strong>of</strong> antioxidants,<br />

Food Chemistry, 97 (2), 217-222.<br />

� Bogdanov, S. (1997) Nature and origin <strong>of</strong> the antibacterial substances in<br />

<strong>honey</strong>, Lebensm-Wiss. U- Technology, 30, pp, 748-753.<br />

� Bogdanov, S. (2002) Harmonized methods <strong>of</strong> the International Honey<br />

Commission, Swiss Bee Research Centre, FAM, Liebefeld, CH-3003 Bern,<br />

Switzerland.<br />

� Bogdanov, S., Rieder, K., and Ruegg, M. (1989) Determination <strong>of</strong><br />

pinocembrin in <strong>honey</strong> using HPLC, J Apicultural Research, 28, pp, 55-57<br />

� Bogdanov, S., Ru<strong>of</strong>f, K. and Oddo. L. (2004) Physico-chemical methods for<br />

charactirisation <strong>of</strong> unifloral <strong>honey</strong>s: a review, Apidologie, 35, S5-S17.<br />

� Borland, M. (2000) Honey proves a sweet remedy in wound care, Nursing<br />

Review (Oct)<br />

250


� Bou, G., Oliver, A. and Martinez-Beltrán, J. (2000) OXA-24, a novel cl<strong>as</strong>s D<br />

β-lactam<strong>as</strong>e with carbapenem<strong>as</strong>e activity in an Acinetobacter baumannii<br />

clinical strain, Antimicrobial Agents and Chemotherapy 44, pp, 1556–1561.<br />

� Booth, I. R. (1985) Regulation <strong>of</strong> Cytopl<strong>as</strong>mic pH in Bacteria,<br />

Microbiological Reviews, 49 (4), pp, 359-378.<br />

� Bradford, P. A. (2001) Extended-spectrum β-lactam<strong>as</strong>es in the 21st century:<br />

characterization, epidemiology, and detection <strong>of</strong> this important resistance<br />

threat, Clinical Microbiology Reviews, 14 (4), pp, 933-951.<br />

� Brady, N. F., Molan, P. C. and Harfoot, C. G. (1996) The sensitivity <strong>of</strong><br />

dermatophytes to the <strong>antimicrobial</strong> activity <strong>of</strong> manuka <strong>honey</strong> and other<br />

<strong>honey</strong>, Pharm. Science, 2(10), pp, 471-473.<br />

� Brauers, J., Frank, U., Kresken, M., Rodl<strong>of</strong>f, A. C. and Seifert, H. (2005)<br />

Activities <strong>of</strong> various beta-lactams and beta-lactam/beta-lactam<strong>as</strong>e inhibitor<br />

combinations against Acinetobacter baumannii and Acinetobacter DNA<br />

group 3 strains, Clinical Microbiology and Infection; 11,pp, 24-30.<br />

� Brooks, G. F., Butel, J. S. and Morse, S. A. (2001) Medical Microbiology,<br />

Twenty two edition, Lange, USA.<br />

� Brudzynski, K. (2006) Effect <strong>of</strong> hydrogen peroxide on antibacterial activities<br />

<strong>of</strong> Canadian <strong>honey</strong>s, Canadian Journal <strong>of</strong> Microbiology, 52, 1228-1237.<br />

� Burdon, R. H. (1995) Superoxide and hydrogen peroxide in relation to<br />

mammalian cell proliferation, Free Rad Biological Medicine, 18, pp, 775-794<br />

� Bush, K., Jacoby, G. A., Medeiros, A. A. (1995) A functional cl<strong>as</strong>sification<br />

scheme for beta-lactam<strong>as</strong>es and its correlation with molecular structure,<br />

Antimicrobial Agents and Chemotherapy, 39, 1211-1233.<br />

251


� Canton, R., Novais, A., Valverde, A., Machado, E., Peixe, L., Baquero, F.<br />

and Coque, T. M. (2008) Prevalence and spread <strong>of</strong> extended- spectrum beta-<br />

lactam<strong>as</strong>e producing Enterobacteriaceae in Europe, Clinical Microbiology &<br />

Infectious Dise<strong>as</strong>es, 14 (1), pp, 144-153<br />

� C<strong>as</strong>ey, J. T., O‟Cleirigh, C., Walsh, P. K. and O‟Shea, D. G. (2004)<br />

Development <strong>of</strong> a robust microtiter plate-b<strong>as</strong>ed <strong>as</strong>say method for <strong>as</strong>sessment<br />

<strong>of</strong> bioactivity, Journal <strong>of</strong> Microbiological Methods, 58, pp, 327-334.<br />

� C<strong>as</strong>tanheira, M., Sader, H. S., Deshpande, L. M et al. (2008) Antimicrobial<br />

activity <strong>of</strong> tigecycline and other broad spectrum <strong>antimicrobial</strong>s tested against<br />

serine carbapenem<strong>as</strong>e- and metallo-b-lactam<strong>as</strong>eproducing<br />

Enterobacteriaceae: report from the SENTRY Antimicrobial surveillance<br />

Program, Antimicrobial Agents Chemotherapy, 52, pp, 570-573.<br />

� Chen, L., Mehta, A., Berenbaum, M., Zangerl, A. R. and Engeseth, N. J.<br />

(2000) Honeys from different floral sources <strong>as</strong> inhibitors <strong>of</strong> enzymatic<br />

browning in fruit and vegetable homogenates, Journal <strong>of</strong> Agriculture and<br />

Food Chemistry, 48, pp, 4997-5000.<br />

� Chiang, D. H. and Wang, C. C. (2008) Risk factors for mortality in patients<br />

with Acinetobacter baumannii bloodstream infection with genotypic species<br />

identification, Journal <strong>of</strong> Microbiology and Immunological Infection, 41(5),<br />

pp, 397-402.<br />

� CLSI <strong>antimicrobial</strong> susceptibility testing standards M2-A9 and M7-A7<br />

(2008) Performance standard for <strong>antimicrobial</strong> susceptibility testing;<br />

Eighteen informational supplement, Vol 28, No 1<br />

252


� Cooper, R. (2005) The Antimicrobial Activity <strong>of</strong> Honey, in White, R.,<br />

Cooper, R. and Molan, P. C. (ed.) Honey: A modern Wound Management<br />

product, Aberdeen, UK.<br />

� Cooper, R. A., Hal<strong>as</strong>, E. and Molan, P. C. (2002b) The efficacy <strong>of</strong> <strong>honey</strong> in<br />

inhibiting strains <strong>of</strong> Pseudomon<strong>as</strong> aeruginosa from infected burns, J Burn<br />

Care Rehabil, 23(6):366-370.<br />

� Cooper, R. A., Molan, P. C. and Harding, K. G. (2002a) The sensitivity to<br />

<strong>honey</strong> <strong>of</strong> Gram-positive cocci <strong>of</strong> clinical significance isolated from wounds, J<br />

Appl Microbiol, 93, pp, 857-863.<br />

� Cooper R. A & Jenkins L, (2009) Comparison between Medical Grade<br />

Honey and table <strong>honey</strong>s in relation to <strong>antimicrobial</strong> efficacy, Wound,<br />

21(2):29-36.<br />

� Cooper, R. A., Molan, P. C. and Harding, K. G. (1999) Antibacterial activity<br />

<strong>of</strong> <strong>honey</strong> against strains <strong>of</strong> Staphylococcus aureus from infected wounds.<br />

Journal <strong>of</strong> the Royal Society <strong>of</strong> Medicine 92 (6), pp, 283-285.<br />

� Cooper, R.A., Jenkins, L., Henriques, A. F., Duggan, R.S. and Burton. N. F.<br />

(2010) „Absence <strong>of</strong> bacterial resistance to medical-grade manuka <strong>honey</strong>‟,<br />

Eur J Clin Microbiol Infect Dis, 29(10), pp, 1237-12341.<br />

� Cooper, R. A., Wigley, P. and Burton, N. F. (2000) Susceptibility <strong>of</strong> multi-<br />

resistant strains <strong>of</strong> Burkholderia cepacia to <strong>honey</strong>, Letters in Applied<br />

Microbiology, 31 (1), pp, 20-24.<br />

� Cootz, T. D. and Marra, A. (2008) Acinetobacter baumannii: an emerging<br />

multidrug-resistant threat, Expert Review <strong>of</strong> Anti Infection Therapy, 6 (3), pp,<br />

309-325.<br />

253


� Costa, S. F., Woodcook, J. and Gill, M. (2000) Outer-membrane protein<br />

pattern and detection <strong>of</strong> beta-lactam<strong>as</strong>es in clinical isolates <strong>of</strong> imipenem-<br />

resistant Acinetobatcer baumannii from Brazil, International Journal <strong>of</strong><br />

Antimicrobial Agent, 13, pp, 175-182.<br />

� Cotter, P. D and Hill, C. (2003) Surviving the Acid Test: Responses <strong>of</strong> Gram-<br />

Positive Bacteria to Low pH, Microbiology and Molecular Biology Reviews,<br />

67(3), pp, 429-453. DOI: 10.1128/MMBR.<br />

� Crane, E. (1979) Honey: A Comprehensive Survey, Heinemann, London, UK.<br />

� Crovell, K. (2006) Comparison <strong>of</strong> the effect <strong>of</strong> manuka <strong>honey</strong> and its main<br />

constituent organic acid on artificial membranes, BSC (hons) BMS,<br />

<strong>University</strong> <strong>of</strong> Wales Institute, Cardiff.<br />

� Dallo, S. F. and Weitao, T. (2010) Insights into Acinetobacter war-wound<br />

infections, bi<strong>of</strong>ilms, and control, Advances in skin and wound care, 23 (4),<br />

pp, 169-174.<br />

� Davis, K. A., Moran, K. A., McAllister, C. K. and Gray, P. J. (2005)<br />

Multidrug-Resistant Acinetobacter Extremity Infections in Soldiers, Journal<br />

<strong>of</strong> Emerging Infectious Dise<strong>as</strong>es, 11(8), pp, 1218-1224.<br />

� Dumronglert, E. (1983) A follow-up study <strong>of</strong> chronic wound healing dressing<br />

with pure natural <strong>honey</strong>, J Nat Res Counc Thail, 15(2), pp, 39-66.<br />

� Dustmann, J. H. (1979) Antibacterial effect <strong>of</strong> <strong>honey</strong>, Apiacta, 14 (1), pp, 7-<br />

11.<br />

� Eddy, J. J. and Gideonsen, M. D. (2005) Topical <strong>honey</strong> for diabetic foot<br />

ulcers, J Fam Pract, 54(6), pp, 533-535.<br />

� Efem, S. E. (1988) Clinical observations on the wound healing properties <strong>of</strong><br />

<strong>honey</strong>, Br. J. Surg, 75, pp, 679-681.<br />

254


� Efem, S. E., Udoh, K. T. and Iwara, C. I. (1992) The <strong>antimicrobial</strong> spectrum<br />

<strong>of</strong> <strong>honey</strong> and its clinical significance, Infection, 20(4), pp, 227-229.<br />

� Efem, S. E. (1993) Recent advances in the management <strong>of</strong> Fournier's<br />

gangrene: Preliminary observations, Surgery, 113 (2), pp, 200-204.<br />

� Emarah, M. H. (1982) A clinical study <strong>of</strong> the topical use <strong>of</strong> bee <strong>honey</strong> in the<br />

treatment <strong>of</strong> some occular dise<strong>as</strong>es, Bull Islamic Med, 2(5), pp, 422-425.<br />

� Engelkirk, P. G. (2007) Laboratory Diagnosis <strong>of</strong> Infectious Dise<strong>as</strong>es:<br />

Essential <strong>of</strong> Diagnostic Microbiology, Philadelphia, USA, Lippincott<br />

Williams & Wilkins.<br />

� Enoch, D. A., Summers, C., Brown, N. M, Moore, L., Gillham, M. I.,<br />

Burnstein, R. M., Thaxter, R. et al., (2008), Investigation and management <strong>of</strong><br />

an outbreak <strong>of</strong> multidrug-carbapenem-resistant Acinetobacter baumannii in<br />

Cambridge, UK, Journal <strong>of</strong> Hospital Infection, 70, pp, 109-118.<br />

� Estevinho, L., Pereira, A. P., Moreira, L., Di<strong>as</strong>, L. G. and Pereira, L. (2008)<br />

Antioxidant and <strong>antimicrobial</strong> effects <strong>of</strong> phenolic compounds extracts on<br />

Northe<strong>as</strong>t Portugal <strong>honey</strong>, Food and Chemical Toxicology, 46, pp, 3774-<br />

3779.<br />

� Finch, R. G., Greenwood, D., Norrby, S. R. and Whitley, R. J. (2003)<br />

Antibiotics and Chemotherapy: Anti-infective Agent and their use in Therapy,<br />

Eight edition, Churchil Livingstone, UK.<br />

� Fontana, C. and Favaro, M. (2008) Acinetobacter baumannii in intensive care<br />

unit: a noval system to study clonal relationship among the isolates, BMC<br />

Infectious dise<strong>as</strong>e, 8, pp,79.<br />

255


� Forbes, B. A., Sahm, D. F., Weissfeld, A. S., Bailey, W. R., Trevino, E. A.<br />

and Scott, E. G. (2007) Bailey & Scotts diagnostic microbiology, Twelve<br />

edition, London, UK, Elsevier Mosby.<br />

� French, V. M., Cooper, R. A. and Molan, P. C. (2005) The antibacterial<br />

activity <strong>of</strong> <strong>honey</strong> against coagul<strong>as</strong>e-negative staphylococci, J Antimicrobial<br />

Chemotherapy, 56(1), pp, 228-231.<br />

� Gheld<strong>of</strong>, N., Wang, X. H, and Engeseth, N. J. (2002) Identification and<br />

quantification <strong>of</strong> antioxidant components <strong>of</strong> <strong>honey</strong>s from various floral<br />

sources, Journal <strong>of</strong> Agricultural and Food Chemistry, 50, (21), pp, 5870-<br />

5877.<br />

� George, N. M. and Cutting, K. F. (2007) Antibacterial <strong>honey</strong> (Medi<strong>honey</strong> TM ):<br />

in-vitro activity against clinical isolates <strong>of</strong> MRSA, VRE, and other multi-<br />

resistant gram negative, Wounds, 19 (9): 231-336.<br />

� Gerisher, U. (2008) Acinetobacter Molecular Biology, Caister Academic<br />

Press, UK<br />

� Gethin, G.T., Cowman, S. and Conroy, R.M. (2008) The impact <strong>of</strong> Manuka<br />

<strong>honey</strong> dressings on the surface pH <strong>of</strong> chronic wounds, International Wound<br />

Journal, 5, pp, 185–194. doi:10.1111/j.1742-481X.2007.00424.x<br />

� Gillespie, S. H. (2004) Management <strong>of</strong> Multiple Drug Resistant Infections.<br />

New York, USA, Humana Press.<br />

� Greenwood, D. (2000) Antimicrobial Chemotherapy, Forth edition, Oxford<br />

<strong>University</strong> press, USA<br />

� Greenwood, D., Finch, R. G., Whintley, R. J. (2003) Antibiotic and<br />

Chemotherapy, 8 th edition, Edinburgh, Churchill Livingstone.<br />

256


� Greenwood, D., Finch, P., Davey, P. and Wilcox, M. (2007) Antimicrobial<br />

Chemotherapy, Fifth edition, Oxford <strong>University</strong> press, UK.<br />

� Haffejee, I. E. and Moosa, A. (1985) Honey in the treatment infantile<br />

g<strong>as</strong>troenteritis, British Medical Journal, 290, pp, 1866-1867.<br />

� Hawkey, P. M. (2008) The growing burden <strong>of</strong> <strong>antimicrobial</strong> resistance,<br />

Journal <strong>of</strong> Antimicrobial Chemotherapy, 62, Suppl. 1, i1–i9.<br />

� Hawkey, P. and Bergogne-Berezin, E. (2006) Acinetobacter spp. in Gillespie,<br />

S. H. and Hawkey, P. M. (ed.) Principle and Practical <strong>of</strong> Clinical<br />

Bacteriology, second edition, John Wiley & Sons, Ltd, UK.<br />

� Hawkey, P. M. and Jones, A. M. (2009) The changing epidemiology <strong>of</strong><br />

resistance, Journal <strong>of</strong> Antimicrobial Chemotherapy, 64, Suppl 1:i3-10<br />

� Health Protection Agency, (2008) Antimicrobial Resistance and Prescribing<br />

in England, Wales and Northern Ireland, London: Health Protection Agency<br />

(HPA), July.<br />

� Henriques, A., Burton, N. and Cooper, R. (2005) Antibacterial activity <strong>of</strong><br />

selected Portuguese <strong>honey</strong>s, Journal <strong>of</strong> Apicultural Research, 44(3), pp, 119-<br />

123.<br />

� Henriques, A., Jackson, S., Cooper, R. and Burton, N. (2006) Free radical<br />

production and quenching in <strong>honey</strong>s with wound healing potential, Journal<br />

<strong>of</strong> Antimicrobial Chemotherapy, 58, pp, 773–777.<br />

� Henriques, A., Jenkins, R., Burton, N. and Cooper, R. (2009) The<br />

intracellular effects <strong>of</strong> manuka <strong>honey</strong> <strong>of</strong> Staphylococcus aureus, Eur J Clin<br />

Microbial Infect Dise<strong>as</strong>e, Springer Sciences.<br />

257


� Henriques, A., Jenkins, R., Burton, N. and Cooper, R. (2010) The effects <strong>of</strong><br />

manuka <strong>honey</strong> on the structure <strong>of</strong> Pseudomon<strong>as</strong> aeruginosa, Eur J Clin<br />

Microbial Infect Dise<strong>as</strong>e, Springer.<br />

� Hobot, J. A. (2002) Bacterial ultr<strong>as</strong>tructure, , in Sussman, M. (ed.) Molecular<br />

medical microbiology, Vol 2, Cambridge <strong>University</strong> press, UK.<br />

� Ingle, R., Levin, J. and Polinder, K. (2006) Wound healing with <strong>honey</strong>- a<br />

randomised controlled trial, South African Medical Journal, 96(9), pp, 831–<br />

835.<br />

� Irish, J., Blair, S. and Carter, D. A. (2011) The Antibacterial Activity <strong>of</strong><br />

Honey Derived from Australian Flora, PLoS ONE, 6(3), e18229.<br />

doi:10.1371/journal.pone.0018229<br />

� Irish, J., Carter, D. A., Shokohi, T. and Blair, S. E. (2006) Honey h<strong>as</strong> an<br />

antifungal effect against Candida species, Medical Mycology, 44, pp, 289-<br />

291.<br />

� Jacoby, G. A. and Munoz-Price, L. S. (2005) The new beta lactam<strong>as</strong>es, New<br />

England Journal Medicine, 352, pp, 380-391<br />

� Jenkins, R., Burton, N. and Cooper, R. (2011) Effect <strong>of</strong> manuka <strong>honey</strong> on the<br />

expression <strong>of</strong> universal stress protein A in methicillin-resistant<br />

Staphylococcus aureus, International Journal <strong>of</strong> <strong>antimicrobial</strong> <strong>agent</strong>s, 37 (4),<br />

pp, 373-376.<br />

� Joly-Guillou, M. L. (2005) Clinical impact and pathogenicity <strong>of</strong><br />

Acinetobacter, Clinical Microbiology and Infection, CMI, 11, pp, 868-873.<br />

� Jul, A. B., Rodgers, A. and Walker, N. (2009) Honey <strong>as</strong> a topical treatment<br />

for wounds, (Review), The Cochrane Library, Issue 4,<br />

http://www.thecochranelibrary.com<br />

258


� Kagan, B.L., Ganz, T. and Lehrer, R. I. (1994) Defensins: a family <strong>of</strong><br />

<strong>antimicrobial</strong> and cytotoxic peptides, Toxicology, 87 (1-3), pp, 131-149.<br />

� Klepser, M., Ernst, E. J., Lewis, R. E., Ernst, M. E. and Pfaller, M. A. (1998)<br />

Influence <strong>of</strong> Test Conditions on Antifungal Time-Kill Curve Results:<br />

Proposal for Standardized Methods, Antimicrobial Agent and Chemotherapy,<br />

42 (5), pp, 1207-1212.<br />

� Knoneman, E. W., Alen, S. D., Janda, W. M., Schereckrenberger, P. C. and<br />

Winn, W. C. (1997) Colour atl<strong>as</strong> and Text book <strong>of</strong> Diagnostic Microbiology,<br />

Fifth edition, Lippincott, USA.<br />

� Konig, H., Claus, H. and Varma, A. (2010) Prokayotic cell wall compunds:<br />

Structure and biochemistry, Springer.<br />

� Kuda, T., Shimizu, K., Yano, T. (2004) Comparision <strong>of</strong> rapid and simple<br />

colorimetric microplate <strong>as</strong>says <strong>as</strong> an index <strong>of</strong> bacterial count, Journal <strong>of</strong><br />

Food Control, 15, pp, 421-425.<br />

� Kumar<strong>as</strong>amy, K.K., Toleman, M. A. and Walsh, T. R. (2010) emergence <strong>of</strong> a<br />

new antibiotic resistance mechanism in India, Pakistan, and the UK: A<br />

Molecular, biological, and epidemiological study, Lancet Infection Dise<strong>as</strong>e,<br />

10, pp, 597-602.<br />

� Kwakman, P. H. S., Johannes, P. C., Van den Akker, J. P., Ahmet, G.,<br />

Aslami, H., Binnekade, J. M., et al. (2008) Medical-grade <strong>honey</strong> kills<br />

antibiotic-resistant bacteria in-vitro and eradicates skin colonization, Clin<br />

Infect Dis,46, pp 1677-1682. DOI: 10.1086/587892.<br />

� Kwakman, P. H. S., Velde, A. A., de Boer, L., Speijer, D., Vandenbroucke-<br />

Grauls C. M. and Zaat, S. A. J. (2010) How <strong>honey</strong> kills bacteria, The FASEB<br />

Journal article, 24, pp, 2576-2582.<br />

259


� Kwakman, P. H. S., te Velde, A. A., de Boer, L., Vandenbroucke-Grauls, C.<br />

M. and Zaat, S. A. (2011) Two Major Medicinal Honeys Have Different<br />

Mechanisms <strong>of</strong> Bactericidal Activity, PLoS ONE , 6(3): e17709.<br />

doi:10.1371/journal.pone.0017709<br />

� Levin, A. S. (2001) Multiresistant Acinetobacter infections: a role for<br />

sulbactam combinations in overcoming an emerging worldwide problem,<br />

Clinical Microbiology Infection, 8, pp, 144-153.<br />

� Levison, M. E. (2004) Pharmacodynamics <strong>of</strong> <strong>antimicrobial</strong> drugs, Infectious<br />

Dise<strong>as</strong>e Clinics <strong>of</strong> North America, 18, pp, 451-465.<br />

� Lichtenstein, A. (1991) Mechanism <strong>of</strong> mammalian cell lyses mediated by<br />

peptide defensins: Evidence for an initial alteration <strong>of</strong> the pl<strong>as</strong>ma membrane,<br />

J. Clin. Invest. 88, pp, 93-100.<br />

� Loveaux, J., Maurizio, A. and Vorwohl, G. (1978) Methods <strong>of</strong><br />

Melissopalynology, Bee World, 59, pp, 139-157.<br />

� Lusby, P. E. (2002) Honey: a potent <strong>agent</strong> for wound healing?, Journal <strong>of</strong><br />

Wound, Ostomy and Continence Nursing, 29(6), pp, 295–300.<br />

� Lusby, P. E., Coombes, A. L. and Wilkinson, J. M. (2005) Bacterial activity<br />

<strong>of</strong> different <strong>honey</strong>s against pathogenic bacteria, Archives <strong>of</strong> medical<br />

Research, 36, 464-467.<br />

� Madigan, M. T., Martinko, J. M., Dunlap, P. V. and Clark, D. P. (2009)<br />

Brock, Biology <strong>of</strong> Microorganisms, 12 th edition, Pearson International<br />

Edition, USA.<br />

� Maedaa, Y. B., Loughreya, A., Earlec, P., Millara, C. et al, (2008)<br />

Antibacterial activity <strong>of</strong> <strong>honey</strong> against community-<strong>as</strong>sociated methicillin-<br />

260


esistant Staphylococcus aureus (CA-MRSA), Complementary Therapies in<br />

Clinical Practice 14, pp, 77–82.<br />

� Mavric, E., Wittmann, S., Barth, G., and Henle, T. (2008) Identification and<br />

quantification <strong>of</strong> methylglyoxal <strong>as</strong> the dominant antibacterial constituent <strong>of</strong><br />

manuka (Leptospermum scoparium) <strong>honey</strong>s from New Zealand, Mol Nutr<br />

Foods Res, 52(4), pp, 483-489.<br />

� Mohamed, M.1., Sirajudeen, K.N., Swamy, M., Yaacob, N.S. and Sulaiman,<br />

S.A. (2010) Studies on the antioxidant properties <strong>of</strong> Tualang <strong>honey</strong> <strong>of</strong><br />

Malaysia, Afr. J. Trad CAM, 7 (1), pp, 59 – 63, ISSN 0189-6016©2009.<br />

� Molan gold standard (http://www.molangoldstandard.co.nz/article).<br />

� Molan, P. C. (1992a) The antibacterial activity <strong>of</strong> <strong>honey</strong>. 1. The nature <strong>of</strong> the<br />

antibacterial activity, Bee World, 73(1), pp, 5-28.<br />

� Molan, P. C. (1992b) The antibacterial activity <strong>of</strong> <strong>honey</strong>. 2. Variation in the<br />

potency <strong>of</strong> the antibacterial activity, Bee World, 73(2), pp, 59-76.<br />

� Molan, P. C. (1999) The role <strong>of</strong> <strong>honey</strong> in the management <strong>of</strong> wounds,<br />

Journal <strong>of</strong> Wound Care, 8(8), pp, 423- 426<br />

� Molan, P. C. (2000a) Selection <strong>of</strong> <strong>honey</strong> for use <strong>as</strong> a wound dressing,<br />

Primary Intention, 8 (3), pp, 87-92.<br />

� Molan, P. C. (2001b) Why <strong>honey</strong> is effective <strong>as</strong> a medicine. 2. The scientific<br />

explanation <strong>of</strong> its effects, Bee World, 82 (1), pp, 22-40.<br />

� Molan, P. C. (2002) Re-introducing <strong>honey</strong> in the management <strong>of</strong> wounds and<br />

ulcers: theory and practice, Ostomy Wound Management, 48, pp, 28-40.<br />

� Molan, P. C. (2006) The evidence supporting the use <strong>of</strong> <strong>honey</strong> in wound<br />

healing, International Journal <strong>of</strong> Lower Extremity Wounds, 5(1), pp, 40–54.<br />

DOI: 10.1177/15347346052860145: 40<br />

261


� Molan, P. C. and Betts, J. A. (2001) Dressing wounds with <strong>honey</strong>, NZ<br />

Nursing Review (March issue), pp, 19-20.<br />

� Molan, P. C. and Russell, K. M. (1988) Non-peroxide antibacterial activity in<br />

some New Zealand <strong>honey</strong>s, J Apic Res, 27, pp, 62–67.<br />

� Moland, E. S., Kim, S. Y., Hong, S. G. and Thomson, K. S. (2008). Newer<br />

beta lactam<strong>as</strong>es: Clinical and laboratory implications, Part 1, Clinical<br />

Microbiology Newsletter, 30(10), pp, 71-77.<br />

� Moore, O. A., Smith, L. A., Campbell, F., Seers, K., McQuay, H. J. and<br />

Moore, R. A. (2001) Systematic review <strong>of</strong> the use <strong>of</strong> <strong>honey</strong> <strong>as</strong> a wound<br />

dressing, BMC, Complement Alternative Medicine, 1:2.<br />

� Mullai, V. and Menon, T. (2005) Antibacterial activity <strong>of</strong> <strong>honey</strong> against<br />

Pseudomon<strong>as</strong> aeruginosa, Indian Journal <strong>of</strong> Pharmacology, 37, pp, 403<br />

� Mullai V, Menon T. (2007) Bacterial activity <strong>of</strong> different types <strong>of</strong> <strong>honey</strong><br />

against clinical and environment isolates <strong>of</strong> Pseudomon<strong>as</strong> aeruginosa, The<br />

Journal <strong>of</strong> Alternative and Complementary Medicine, 13 (4), pp, 439–441,<br />

DOI: 10.1089/acm.2007.6366.<br />

� Mulu, A., Tessema, B. and Derbie, F. (2004) In vitro <strong>as</strong>sessment <strong>of</strong> the<br />

<strong>antimicrobial</strong> potential <strong>of</strong> <strong>honey</strong> on common human pathogens, Ethiopian<br />

Jorunal <strong>of</strong> Health Dev. 18(2), pp, 107-111.<br />

� Murray, C., Scott, P., Moran, K. and Craft, D. (2008) US army experience<br />

with Acinetobacter in operation Iraqi freedom, in Bergogne-Bérézin, E.,<br />

Friedman, H. and Bendinelli, M. (ed.) Acinetobacter: Biology and<br />

Pathogenesis (Infectious Agent and Pathogenesis), Springer Sciences and<br />

Business Media.<br />

262


� Murray, P. R., Baron, E. J., Jorgensen, J. and Landry, M. L. (2007) Manual<br />

<strong>of</strong> Clinical Microbiology, Nine edition, W<strong>as</strong>hington, USA, ASM Press.<br />

� Nagai, T., Sakai, M., Inoue, R., Inoue, H. and Suzuki, N. (2001)<br />

Antioxidative activities <strong>of</strong> some commercially <strong>honey</strong>s, royal jelly and<br />

propolis, Food Chemistry, 75, pp, 237-240.<br />

� National Honey Board (NHB), Honey technical information,<br />

http://www.<strong>honey</strong>.com/nhb/technical/.<br />

� Nzeako, B. C. and Hamdi, J. (2000) Anti microbial potential <strong>of</strong> <strong>honey</strong> on<br />

some microbial isolates, Journal <strong>of</strong> Science Research Medicine, 2, pp, 75-79.<br />

� Okeniyi, J. A., Olubanjo, O., Ogunlesi, T. A. and Oyelami, O. A. (2005)<br />

Comparesion <strong>of</strong> healing <strong>of</strong> incised abscess wound with <strong>honey</strong> and EUSOL<br />

dressing, Journal <strong>of</strong> Alternative and Complementary Medicine, 11 (3), pp,<br />

511-513.<br />

� Okhiria, O. A., Henriques, A. F., Burton, N. F., Peters, A. and Cooper R. A.<br />

(2009) Honey modulates bi<strong>of</strong>ilms <strong>of</strong> Pseudomon<strong>as</strong> aeruginosain a time and<br />

dose dependent manner, Journal <strong>of</strong> ApiProduct and ApiMedical Science, 1<br />

(1), pp, 6- 10.<br />

� Paterson, D. L. and Bonomo, R. A. (2005) Extended spectrum β-lactam<strong>as</strong>e: a<br />

clinical update, Clinical Microbiology Reviews, 18 (4), pp, 657-686.<br />

� Patrzykat, A., Friedrich, C. L., Zhang, L., Mendoza, V. and Hancock, R.E.<br />

(2002) Sublethal Concentrations <strong>of</strong> Pleurocidin-Derived Antimicrobial<br />

Peptides Inhibit Macromolecular Synthesis in Escherichia coli, Antimicrobial<br />

Agent and Chemotherapy, 46 (3), pp, 605-614. DOI:<br />

10.1128/AAC.46.3.605–614.2002.<br />

263


� Patton, T., Barrett, J., Bernnan, J. and Moran, N. (2006) Use <strong>of</strong> a<br />

spectrophotometric bio<strong>as</strong>say for determination <strong>of</strong> microbial sensitivity to<br />

manuka <strong>honey</strong>, Journal <strong>of</strong> Microbiological Methods, 64, pp, 84-95.<br />

� Perez, F., Hujer, A. M., Hujer, K. M., Decker, B. K., Rather, P.N and<br />

Bonomo, R.A. (2007) The Global Challenge <strong>of</strong> Multidrug Resistant (MDR)<br />

Acinetobacter baumannii, Antimicrobial Agents and Chemotherapy, 51 (10),<br />

pp, 3471–3484, doi:10.1128/AAC.01464-06.<br />

� Poirel, L., Al-M<strong>as</strong>kari, Z., Al-R<strong>as</strong>hdi, F., Bernabeu, S. and Nordmann, P.<br />

(2010) NDM-1- producing Klebsiella pneumoniae isolated in the Sultanate <strong>of</strong><br />

Oman, Journal <strong>of</strong> Antimicrobial Chemotherapy, doi:10.1093/jac/dkq428.<br />

� Pyrznska, K. and Biesaga, M. (2009) Analysis <strong>of</strong> phenolic acid and<br />

flavonoids in <strong>honey</strong>, Trends in Analytical Chemistry, 28 (7), pp, 893-902.<br />

� Russell, K. M., Molan, P. C., Wilkins, A. L. and Holland, P. T. (1988) The<br />

identification <strong>of</strong> some antibacterial constituents <strong>of</strong> New Zealand manuka<br />

<strong>honey</strong>, Journal <strong>of</strong> Agriculture Food Chemistry, 38, pp, 10-13.<br />

� Sajwani, A., Farooq, S. A., Eltayeb, E. A. and Bryant, V. (2007a)<br />

Melissopalynological studies from Oman, Palynology, 31, pp, 63-79.<br />

� Sajwani, A., Eltayeb, E. A., Farooq, S. A., and Patzelt, A. (2007b) Sugar and<br />

protein pr<strong>of</strong>iles <strong>of</strong> Omani <strong>honey</strong> from Muscat and Batina regions <strong>of</strong> Oman,<br />

International Journal <strong>of</strong> Food Properties, 10, pp, 675-690.<br />

� Sammataro, D. and Avitabile, A. (1998) The beekeeper’s handbook, third<br />

edition, Cornell <strong>University</strong> Press, USA.<br />

� Sanz, M.L., del C<strong>as</strong>tillo, M.D., Corzo, N. and Olano, A. (2003) 2-<br />

Furoylmethyl amino acids and hydroxymethylfurfural <strong>as</strong> indicators <strong>of</strong> <strong>honey</strong><br />

quality, Journal <strong>of</strong> Agriculture Food Chemistry, 51 (15), pp, 4278-4283.<br />

264


� Scholar, E. M. and Pratt, W. B. (2000) The Antimicrobial Drugs, Second<br />

edition, Oxford <strong>University</strong> press, UK.<br />

� Schumacher, H. H. (2004) Use <strong>of</strong> medical <strong>honey</strong> in patients with chronic<br />

venous leg ulcers after split-skin grafting. Journal <strong>of</strong> Wound Care, 13, pp,<br />

451-452.<br />

� Sefton, A. M. (2002) Mechanisms <strong>of</strong> <strong>antimicrobial</strong> resistance: Their clinical<br />

relevance in the new millennium, Drugs, 62, pp, 557-566<br />

� Sherlock, O., Athman, D. A., Power, D. A., Gethin. G., Cowman, S, et al.<br />

(2010) Comparison <strong>of</strong> the <strong>antimicrobial</strong> activity <strong>of</strong> ulmo <strong>honey</strong> from Chile<br />

and manuka <strong>honey</strong> against methicillin-resistant Staphylococcus aureus,<br />

Escherichia coli and Pseudomon<strong>as</strong> aeruginosa, BMC Complement Alternat<br />

Med, 10, pp, 47.<br />

� Shimoda, M., OHKI, K., Shimamoto, Y. and Koh<strong>as</strong>hi, O. (1995)<br />

Morphology <strong>of</strong> Defensin-Treated Staphylococcus aureus, Infection and<br />

Immunology, 63 (8), pp, 2886-2891.<br />

� Shriv<strong>as</strong>tava, S. M., Kumar, S. and Chaudhary, M. (2009) Time-kill curve<br />

studies <strong>of</strong> Ampucare against Escherichia coli, Staphylococcus aureus,<br />

Klebsiella pneuminiae and Proteus vulgaris, Reseach Journal <strong>of</strong> medicinal<br />

plant, ISSN 1819-3455<br />

� Silberman, S. (2007) The invisible enemy, WIRED, 15(2), on-line edition<br />

� Singleton, P. (2004) Bacteria, in Biology, Biotechnology and Medicine, Sixth<br />

edition, John Wiley & Sons, Ltd, Spain.<br />

� Simon, A., S<strong>of</strong>ka, K.,Wiszniewsky, G., et al. (2005) Wound care with<br />

antibacterial <strong>honey</strong> (Medi<strong>honey</strong>) in pediatric hematology-oncology, Support<br />

Care Cancer<br />

265


� Snow, M. J. and Harris, M. M. (2003) Analytical, Nutritional and Clinical<br />

Methods On the nature <strong>of</strong> non-peroxide antibacterial activity in New Zealand<br />

manuka <strong>honey</strong>, Food Chemistry, 84 (1), pp, 145-147<br />

� Song, Y., Yang, Q., Kong, H. and Zhong, B. (2004) Typing and<br />

characterization <strong>of</strong> carbapenem resistance Acinetobacter calcoaceticus-<br />

baumannii complex in a Chinese hospital, Journal <strong>of</strong> Medical Microbiology,<br />

53, pp, 653-656.<br />

� Stephen-Haynes, J. (2004) Evaluation <strong>of</strong> <strong>honey</strong>-impregnated tulle dressing in<br />

primary care, British Journal <strong>of</strong> Community Nursing, 9 (6) Suppl, S21-27.<br />

� Stephens, J. M., Schlothauer, R. C., Morris, B. D., Yang, D., Fearnley, L. and<br />

Loomes, K. M. (2009) Phenolic compounds and methylglyoxal in some New<br />

Zealand manuka and Kanuka <strong>honey</strong>s, Food Chemistry, 120 (1), pp, 78-86.<br />

� Subrahmanyam, M. (1993) Honey impregnated gauze versus polyurethane<br />

film (OpSite®) in the treatment <strong>of</strong> burns-a prospective randomised study,<br />

Burn Journal <strong>of</strong> Pl<strong>as</strong>tic Surgry, 46(4) pp, 322-323.<br />

� Subrahmanyam, M. (1994) Honey-impregnated gauze versus amniotic<br />

membrane in the treatment <strong>of</strong> burns, Burns, 20(4), pp, 331-333.<br />

� Subrahmanyam, M. (1996) Addition <strong>of</strong> antioxidants and polyethylene glycol<br />

4000 enhances the healing property <strong>of</strong> <strong>honey</strong> in burns, Annual Burns Fire<br />

Dis<strong>as</strong>ters, 9(2), pp, 93-95.<br />

� Subrahmanyam, M. (1996) Honey dressing versus boiled potato peel in the<br />

treatment <strong>of</strong> burns: a prospective randomized study, Burns, 22(6), pp, 491-<br />

493.<br />

266


� Subrahmanyam, M. (1998) A prospective randomised clinical and<br />

histological study <strong>of</strong> superficial burn wound healing with <strong>honey</strong> and silver<br />

sulfadiazine, Burns, 24(2), pp, 157-161.<br />

� Subrahmanyam, M., Hemmady. A., Pawar, S. G. (2001) Antibacterial<br />

activity <strong>of</strong> <strong>honey</strong> on bacteria isolated from wounds, Annals <strong>of</strong> Burns and Fire<br />

Dis<strong>as</strong>ters, XIV<br />

� Subrahmanyam, M., Shahapure, A. G., Nagane, N.S., Bhagwat, V. R. and<br />

Ganu, J. V. (2003) Free radical control-the main mechanism <strong>of</strong> the action <strong>of</strong><br />

<strong>honey</strong> in burns, Annals <strong>of</strong> Burns and Fire Dis<strong>as</strong>ter, XVI, pp, 135–137.<br />

� Subrahmanyam, M. (2007) Topical application <strong>of</strong> <strong>honey</strong> for burn wound<br />

treatment-An overview, Annals <strong>of</strong> Burns and Fire Dis<strong>as</strong>ters, XX (3).<br />

� Sussman, M. (ed.) Molecular medical microbiology, Vol 2, Cambridge<br />

<strong>University</strong> press, UK.<br />

� Tan, H. T., Abdul Rahman, R., Gan, S. H., Halim, A. S., H<strong>as</strong>san, A.,<br />

Sulaiman, A. and Kaur, K. (2009) The antibacterial properties <strong>of</strong> Malaysian<br />

tualang <strong>honey</strong> against wound and enteric microorganisms in comparison to<br />

manuka <strong>honey</strong>, BMC Complementary and Alternative Medicine, 9, pp, 34<br />

� Terrab, A., Diez, M. J. and Heredia, F. J. (2002a) Chromatic charactrisation<br />

<strong>of</strong> Moroccan <strong>honey</strong>s by diffuse reflectance tristimulus colorimetry-Non<br />

uniform and uniform colour spaces, Food Sciences Tech Int, 8, pp, 189-195.<br />

� Tonks, A. J., Cooper, R. A., Jones, K. P., et al. (2003) Honey stimulates<br />

inflammatory cytokine production from monocytes, Cytokine, 21(5), pp, 242-<br />

247.<br />

267


� Torres, M. V., Munoz, P. and Bouza, E. (2007) Hospital-Acquired<br />

Pneumonia, in Gould, I. M. and Van Deer Meer, J. W. M. (ed.) Antibiotic<br />

policies: fighting resistance, Springer Sciences<br />

� Towner, K. J. (1997) Clinical importance and antibiotic resistance <strong>of</strong><br />

Acinetobacter spp, Journal <strong>of</strong> Medical Microbiology, 46, pp, 721-746.<br />

� Towner, K. J. (2002) Acinetobacter, in Sussman, M. (ed.) Molecular medical<br />

microbiology, Vol 2, Cambridge <strong>University</strong> press, UK.<br />

� Townsend, G. F. (1969) Optical density <strong>as</strong> a means <strong>of</strong> colour cl<strong>as</strong>sification <strong>of</strong><br />

<strong>honey</strong>, Journal Apic. Research, 8 (1), pp, 29-36.<br />

� Tur, E., Bolton, L. and Constantine, B. E. (1995) Topical hydrogen peroxide<br />

treatment <strong>of</strong> ischemic ulcers in the guinea pig: Blood recruitment in multiple<br />

skin sites, Journal Am Academic Dermatology, 33 (2) pt 1, pp, 217-221<br />

� Turkmen, N., Sari, F., Poyrazoglu, E. S. and Velioglu, Y. S. (2006) Effect <strong>of</strong><br />

prolonged heating on antioxidant activity and colour <strong>of</strong> <strong>honey</strong>, Food<br />

Chemistry, 95 (4), pp, 653-657.<br />

� Van den Berg, A. J., Van den Worm, E., Van Ufford, H. C., Halkes, S.B,<br />

Hoekstra, M. J. and Beukelman ,C. J. (2008) An in vitro examination <strong>of</strong> the<br />

antioxidant and anti-inflammatory properties <strong>of</strong> buckwheat <strong>honey</strong>, J Wound<br />

Care, 17 , pp, 172-78.<br />

� Vila, J. (1998) Mechanisms <strong>of</strong> <strong>antimicrobial</strong> resistance in Acinetobacter<br />

baumannii, Review <strong>of</strong> Medical Microbiology, 9, pp, 87–97.<br />

� Viuda-Martos, M., Ruiz-Navaj<strong>as</strong>, Y., Fernandez-Lopez, J. and Perez-<br />

Alvarez, J. A. (2008) Function properties <strong>of</strong> <strong>honey</strong>, propolis, and royal jelly,<br />

Food Sciences, 73 (9), pp, 117-124.<br />

268


� Wadi, M., Al-Amin, H., Farouq, A., K<strong>as</strong>hef, H. and Khaled, S. A. (1987)<br />

Sudanese bee <strong>honey</strong> in the treatment <strong>of</strong> suppurating wounds, Arab Medico,<br />

3, pp, 16-18.<br />

� Wahdan, H. A. (1998) Causes <strong>of</strong> the <strong>antimicrobial</strong> activity <strong>of</strong> <strong>honey</strong>,<br />

Infection, 26 (1), pp, 26-30.<br />

� Weston, R.J. (2000) The contribution <strong>of</strong> catal<strong>as</strong>e and other natural products<br />

to the antibacterial activity <strong>of</strong> <strong>honey</strong>: a review, Food Chemistry, 71, pp, 235 –<br />

239.<br />

� Weston, R. J., Mitchell, K. R. and Allen, K. L. (1999) Antibacterial phenolic<br />

components <strong>of</strong> New Zealand <strong>honey</strong>, Food Chemistry, 64, pp, 295-301.<br />

� Wheat, E. J. (2004) The antibacterial activity <strong>of</strong> Welsh <strong>honey</strong>s, Mphil,<br />

<strong>University</strong> <strong>of</strong> Wales Institute, Cardiff.<br />

� White, J. W. (1975) Composition <strong>of</strong> <strong>honey</strong>, in: Crane, E. (ed.) Honey: a<br />

Comprehensive Survey, London: Heinemann, pp, 157-206.<br />

� White, J. W. and Rudji, O. N. (1978) The protein content <strong>of</strong> <strong>honey</strong>, Journal<br />

Apic. Research, 17 (4), pp, 234-238.<br />

� Williams, D. B and Carter, C. B. (2009) Transmission electron microscopy. A<br />

text book for materials science, 2 nd edition, Springer.<br />

� Wilkinson, J. M. and Cavanagh, H. M. (2005) Antimicrobial activity <strong>of</strong> 13<br />

<strong>honey</strong>s against Escherichia coli and Pseudomon<strong>as</strong> aeruginosa, Journal <strong>of</strong><br />

Medicinal Food, 8 (1), pp, 100-103.<br />

� Wilson, M., McNab, R. and Hendeson, B. (2002) Bacterial dise<strong>as</strong>e<br />

mechanisms- An introduction to cellular microbiology, Cambridge<br />

<strong>University</strong> press, UK<br />

269


� Winn, W. C., Koneman, E. W., Allen, S. D., Janda, W. M., Schreckenberger,<br />

P. C., Procop, G. W. and Woods, G. L. (2006) Koneman’s Colour Atl<strong>as</strong> and<br />

text book <strong>of</strong> Diagnostic Microbiology, Sixth edition, Lippincott Williams &<br />

Wilkins, Philadelphia, USA.<br />

� Yao, L., Datta, N., Barberan, F. A., Ferreres, F., Martos, I. and Singanusong,<br />

R. (2003) Flavonoids, phenolic acids and abscisic acid in Australian and New<br />

Zealand Leptosperdium <strong>honey</strong>s, Food chemistry, 80.<br />

� Yong, D., Toleman, M. A., Giske, C. G., Cho, S. H., Sundman, K., Lee, K.<br />

and Walsh, T. R. (2009) Characterization <strong>of</strong> a new metallo-Lactam<strong>as</strong>e gene,<br />

blaNDM-1, and a novel erythromycin ester<strong>as</strong>e gene carried on a unique<br />

genetic structure in Klebsiella pneumoniae sequence type 14 from India,<br />

Antimicrobial Agent and Chemotherapy, 53(12), pp, 5046-5054.<br />

� Zong and Yu. (2010) Escherichia coli carrying the blaCTX-M-15 gene <strong>of</strong><br />

ST648, J Med Microbiol, 59, pp,1536-1537<br />

� Zumla, A. and Lulat, A. (1989) Honey- a remedy rediscovered, Journal <strong>of</strong><br />

the Royal Society <strong>of</strong> Medicine, 82, pp, 384-385<br />

270

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!