15.12.2012 Views

Honours Project Book - Faculty of Health Sciences - University of ...

Honours Project Book - Faculty of Health Sciences - University of ...

Honours Project Book - Faculty of Health Sciences - 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.

SCHOOL OF PAEDIATRICS AND<br />

REPRODUCTIVE HEALTH<br />

2013<br />

In Paediatrics and Child <strong>Health</strong>, Reproductive Biology and Regenerative Medicine


Research opportunities for <strong>Honours</strong><br />

students in 2013<br />

School <strong>of</strong> Paediatrics and<br />

Reproductive <strong>Health</strong><br />

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

International research leaders in Paediatrics & Child<br />

<strong>Health</strong>, Reproductive Biology & <strong>Health</strong> and<br />

Regenerative Medicine<br />

Contents<br />

Introduction .............................................................................................................................. 3<br />

Expression <strong>of</strong> Interest in an <strong>Honours</strong> <strong>Project</strong> ......................................................................... 6<br />

Scholarship Application Form ................................................................................................ 9<br />

<strong>Project</strong>s<br />

Discipline <strong>of</strong> Obstetrics and Gynaecology........................................................................ 11<br />

Discipline <strong>of</strong> Paediatrics ........................................................................................................ 48<br />

Index........................................................................................................................................ 74<br />

Last updated 5 November 2012<br />

2 |


Make a difference<br />

"The recent Excellence in Research Australia (ERA)<br />

results have rated the School <strong>of</strong> Paediatrics and<br />

Reproductive <strong>Health</strong> at the highest level (5/5), a feat<br />

unmatched by any other Paediatrics and<br />

Reproductive <strong>Health</strong> Department in Australia. Our<br />

School is internationally recognised for our research<br />

in Reproduction, Maternal and Child <strong>Health</strong>. Our<br />

graduates are highly sought after and have<br />

outstanding career opportunities. We invite you to<br />

take up one <strong>of</strong> the <strong>Honours</strong> research opportunities<br />

outlined in this booklet and join us in making a<br />

difference."<br />

Pr<strong>of</strong>essor Julie Owens,<br />

Head <strong>of</strong> School,<br />

School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong><br />

The School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong> invites you to join us for <strong>Honours</strong> study in 2013.<br />

Our School and its members have international reputations as research leaders in reproductive biology and<br />

medicine, maternal, fetal and child health, and reproductive and regenerative medicine. We are recognised for<br />

our strong links between cutting-edge discoveries in basic research with bridges through to clinical application and<br />

evaluation in evidence-based medicine. The School’s two Disciplines; Obstetrics & Gynaecology and Paediatrics<br />

include 300 staff and postgraduate students and over 200 clinical affiliates making our School one <strong>of</strong> the largest<br />

and most productive peadiatrics and reproductive health departments in the world. You have the chance to work,<br />

be educated and graduate from an internationally renowned School to the benefit <strong>of</strong> your future career.<br />

The School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong> consists <strong>of</strong> two disciplines, Paediatrics and Obstetrics and<br />

Gynaecology. The School hosts and leads one research institute, the Robinson Institute, consisting <strong>of</strong> four <strong>University</strong><br />

recognised and supported centres and one affiliated <strong>University</strong> research centre:<br />

• The Research Centre for Reproductive <strong>Health</strong><br />

• Early Origins <strong>of</strong> <strong>Health</strong> and Disease<br />

Centre for Stem Cell Research<br />

• The Australian Research Centre for the <strong>Health</strong> <strong>of</strong> Women and Babies<br />

• The affiliated Children’s Research Centre<br />

Our five major locations are in the Medical School, Women’s and Children’s Hospital, Lyell McEwin Hospital, Royal<br />

Adelaide Hospital and Robinson Institute on King William Road at North Adelaide. Students can also study with our<br />

affiliates at these locations and other including the CSIRO and Flinders Medical Centre.<br />

We are an extremely research-active School, with major project funding from the National <strong>Health</strong> & Medical<br />

Research Council, Australian Research Council and the National Institute <strong>of</strong> <strong>Health</strong> (US) amongst others. We host 12<br />

NHMRC Senior Fellows, 2 ARC Future Fellows and numerous Training and Career Development Fellows. Members <strong>of</strong><br />

our School published over 400 peer-reviewed articles in 2011 in the top journals in their fields, including Nature, the<br />

Cochrane Database <strong>of</strong> Systemic Reviews, Endocrine Reviews, Journal <strong>of</strong> Clinical Endocrinology & Metabolism,<br />

Journal <strong>of</strong> Paediatrics & Child <strong>Health</strong>, Lancet, Nature Genetics, New England Journal <strong>of</strong> Medicine, Obstetrics &<br />

Gynaecology and Theriogenology.<br />

This booklet contains information about our <strong>Honours</strong> course, enrolment and available scholarships, as well as a<br />

listing <strong>of</strong> the <strong>Honours</strong> research projects we are <strong>of</strong>fering for study. Please remember that the research groups within<br />

the School and its affiliates may also have additional projects available, and <strong>of</strong>ten projects can be adapted to suit<br />

the interests and skills <strong>of</strong> the student. You should use the outlines in this book as a starting point in developing your<br />

project with a supervisor.<br />

We invite you to join us for <strong>Honours</strong> and develop your career.<br />

3 |


Eligibility and study options<br />

Students who have completed an undergraduate degree in a science course, including B Science, B Science<br />

(Biomedical Science), B <strong>Health</strong> Science, B Biotechnology, B Agricultural Science, or B Animal Science, or who have<br />

completed 3 years or more <strong>of</strong> the MBBS program, are invited to apply for <strong>Honours</strong> positions within our School. To be<br />

eligible for <strong>Honours</strong>, a candidate must have completed the requirements for a Bachelor degree or equivalent to a<br />

standard that is acceptable to the <strong>Faculty</strong> for the purposes <strong>of</strong> admission into the <strong>Honours</strong> program. For more<br />

information please visit:<br />

http://health.adelaide.edu.au/programs/honours/<br />

A credit average within relevant subjects is the usual standard expected, but students with a strong interest in a<br />

particular field and the support <strong>of</strong> their proposed supervisor/s are also encouraged to apply, and <strong>of</strong>ten find their<br />

<strong>Honours</strong> year very stimulating and successful.<br />

<strong>Honours</strong> students are invited to enrol either full-time (1 year <strong>of</strong> study), or part-time and study over two/three years.<br />

The main <strong>Honours</strong> intake is in February, and students may also commence mid-year.<br />

Facilities and training opportunities<br />

<strong>Honours</strong> students have access to the core facilities <strong>of</strong> the <strong>University</strong> and School as required for their specific projects.<br />

These include state-<strong>of</strong>-the-art molecular biology and imaging equipment, confocal microscopes, large and small<br />

animal surgeries and in vivo facilities, cell culture laboratories and an embryology laboratory. Some projects have<br />

strong clinical components with patient involvement, epidemiological methodologies and genomic linkage. We<br />

also encourage our <strong>Honours</strong> students to participate in the School’s workshop series including areas such as<br />

bioinformatics, new molecular biology techniques and research ethics. Of course, <strong>Honours</strong> students will also get in<br />

depth training in their research field and its techniques within their research group throughout their research<br />

projects.<br />

Our <strong>Honours</strong> program<br />

Our program is designed for you to develop specific knowledge and skills in your chosen research field as well as<br />

general skills in working independently, critical reasoning, scientific writing and presentation. Our <strong>Honours</strong> students<br />

each undertake a full-year research project (or part-time equivalent), with the majority <strong>of</strong> assessments structured<br />

around this. In addition, our <strong>Honours</strong> structured program teaches <strong>Honours</strong> students the skills they need to succeed in<br />

research, including experimental design and statistics, research ethics, an understanding <strong>of</strong> the scientific method,<br />

good record keeping and laboratory practice, literature searching and bibliographic s<strong>of</strong>tware and critical<br />

evaluation <strong>of</strong> papers during “journal club” tutorial discussions. The overall objective <strong>of</strong> the program is for students to<br />

demonstrate a deep understanding and interpretation <strong>of</strong> their subject area and the ability to clearly and<br />

thoroughly present the project.<br />

Aims <strong>of</strong> the <strong>Honours</strong> Program<br />

• To provide mentored training in scientific research methods.<br />

• To develop skills for independent critical thought and learning.<br />

• To develop logical reasoning and written and oral presentation skills.<br />

• To encourage interest in a broad range <strong>of</strong> child health and reproduction related science.<br />

One on one contact with the supervisor and other members <strong>of</strong> the research team is the main method by which<br />

students gain these skills during <strong>Honours</strong>. In addition, hands on experience, communication with other members <strong>of</strong><br />

the <strong>Honours</strong> class, the <strong>Honours</strong> coordinator, and the School is invaluable.<br />

Assessment<br />

The main component <strong>of</strong> the <strong>Honours</strong> course is the research project carried out with your supervisor/s. Four points <strong>of</strong><br />

assessment are designed to evaluate and provide feedback on student skills relating to research aptitude,<br />

knowledge <strong>of</strong> their field, as well as written and verbal presentation style.<br />

1. Critical Literature Review (3000 words 15%)<br />

Students discuss and critically appraise the 10-15 papers most salient to their project.<br />

2. Examination <strong>of</strong> Tutorial Course material (2 hours open book 15%, Tutorial Participation 5%)<br />

Problem/scenario based open book written exam that draws on the issues discussed in the tutorial course.<br />

3. Thesis (5000 words 50%)<br />

Demonstrates your ability to undertake scientific research and present your findings as a scientific manuscript.<br />

4. Final seminar (20 minutes 15%<br />

Oral presentation <strong>of</strong> the student research project.<br />

4 |


<strong>Honours</strong> graduate career pathways<br />

Our previous <strong>Honours</strong> students have gone on to successful careers in a wide range <strong>of</strong> roles. The skill base that<br />

<strong>Honours</strong> gives you is essential for those wanting to undertake postgraduate research training, and valuable across a<br />

wide range <strong>of</strong> areas. Approximately half <strong>of</strong> our students go on to further study, including PhD and postgraduate<br />

Medicine. Others are now employed as research assistants and <strong>of</strong>ficers, journal editors, patent attorneys and<br />

investment managers.<br />

Interested?<br />

The process to undertake <strong>Honours</strong> within the School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong> involves:<br />

Browse this booklet to determine a research area or project that interests you.<br />

Discuss potential projects with Supervisors at our annual <strong>Honours</strong> Information Session in August or contact<br />

them directly using the information provided in this booklet.<br />

If you have any questions about your eligibility, part-time study or other queries, you should contact:<br />

Dr Kathy Gatford<br />

<strong>Honours</strong> Coordinator<br />

School <strong>of</strong> Paediarics and Reproductive <strong>Health</strong><br />

Phone: (08) 8313 4158<br />

Email: kathy.gatford@adelaide.edu.au<br />

Discuss your intention to apply for the project with the appropriate supervisor and then complete the<br />

‘Expression <strong>of</strong> interest in <strong>Honours</strong>’ form on the following page.<br />

You will be notified via email <strong>of</strong> the outcome <strong>of</strong> your application by the School <strong>of</strong> Paediatrics and<br />

Reproductive <strong>Health</strong> <strong>Honours</strong> Coordinator following the release <strong>of</strong> any outstanding results. If your application<br />

is accepted you will receive an acceptance letter with application to enrol forms. You will be able to enrol<br />

following once enrolments open (usually late January for Semester 1 or late July for Semester 2). For more<br />

information regarding the enrolment process contact:<br />

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

Plaza Building, Level 2<br />

North Terrace Campus<br />

(08) 8313 5336<br />

Or visit the <strong>Faculty</strong> <strong>of</strong> <strong>Health</strong> <strong>Sciences</strong> website: http://www.health.adelaide.edu.au/programs/honours/<br />

“The option <strong>of</strong> undertaking an <strong>Honours</strong> degree on a full-time (one year)<br />

or part-time (two year) basis as a medical student is becoming<br />

increasingly popular. The honours programme is engaging and<br />

rewarding in gaining thorough 'ground-level' understanding <strong>of</strong> the<br />

principles <strong>of</strong> good clinical research practice. The School <strong>of</strong> Paediatrics<br />

and Reproductive <strong>Health</strong> has been very supportive and flexible with a<br />

fantastic series <strong>of</strong> statistics and research tutorials. I would encourage<br />

those interested in exploring the exciting prospects <strong>of</strong> honours in the<br />

School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong> to seek helpful guidance<br />

from Dr Kathy Gatford as soon as possible”<br />

Brenton Systermans & Tom Cundy (completed <strong>Honours</strong> 2010, School <strong>of</strong><br />

Paediatrics and Reproductive <strong>Health</strong>).<br />

Tom has since been awarded a $150,000 2011 General Sir John Monash<br />

scholarship to study for his PhD in robotic surgery.<br />

5 |


Expression <strong>of</strong> Interest in an <strong>Honours</strong> <strong>Project</strong><br />

Personal Details<br />

Student ID Name<br />

Address Postcode<br />

Phone Email<br />

Alternate contact details (if we need to contact you at a different place/number/ e-mail):<br />

Dates to use alternate contact details Start date End date<br />

Address Country<br />

Telephone Email<br />

Candidature Details<br />

Undergraduate degree<br />

Citizenship (Tick one)<br />

Australian citizen<br />

New Zealand citizen<br />

permanent resident<br />

I will hold a visa whilst studying honours<br />

(international students)<br />

Visa type: .........................................................<br />

Start date (Year/Semester) 20 ...... Semester 1 Semester 2<br />

Enrolment load Full time Part time<br />

<strong>Honours</strong> <strong>Project</strong> Details<br />

Title<br />

Discipline (Tick one) Paediatrics Obstetrics & Gynaecology<br />

Supervision details<br />

Primary<br />

Supervisor<br />

Location<br />

Email Phone<br />

Signature Date<br />

Cosupervisor Email<br />

ATTACH a copy <strong>of</strong> your current academic transcript when you return this form. If you have completed<br />

undergraduate studies at an institution other than the <strong>University</strong> <strong>of</strong> Adelaide you will also need to<br />

provide an <strong>of</strong>ficial copy <strong>of</strong> your transcript once your final marks are available.<br />

DUE DATE: 4 November for first round Semester 1 <strong>of</strong>fers <strong>of</strong> the following year.<br />

18 June for mid year <strong>of</strong>fers (Semester 2 <strong>of</strong> the same year).<br />

Forms will still be accepted after these dates but will be treated on an ad hoc basis.<br />

SUBMIT via: Email: sprh.l_and_t@adelaide.edu.au<br />

In person/ by post to: <strong>Honours</strong> Coordinator, School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong><br />

Level 3, Medical School South, Frome Road, <strong>University</strong> <strong>of</strong> Adelaide, Adelaide SA 5005.<br />

6 |


<strong>Honours</strong> Scholarships and Support<br />

Scholarships are available to support full time <strong>Honours</strong> study in the School <strong>of</strong> Paediatrics and<br />

Reproductive <strong>Health</strong>. Different projects will be eligible for different scholarships, and these<br />

are indicated by the following symbols next to the project description:<br />

Robinson Institute .R.<br />

Discipline <strong>of</strong> Obstetrics and Gynaecology O&G.<br />

Eligibility for scholarships<br />

Applicants must:<br />

▪ be enrolled in their <strong>Honours</strong> degree full time.<br />

▪ be Australian citizens, permanent residents <strong>of</strong> Australia or hold a suitable Visa<br />

▪ have completed an undergraduate degree in a relevant area before scholarship<br />

commencement<br />

▪ meet the specific requirements for eligibility for specific scholarships (see below)<br />

<strong>Honours</strong> scholarship conditions:<br />

The following conditions apply to all <strong>Honours</strong> scholarships awarded through our School:<br />

1. The award <strong>of</strong> the scholarship is subject to acceptance into an <strong>Honours</strong> program at<br />

the <strong>University</strong> <strong>of</strong> Adelaide through the School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong><br />

and commencement <strong>of</strong> the project within 12 months <strong>of</strong> approval.<br />

2. Awardees may only hold one <strong>Honours</strong> scholarship funded from within The <strong>University</strong> <strong>of</strong><br />

Adelaide.<br />

3. The awardee must complete their <strong>Honours</strong> degree. Should the awardee withdraw<br />

from the <strong>Honours</strong> program the scholarship will be cancelled and all scholarship<br />

monies are repayable to the sponsor (i.e. Discipline, Robinson Institute, Children’s<br />

Research Centre or <strong>Project</strong> supervisor).<br />

4. You must notify the project sponsor in writing if you:<br />

a. discontinue your studies<br />

b. cease to study or cease to be enrolled<br />

c. change your course <strong>of</strong> study<br />

d. change your address<br />

Selection<br />

The Scholarships selection committee will review applications, considering the following:<br />

▪ eligibility criteria<br />

▪ student Grade Point Average (GPA)<br />

▪ the strength <strong>of</strong> the potential <strong>Honours</strong> project<br />

▪ endorsement from the potential supervisor<br />

The selection committee may choose not to award scholarships based on the quality <strong>of</strong><br />

applications.<br />

Discipline <strong>of</strong> Obstetrics and Gynaecology <strong>Honours</strong> Scholarships<br />

The Discipline <strong>of</strong> Obstetrics and Gynaecology is <strong>of</strong>fering up to three <strong>Honours</strong> scholarships,<br />

each valued at $4000 pa, for <strong>Honours</strong> students enrolled through our Discipline. These<br />

scholarships will be awarded to students who have a Disctiction average in their previous<br />

undergraduate studies. The Discipline <strong>of</strong> Obstetrics and Gynaecology also provide<br />

dedicated computers for <strong>Honours</strong> student use (one computer per two students) to facilitate<br />

your studies.<br />

<strong>Project</strong>s eligible for these scholarships are marked with O&G.<br />

7 |


Robinson Institute <strong>Honours</strong> Scholarships<br />

2011 Jeffrey Robinson <strong>Honours</strong><br />

Scholarship Recipent<br />

The Jeffrey Robinson <strong>Honours</strong> Scholarship<br />

The purpose <strong>of</strong> The Jeffrey Robinson <strong>Honours</strong> Scholarship is to attract high quality students to<br />

the research programs <strong>of</strong> The Robinson Institute and its constituent centres. Emeritus Pr<strong>of</strong>essor<br />

Jeffrey Robinson, born in Northern Ireland, was appointed Pr<strong>of</strong>essor <strong>of</strong> Obstetrics and<br />

Gynaecology at the <strong>University</strong> <strong>of</strong> Adelaide in 1986. Under his leadership the <strong>University</strong><br />

developed its outstanding reputation for research into fetal-maternal physiology and<br />

reproductive medicine and biology. In 2006 he was awarded a Commander <strong>of</strong> the British<br />

Empire (CBE) for his services to maternal and fetal heath. Jeffrey continues to be involved in<br />

research at The Robinson Institute.<br />

The Robinson Institute will be awarding one Jeffrey Robinson Scholarship to an exceptional<br />

student. The successful applicant will:<br />

▪ Undertake a research project within a successful research team<br />

▪ Receive a tax-free scholarship to the value <strong>of</strong> $7,000<br />

▪ Be enrolled in The Robinson Institute <strong>Honours</strong> Mentoring Program, which provides<br />

students one-on-one coaching with our leading researchers across areas <strong>of</strong><br />

reproductive health and regenerative medicine.<br />

<strong>Project</strong>s eligible for these scholarships are marked with .R.<br />

The Robinson Institute brings together over 400<br />

cutting edge researchers in the areas <strong>of</strong><br />

reproductive health, the health <strong>of</strong> women and<br />

babies, origins <strong>of</strong> health <strong>of</strong> disease and stem cell<br />

research.<br />

By focusing on the earliest stages <strong>of</strong> life, the<br />

Robinson Institute and its centres are looking at<br />

preventing disease and promoting health in<br />

children and adults across generations. The Institute<br />

bridges the gap between research discoveries and<br />

medical practice. Many <strong>of</strong> the Institute's senior<br />

researchers are leading clinicians in their fields,<br />

enabling a strong translation <strong>of</strong> research<br />

discoveries, with immeasurable benefits, into the<br />

community.<br />

As a member <strong>of</strong> the Robinson Institute you are<br />

eligible to apply for the Jeffrey Robinson <strong>Honours</strong><br />

Scholarship. The Institute also <strong>of</strong>fers its members the<br />

opportunity to participate in mentoring and<br />

pr<strong>of</strong>essional development programs and gain<br />

research support, such as assistance in applying for<br />

awards and external scholarships.<br />

8 |


Discipline <strong>of</strong> Obstetrics and Gynaecology<br />

Scholarship Application Form<br />

APPLICANT DETAILS<br />

First name: Surname: Student no:<br />

Undergraduate Degree: Institution:<br />

Postal Address:<br />

Citizenship: (please circle)<br />

Australian citizen New Zealand citizen Permanent Resident Visa Holder<br />

Email: Phone: Mobile:<br />

PROJECT DETAILS<br />

Proposed <strong>Honours</strong> <strong>Project</strong> Title:<br />

Brief Description: (250 word maximum)<br />

PRIMARY SUPERVISOR<br />

Email: Phone:<br />

Location:<br />

Signature: Date:<br />

Co-supervisor: Signature:<br />

9 |


Scholarship/s being applied for: (Please tick appropriate box/es)<br />

Robinson Institute .R.<br />

Discipline <strong>of</strong> Obstetrics and Gynaecology O&G.<br />

Please explain how this research project aligns to the Discipline, Robinson Institute or Research Centre<br />

strategic research objectives. (250 word maximum).<br />

Declaration<br />

I declare to the best <strong>of</strong> my knowledge and belief that the information I have supplied in this application is correct<br />

and complete.<br />

I understand that selection for these <strong>Honours</strong> scholarships is competitive and not all applicants who meet the<br />

basic eligibility criteria are necessarily awarded scholarships. I have read and understand the conditions <strong>of</strong> these<br />

scholarships as outlined in the School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong> ‘<strong>Honours</strong> Research Opportunities<br />

booklet’. I recognise that it is my responsibility to provide all necessary documentation and applications with<br />

incomplete documentation cannot be considered.<br />

Applicant signature: Date:<br />

ATTACH to this application:<br />

1. A cover letter.<br />

2. Your ‘Expression <strong>of</strong> interest in <strong>Honours</strong>’ form and transcript.<br />

3. A one page current CV.<br />

DUE DATE: 4 November, 2012.<br />

SUBMIT via: Email: sprh.l_and_t@adelaide.edu.au<br />

OR<br />

In person/ by post to:<br />

<strong>Honours</strong> Coordinator<br />

School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong><br />

Level 3, Medical School South, Frome Road<br />

<strong>University</strong> <strong>of</strong> Adelaide, Adelaide SA 5005<br />

10 |


Discipline <strong>of</strong> Obstetrics and Gynaecology<br />

Medical School<br />

Women’s and Children’s Hospital<br />

Lyell McEwin Hospital<br />

Royal Adelaide Hospital<br />

The Robinson Institute<br />

“The Discipline <strong>of</strong> Obstetrics and Gynaecology is a<br />

leading centre <strong>of</strong> academic research with an<br />

international reputation in the areas <strong>of</strong> maternal-fetal<br />

physiology, circadian physiology, reproductive<br />

immunology, embryology, advanced reproductive<br />

technology, feto-maternal medicine, the fetal origins<br />

<strong>of</strong> adult disease, perinatology, placentology, the<br />

genomics <strong>of</strong> pregnancy disorders e.g. preterm<br />

delivery, regenerative medicine and stem cell<br />

research, pre-eclampsia and cerebral palsy,<br />

urogynaecology and postmenopausal health . We<br />

are the largest and most research-active Discipline <strong>of</strong><br />

Obstetrics and Gynaecology in the world and our<br />

range <strong>of</strong> interests from basic science through to<br />

clinical practice creates multiple and successful<br />

career paths for our graduates.”<br />

Dr Paul Duggan<br />

Head <strong>of</strong> Discipline <strong>of</strong> Obstetrics and Gynaecology<br />

11 |


The Australian Research Centre for <strong>Health</strong> <strong>of</strong> Women and Babies<br />

Supervisors (photographs left to right)<br />

Pr<strong>of</strong>essor Caroline Crowther<br />

Pr<strong>of</strong>essor Jodie Dodd<br />

Philippa Middleton<br />

Emeritus Pr<strong>of</strong>essor Jeffrey Robinson<br />

Associate Pr<strong>of</strong>essor Ross Haslam<br />

Dr Andrew McPhee<br />

Dr Rosalie Grivell<br />

Dr Chad Andersen<br />

Dr Helena Oakey<br />

Associate Pr<strong>of</strong>essor Dominic Wilkinson<br />

Dr Lisa Yelland<br />

Dr Thach Son Tran<br />

Pr<strong>of</strong>essor Alastair MacLennan<br />

Dr Catherine Gibson<br />

Dr Michael O’Callaghan<br />

Dr Lisa Moran<br />

Dr Yee Khong<br />

Location: Women’s and Children’s Hospital<br />

The Australian Research Centre for <strong>Health</strong> <strong>of</strong> Women and Babies (ARCH) conducts high quality and timely maternal<br />

and perinatal research encompassing the spectrum from preconception through pregnancy and childbirth, infancy<br />

and later life through its six research divisions www.adelaide.edu.au/arch:<br />

Research Synthesis<br />

o Through the Research Synthesis Division we conduct, promote and support the preparation and<br />

updating <strong>of</strong> high quality systematic reviews <strong>of</strong> the existing evidence on questions <strong>of</strong> relevance to<br />

women and babies in Australia, regionally in South East Asia, and internationally.<br />

• Clinical Studies and Trials<br />

o Within the Clinical Studies and Trials Division we focus our research around nine key themes; Preterm<br />

Birth, Obesity, Diabetes, Maternal and Fetal Medicine, Term Birth, Multiple Pregnancy, Cerebral Palsy,<br />

Obstetric Medicine and Perinatal Ethics and Decision Making.<br />

• Translational Research<br />

o Our Translation Research <strong>Health</strong> Division focuses on promoting evidence-based practice in women’s<br />

and babies’ health by the dissemination and implementation <strong>of</strong> clinical research findings into clinical<br />

practice. We are achieving this by participating in guideline development, guideline methods and<br />

conducting implementation studies.<br />

• Indigenous Maternal and Perinatal <strong>Health</strong><br />

o Through our Indigenous Maternal and Perinatal <strong>Health</strong> Division we continue to maintain and build our<br />

collaborations with indigenous organisations, contribute to policy knowledge and endeavour to seek<br />

collaborative research and evaluation opportunities.<br />

• Research Networks and Education<br />

o We continue to provide high pr<strong>of</strong>ile research opportunities by identifying research gaps and defining<br />

research questions <strong>of</strong> major importance in maternal and perinatal health through our Research<br />

Networks and Education Division.<br />

• International Maternal and Perinatal <strong>Health</strong><br />

o Within the International Maternal and Perinatal <strong>Health</strong> Division we have widened our international<br />

network <strong>of</strong> collaborators through conducting individual participant data (IPD) meta-analyses. An<br />

integral component is the formation <strong>of</strong> an international collaborative group <strong>of</strong> trialists where<br />

researchers provide individual data from their trials possibly creating new knowledge not available<br />

through aggregate meta-analysis.<br />

12 |


Clinical Studies and Trials<br />

The Obesity Study Group is focussed on investigating<br />

whether lifestyle factors can reduce the risks<br />

associated with obesity in pregnancy.<br />

1: Assessment <strong>of</strong> the maternal fatty acid pr<strong>of</strong>ile<br />

during pregnancy and relationship to depression,<br />

anxiety and quality <strong>of</strong> life during pregnancy and<br />

post-partum.<br />

PROJECT: (Clinical) The relationship <strong>of</strong> depression and<br />

anxiety with n-3 long-chain polyunsaturated fatty<br />

acids during pregnancy and post-partum.<br />

R. O&G.<br />

Supervisors<br />

Dr Lisa Moran<br />

Pr<strong>of</strong>essor Jodie Dodd<br />

Pr<strong>of</strong>essor Maria Makrides<br />

Pr<strong>of</strong>essor Robert Gibson<br />

Contact Person<br />

Lisa Moran<br />

Phone: 8313 1352<br />

lisa.moran@adelaide.edu.au<br />

http://www.adelaide.edu.au/directory/lisa.moran<br />

<strong>Project</strong> Background/Aims<br />

Overweight and obesity are recognised risk factors<br />

for depression, anxiety and other psychological<br />

conditions. Although pregnancy is <strong>of</strong>ten thought <strong>of</strong><br />

as a time <strong>of</strong> emotional well-being, longitudinal studies<br />

have demonstrated that for many women,<br />

pregnancy is associated with increased levels <strong>of</strong><br />

depression and anxiety, including the onset or<br />

relapse <strong>of</strong> major depressive illness. Depression is<br />

estimated to affect approximately 9% <strong>of</strong> women in<br />

the antenatal period, increasing to 16% in the postpartum<br />

period, although a far greater proportion <strong>of</strong><br />

women commonly report negative mood during over<br />

the course <strong>of</strong> their pregnancy. Both obstetric and<br />

neonatal complications associated with maternal<br />

depression are well described, with more extreme<br />

adverse events including suicide and infanticide. The<br />

relative contribution <strong>of</strong> factors including diet and<br />

physical activity to psychological function during<br />

pregnancy and the post-partum period is unclear.<br />

Psychological wellbeing may be associated with the<br />

long-chain n-3 polyunsaturated fatty acids (n-3 LC<br />

PUFA), specifically the omega-3 fatty acids<br />

docosahexaenoic acid (DHA) and eicoapentaenoic<br />

acid (EPA) with some research indicating DHA<br />

deficiency may be related to depressive symptoms.<br />

Furthermore, a depletion <strong>of</strong> maternal plasma DHA<br />

also occurs during pregnancy and persists following<br />

birth and higher intake <strong>of</strong> n-3 LCPUFA from fish and<br />

seafood during pregnancy are associated with a<br />

reduced risk <strong>of</strong> postnatal depressive symptoms.<br />

However, the effect <strong>of</strong> maternal overweight and<br />

obesity on risk <strong>of</strong> psychological function including<br />

depression, anxiety and quality <strong>of</strong> life and the relative<br />

contribution <strong>of</strong> dietary factors including n-3 LCPUFA<br />

has been poorly documented to date.<br />

<strong>Project</strong>s available<br />

1: Assessment <strong>of</strong> association between n-3 LC PUFA<br />

from maternal blood, cord blood and dietary<br />

questionnaires with psychological function<br />

(depression, anxiety and quality <strong>of</strong> life) in overweight<br />

and obese women during pregnancy and the postpartum<br />

period.<br />

References<br />

Jans LA, Giltay EJ, Van der Does AJ. The efficacy <strong>of</strong> n-<br />

3 fatty acids DHA and EPA (fish oil) for perinatal<br />

depression. Br J Nutr 2010; 104:1577–1585.<br />

Bennett HA, Einarson A, Taddio A, Koren G, Einarson<br />

TR. Prevalence <strong>of</strong> depression during pregnancy:<br />

systematic review. Obstet Gynecol. 2004;103(4):698-<br />

709.<br />

2: Assessment <strong>of</strong> an antenatal lifestyle intervention<br />

aimed at preventing excessive gestational weight<br />

gain in overweight and obese women.<br />

PROJECT (Clinical): The effect <strong>of</strong> optimising excessive<br />

gestational weight gain on risk factors for diabetes<br />

and cardiovascular disease, placental function and<br />

body composition in mothers and infants.<br />

R. O&G.<br />

Supervisors<br />

Dr Lisa Moran<br />

Pr<strong>of</strong>essor Jodie Dodd<br />

Lisa Moran<br />

Phone: 8313 1352<br />

lisa.moran@adelaide.edu.au<br />

http://www.adelaide.edu.au/directory/lisa.moran<br />

<strong>Project</strong> Background/Aims<br />

Overweight and obesity are present in over 50% <strong>of</strong><br />

Australian women and are associated with an<br />

increased risk <strong>of</strong> obesity-associated metabolic<br />

diseases such as cardiovascular disease and<br />

diabetes. Furthermore, maternal obesity and excess<br />

weight gain during pregnancy are associated with<br />

worsened maternal health outcomes including<br />

gestational diabetes, hypertensive conditions and<br />

pre-eclampsia, infection, thromboembolic events,<br />

need for induction <strong>of</strong> labour, caesarean section,<br />

perinatal death, preterm birth and high birth weight<br />

and an increased risk <strong>of</strong> diabetes and cardiovascular<br />

disease after pregnancy. Infants <strong>of</strong> mothers who are<br />

overweight or obese are more likely to be<br />

macrosomic, require admission to the neonatal<br />

intensive care unit, be born preterm, have a<br />

congenital anomaly, and to require treatment for<br />

jaundice or hypoglycaemia. The effects <strong>of</strong> maternal<br />

obesity may also extend to deleterious effects on<br />

13 |


<strong>of</strong>fspring with regards to adiposity and<br />

cardiometabolic risk factors. In humans, there is an<br />

association between infant birth weight and being<br />

large for gestational age and risk for higher BMI and<br />

adverse cardiovascular risk pr<strong>of</strong>ile in childhood.<br />

Additionally, maternal factors including maternal<br />

obesity, obesity in pregnancy or high gestational<br />

weight gain are associated with risk for childhood or<br />

adult obesity and a worsened cardiovascular risk<br />

pr<strong>of</strong>ile in infancy. Optimising weight management<br />

during pregnancy is therefore crucial for both<br />

improving maternal and infant pregnancy outcomes<br />

and for reducing the risk <strong>of</strong> obesity and obesityassociated<br />

diseases such as diabetes or<br />

cardiovascular disease in mothers and children<br />

during pregnancy and following childbirth. These<br />

projects relate to a randomised controlled trial<br />

comparing standard antenatal care with an<br />

antenatal lifestyle intervention aimed at preventing<br />

excessive gestational weight gain in overweight and<br />

obese women (the LIMIT trial).<br />

<strong>Project</strong>s available<br />

1: Assessment <strong>of</strong> the effect <strong>of</strong> the LIMIT trial on risk<br />

factors for diabetes and cardiovascular disease in<br />

cord blood from infants at birth and maternal risk<br />

factors for diabetes and cardiovascular disease<br />

across pregnancy.<br />

2: Assessment <strong>of</strong> dietary intake, eating behaviour,<br />

physical activity and sedentary behaviour in children<br />

and parents at long-term follow-up (3 years) in LIMIT<br />

and the relationship with parental and childhood<br />

obesity.<br />

3: Evaluate the occurrence <strong>of</strong> structural inflammatory<br />

changes within the placenta among women who<br />

were overweight or obese, and to evaluate the<br />

effect <strong>of</strong> increasing body mass index on these<br />

changes.<br />

4: Increasing exercise and diet in overweight<br />

pregnant women: Validation <strong>of</strong> exercise and dietary<br />

questionnaire data with markers <strong>of</strong> cardiometabolic<br />

function (taken in early pregnancy, 28 weeks<br />

gestation and 36 weeks gestation).<br />

5: Effect <strong>of</strong> an antenatal dietary and lifestyle<br />

intervention to limit weight gain in pregnancy on<br />

neonatal growth and development.<br />

• Evaluation <strong>of</strong> neonatal body composition at<br />

birth as measured by birth weight, length<br />

and head circumference measures, skinfold<br />

thickness, and bioimpedance data.<br />

• Correlation <strong>of</strong> body composition findings<br />

with neonatal cord blood evaluation<br />

(glucose, insulin, adiponectin and<br />

inflammatory cytokines).<br />

6: Detailed body composition evaluation at 6, and 18<br />

months <strong>of</strong> age as measured by weight, length and<br />

head circumference measures, skin-fold thickness,<br />

and bioimpedance data.<br />

7: Correlation <strong>of</strong> body composition findings with<br />

epigenetic changes detected by buccal swab.<br />

References<br />

Dodd JM, Crowther CA, Robinson JS. Dietary and<br />

lifestyle interventions to limit weight gain during<br />

pregnancy for obese or overweight women: a<br />

systematic review. Acta Obstet Gynecol Scand. 2008<br />

May 2:1-5.<br />

Koklu E, Kurtoglu S, Akcakus M, et al. Increased aortic<br />

intima-media thickness is related to lipid pr<strong>of</strong>ile in<br />

newborns with intrauterine growth restriction.<br />

Hormone research 2006;65(6):269-75.<br />

Saarelainen H, Valtonen P, Punnonen K, et al. Flow<br />

mediated vasodilation and circulating<br />

concentrations <strong>of</strong> high sensitive C-reactive protein,<br />

interleukin-6 and tumor necrosis factor-alpha in<br />

normal pregnancy--The Cardiovascular Risk in Young<br />

Finns Study. Clin Physiol Funct Imaging 2009;29(5):347-<br />

52.<br />

Curry AE, Vogel I, Skogstrand K, et al. Maternal<br />

plasma cytokines in early- and mid-gestation <strong>of</strong><br />

normal human pregnancy and their association with<br />

maternal factors. J Reprod Immunol 2008;77(2):152-<br />

60.<br />

Hill C, Llewellyn CH, Saxton J et al. Adiposity and<br />

‘eating in the absence <strong>of</strong> hunger’ in children. Int J<br />

Obes 2008, 32:1499-1505.<br />

PROJECT (CLINICAL): The association between<br />

maternal obesity and placental histology.<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong> Jodie Dodd<br />

Dr Yee Khong<br />

Contact Person<br />

Jodie Dodd<br />

Phone: 8161 7619<br />

lisa.moran@adelaide.edu.au<br />

<strong>Project</strong> Background<br />

Women who are overweight and obese in<br />

pregnancy are at increased risk <strong>of</strong> adverse outcomes<br />

including, hypertensive conditions and preeclampsia,<br />

gestational diabetes, infection, thromboembolic<br />

events, need for induction <strong>of</strong> labour,<br />

caesarean section and perinatal death. Histological<br />

examination <strong>of</strong> the placenta provides important<br />

information that can be used to interpret clinical<br />

outcomes <strong>of</strong> pregnancy and birth. The LIMIT Trial<br />

recruited women who were overweight and obese in<br />

pregnancy and collected information about their<br />

birth outcomes.<br />

Expected Outcome<br />

Histological examination <strong>of</strong> the placentas <strong>of</strong> a subset<br />

<strong>of</strong> women participating in the study may yield<br />

important information regarding the effect <strong>of</strong><br />

overweight and obesity on pregnancy outcome.<br />

14 |


Cerebral Palsy Research Group<br />

The Cerebral Palsy Research group is the world’s<br />

premier group investigating the interaction <strong>of</strong><br />

genetic and environmental causes <strong>of</strong> cerebral palsy.<br />

PROJECT (BASIC): Perinatal Stroke and Cerebral Palsy.<br />

R. O&G.<br />

Supervisors/Contact Persons<br />

Pr<strong>of</strong>essor Alastair MacLennan<br />

alastair.maclennan@adelaide.edu.au<br />

Dr Catherine Gibson<br />

catherine.s.gibson@adelaide.edu.au<br />

adelaide.edu.au/directory/catherine.s.gibson<br />

Dr Michael O’Callaghan<br />

michael.ocallaghan@adelaide.edu.au<br />

adelaide.edu.au/directory/michael.ocallaghan<br />

<strong>Project</strong> Background/Aims<br />

Cerebral palsy is a developmental disorder affecting<br />

movement and posture. It is the result <strong>of</strong> nonprogressive<br />

brain injury occurring before birth or early<br />

in life and there is currently no cure. Cerebral palsy<br />

affects 1 in 500 babies born in Australia with a total <strong>of</strong><br />

33,000 individuals affected in this country alone (1).<br />

Cerebral palsy presents a burden to the Australian<br />

economy <strong>of</strong> $4 billion per annum. The rate <strong>of</strong><br />

diagnosis has not changed in the last 50 years since<br />

records have been kept.<br />

Our group’s ongoing research program has provided<br />

evidence for an association <strong>of</strong> clotting genes with<br />

cerebral palsy (2, 3). The aim <strong>of</strong> this project will be to<br />

seek clinical details, such as MRI reports and<br />

evidence <strong>of</strong> infection for a cohort <strong>of</strong> patients with<br />

cerebral palsy. This clinical information will be<br />

correlated with existing genetic data to help better<br />

understand the role <strong>of</strong> thrombosis in cerebral palsy<br />

causation.<br />

References<br />

1. Access Economics. The Economic Impact <strong>of</strong><br />

Cerebral Palsy in Australia in 2007. 2008.<br />

2. O'Callaghan ME, Maclennan AH, Gibson<br />

CS, et al. Fetal and Maternal Candidate<br />

Single Nucleotide Polymorphism<br />

Associations With Cerebral Palsy: A Case-<br />

Control Study. Pediatrics 2012:DOI:<br />

peds.2011-0739 [pii] 10.1542/peds.2011-<br />

0739.<br />

3. Gibson CS, MacLennan AH, Hague WM, et<br />

al. Associations between inherited<br />

thrombophilias, gestational age, and<br />

cerebral palsy. Am J Obstet Gynecol<br />

2005;193:1437.<br />

Research Synthesis Division<br />

PROJECT (Clinical): Interventions for improving health<br />

<strong>of</strong> women and babies.<br />

Supervisor<br />

Pr<strong>of</strong>essor Caroline Crowther<br />

Pr<strong>of</strong>essor Jodie Dodd<br />

Philippa Middleton<br />

Dr Rosalie Grivell<br />

Contact Person<br />

Pr<strong>of</strong>essor Caroline Crowther<br />

Phone: 8161 7619<br />

caroline.crowther@adelaide.edu.au<br />

<strong>Project</strong> background<br />

There are many care practices in pregnancy and<br />

labour care that require a systematic review <strong>of</strong> the<br />

literature.<br />

Expected outcomes<br />

• publication <strong>of</strong> a Cochrane review on The<br />

Cochrane Library<br />

• a health topic to suit all research interests<br />

• research skills gained in preparing a<br />

systematic review<br />

• research skills gained in preparing umbrella<br />

reviews around health topic<br />

• skills in preparing health statements for<br />

health pr<strong>of</strong>essionals and consumers.<br />

Keywords<br />

Antenatal care, systematic reviews, perinatal health,<br />

diagnostic test reviews, intervention reviews.<br />

References<br />

Tieu J, Crowther CA, Middleton P. Dietary advice in<br />

pregnancy for preventing gestational diabetes<br />

mellitus. Cochrane Database Syst Rev. 2008 Apr<br />

16;(2) Review.<br />

Rumbold A, Duley L, Crowther CA, Haslam RR.<br />

Antioxidants for preventing pre-eclampsia. Cochrane<br />

Database Syst Rev. 2008 Jan 23;(1):CD004227.<br />

Review.<br />

McDonald S, Middleton P. Effect <strong>of</strong> timing <strong>of</strong><br />

umbilical cord clamping <strong>of</strong> term infants on maternal<br />

and neonatal outcomes. Cochrane Database <strong>of</strong><br />

Systematic Reviews 2008:2<br />

Doyle LW, Crowther CA, Middleton P, Marret S, Rouse<br />

D. Magnesium sulphate for women at risk <strong>of</strong> preterm<br />

birth for neuroprotection <strong>of</strong> the fetus. Cochrane<br />

Database Syst Rev (to be published Jan 21 2009).<br />

Translational <strong>Health</strong> Division<br />

PROJECT (Clinical): To identify evidence-practice<br />

gaps in maternity care.<br />

R. O&G.<br />

Supervisor<br />

Pr<strong>of</strong>essor Caroline Crowther<br />

15 |


Philippa Middleton<br />

Contact Person<br />

Pr<strong>of</strong>essor Caroline Crowther<br />

Phone: 8161 7619<br />

caroline.crowther@adelaide.edu.au<br />

<strong>Project</strong> Background<br />

Clinical research results showing treatments to be<br />

effective are worthless if this information is not then<br />

used by health care workers to benefit their patients.<br />

Several projects will by audit techniques identify<br />

evidence-practice gaps in maternity care and<br />

Breast Biology and Cancer<br />

Supervisors<br />

A/Pr<strong>of</strong> Wendy Ingman<br />

Dr Danielle Glynn<br />

explore reasons for these. Audits will be conducted <strong>of</strong><br />

care practices:<br />

• preconception<br />

• antenatal period<br />

Expected outcomes<br />

Identification <strong>of</strong> care practices where there is an<br />

evidence-practice gap, and exploring the reasons<br />

why. This will lead on to planning intervention studies<br />

to improve patient care.<br />

Keywords<br />

Evidence-practice gap, audit, barrier analysis.<br />

Location: Medical School and The Queen Elizabeth Hospital (TQEH)<br />

Contact Person<br />

A/Pr<strong>of</strong> Wendy Ingman<br />

Phone: 8313 4100 (Medical School) or 8222 6141 (TQEH)<br />

wendy.ingman@adelaide.edu.au<br />

Breast cancer is the most prevalent type <strong>of</strong> cancer among women, with approximately 13,000 new cases<br />

diagnosed each year in Australia. The aim <strong>of</strong> our research is to understand the cellular and molecular mechanisms<br />

that underpin this high incidence <strong>of</strong> breast cancer. We study how key risk factors, including menstrual cycling and<br />

breast density, lead to increased susceptibility <strong>of</strong> the mammary gland to cancer, with specific focus on how these<br />

risk factors affect the ability <strong>of</strong> the immune system to protect this unique tissue against carcinogens and other<br />

cancer initiating factors. The overarching objective <strong>of</strong> this research is to provide therapies that reduce a woman's<br />

lifetime risk <strong>of</strong> developing breast cancer.<br />

In addition to studies on breast cancer susceptibility, we also conduct mastitis research. Mastitis is a common<br />

inflammatory disease in lactating women that causes pain, fever, low milk supply and leads many to cease<br />

breastfeeding. This research explores the cellular mechanisms that lead to inflammation, and investigates potential<br />

therapies to quickly and effectively stop the symptoms <strong>of</strong> mastitis.<br />

PROJECT: (Basic) Cytokine expression in breast<br />

epithelial cells.<br />

R. O&G.<br />

<strong>Project</strong> Background<br />

Previous research in our laboratory has shown that<br />

macrophage phenotype and function in the<br />

mammary gland fluctuate in response to the<br />

changing hormone environment during the estrous<br />

cycle (which is the mouse equivalent to the human<br />

menstrual cycle). This is significant because the<br />

number <strong>of</strong> menstrual cycles a woman experiences in<br />

her lifetime is an important determinant <strong>of</strong> breast<br />

cancer risk. Macrophage function is dependent on<br />

the types <strong>of</strong> cytokines the cell is exposed to. This<br />

project will investigate cytokine production by<br />

human breast epithelial cells in vitro, in response to<br />

ovarian hormone stimulation.<br />

PROJECT: (Basic) Inflammation and mastitis.<br />

R. O&G.<br />

<strong>Project</strong> Background<br />

Mastitis is a common inflammatory condition that<br />

affects 15-20% <strong>of</strong> lactating women in the first 6<br />

months postpartum. It causes fever, pain, reduced<br />

milk production and leads many women to cease<br />

breastfeeding. Although bacterial infection is highly<br />

associated with mastitis in dairy cows, the link<br />

between infection and mastitis in women is less clear.<br />

A family <strong>of</strong> inflammatory signalling pathways have<br />

been identified as playing a crucial role in generating<br />

the symptoms <strong>of</strong> mastitis. These pathways are known<br />

to be stimulated by not only bacteria and their by-<br />

16 |


products, but also naturally occurring molecules<br />

found in breast milk. This project will utilise a mouse<br />

model <strong>of</strong> mastitis developed in our laboratory to<br />

further characterise the contribution <strong>of</strong> inflammatory<br />

signalling pathways and the role <strong>of</strong> immune cells in<br />

the pathology and resolution <strong>of</strong> mastitis.<br />

17 |


Circadian Physiology Group<br />

Contact Person<br />

Pr<strong>of</strong>essor David Kennaway<br />

Phone: 8313 4090<br />

david.kennaway@adelaide.edu.au<br />

Supervisors<br />

Pr<strong>of</strong>essor David Kennaway<br />

Dr Tamara Varcoe<br />

Location: Medical School North<br />

It has been known for many years that a wide range <strong>of</strong> physiological functions change across 24 hours in a<br />

predictable manner. These daily or circadian rhythms are entrained to the environmental day/night cycle via the<br />

eyes, the hypothalamus and a suite <strong>of</strong> clock gene transcription factors. The clock genes are rhythmically expressed<br />

in a wide range <strong>of</strong> tissues and organs, including the liver, muscle, adipose tissue, pancreas, mammary glands and<br />

reproductive tract. This raises the question <strong>of</strong> the impact <strong>of</strong> disruption to their expression on health and the<br />

predisposition to diseases. To this end the Circadian Physiology Group is actively pursuing the role <strong>of</strong> clock genes<br />

and proteins in the maintenance <strong>of</strong> metabolic homeostasis, susceptibility to the development <strong>of</strong> breast cancer and<br />

impaired fertility.<br />

PROJECT: (Basic) Is eating a high fat diet during the<br />

night harmful to shiftworkers?<br />

R. O&G.<br />

<strong>Project</strong> Background<br />

There is increasing evidence that abnormal<br />

rhythmicity may contribute to, as well as result from,<br />

diabetes and related disorders. Shift work schedules<br />

disrupt sleep/wake rhythmicity, eating patterns and<br />

light exposure and as a consequence shift workers<br />

are at a high risk <strong>of</strong> becoming obese and developing<br />

diabetes compared to day workers. This strongly<br />

suggests that abnormal rhythmicity may be an<br />

important causal factor in certain diseases.<br />

Direct<br />

evidence for this is currently lacking and the nature <strong>of</strong><br />

the underlying mechanisms responsible is unknown.<br />

We have, however, shown that mutations in clock<br />

genes that disrupt rhythmicity alter glucose control.<br />

The aim <strong>of</strong> this study is to determine the effect <strong>of</strong> mismatching<br />

the availability <strong>of</strong> food and the light dark<br />

cycle on metabolic homeostasis.<br />

Mice will be given access to a high fat diet<br />

exclusively during the light period or during the dark<br />

period (control) for 4 weeks. The effects on circadian<br />

rhythms <strong>of</strong> various organs will be determined,<br />

together with the effects on metabolic function.<br />

Expected Outcome<br />

The continual mis-matching <strong>of</strong> food availability and<br />

the light schedule will lead to glucose intolerance<br />

and insulin resistance.<br />

What you will gain<br />

Wider understanding <strong>of</strong> metabolic control systems,<br />

analytical skills, including Real time RT-PCR and<br />

hormone immunoassay.<br />

Reference<br />

D. J. Kennaway, J. A. Owens, A. Voultsios, M. J. Boden and T.<br />

J. Varcoe (2007) Metabolic homeostasis in mice with<br />

disrupted Clock gene expression in peripheral tissues,<br />

American Journal <strong>of</strong> Physiology 293 (4) R1528-R1537<br />

PROJECT: (Basic) Uterine expression <strong>of</strong> clock genes<br />

and clock controlled genes in early pregnancy.<br />

R. O&G.<br />

<strong>Project</strong> Background<br />

The involvement <strong>of</strong> daily or circadian rhythmicity in<br />

reproductive processes has been recognised for over<br />

30 years and is particularly evident in female rodents.<br />

Laboratory mice and rats have a precisely timed<br />

surge in LH on the afternoon <strong>of</strong> pro-estrus, which<br />

results in ovulation around 6 hours later and<br />

copulation and fertilisation within a few hours <strong>of</strong> the<br />

ovulation. This rhythmicity is driven by a group <strong>of</strong><br />

neurons in the brain called the suprachiasmatic<br />

nucleus (SCN). The contemporary view <strong>of</strong> how the<br />

SCN generates its endogenous rhythmicity is that a<br />

set <strong>of</strong> proteins exerts both positive and negative<br />

feedback actions on their own gene expression. The<br />

two key proteins are CLOCK and BMAL1, which form<br />

heterodimers and bind to an enhancer E-box in the<br />

promoters <strong>of</strong> per1 and per2, driving their<br />

transcription. It is now also clear that peripheral<br />

tissues including the reproductive tract express a<br />

wide range <strong>of</strong> proteins rhythmically and that<br />

18 |


disruption <strong>of</strong> either Clock or Bmal1 has a deleterious<br />

effect on reproduction.<br />

The aim <strong>of</strong> this project will be to determine what<br />

changes occur in early pregnancy in Clock mutant<br />

mice that lead to the fetal loss <strong>of</strong>ten observed in<br />

these animals.<br />

Adult Clock ∆ 19 mutant mice and their respective<br />

control lines will be mated with appropriate control<br />

males. Uteri will be obtained at 4 hour intervals for 48<br />

hours, starting on the morning <strong>of</strong> day 4 after sperm<br />

detection and processed to measure the expression<br />

<strong>of</strong> a range <strong>of</strong> genes at selected times;<br />

For example Clock genes: Per1, Per2, Clock & Bmal1;<br />

Clock controlled genes: Bsg, PAI-1, COX-2, VEGF &<br />

EG-VEGF. Cytokines: TGFβ, TNFα, IL-10, IL-5, IL-11, IL-<br />

11R; Growth factors: Leptin, lif, igf-1, igf-2 and igf-bp1;<br />

Transcription factor: HOXA10<br />

Expected Outcomes<br />

This experiment will provide us for the first time with<br />

information on the role <strong>of</strong> cellular rhythmicity in the<br />

peri-implantation period.<br />

What you will gain<br />

Wider understanding <strong>of</strong> early implantation control<br />

systems, embryology, analytical skills, including Real<br />

time RT-PCR.<br />

Reference<br />

D. J. Kennaway, M. J. Boden and A. Voultsios (2005)<br />

Reproductive performance in female Clock ∆ 19 mutant<br />

mice. Reproduction Fertility and Development 16: 801-810<br />

19 |


Developmental Biology <strong>of</strong> the Ovary<br />

Contact Person<br />

Pr<strong>of</strong>essor Ray Rodgers<br />

Phone: 8313 3932<br />

ray.rodgers@adelaide.edu.au<br />

Supervisors<br />

Pr<strong>of</strong>essor Ray Rodgers<br />

Dr Katja Hummitzsch<br />

Location: Level 2, Medical School North<br />

The ovary produces eggs and hormones, and these functions are dependent upon growth and development <strong>of</strong><br />

follicles and corpora lutea within the ovary. Control <strong>of</strong> these processes is externally via hormones, or internally by<br />

growth factors and extracellular matrix. In particular our laboratory focuses on the role <strong>of</strong> extracellular matrix in<br />

regulating ovarian function as this has been poorly studied. We use molecular and cellular biology approaches. This<br />

area is important as extracellular matrix can be used to regulate ovarian cell function in vitro, as well as aiding in the<br />

understanding <strong>of</strong> ovarian diseases including infertility and hormone imbalances.<br />

Recently we discovered that the follicular basal lamina structure is related to the quality <strong>of</strong> the oocyte in the follicles<br />

(1) and we are hunting biomarkers <strong>of</strong> such follicles with a view to improving success or assisted reproductive<br />

technologies. We also discovered that a specialized extracellular matrix, focimatrix, is not only developmentally<br />

regulated in follicles but may be part <strong>of</strong> the mechanism by which a dominant follicle is selected to ovulate (2).<br />

Currently we do not understand how one follicle is selected or gains dominance over other follicles as a means <strong>of</strong><br />

controlling ovulation rate, and our discovery could be valuable in unravelling this phenomenon.<br />

References<br />

Irving-Rodgers HF, Morris S, Collett RA, Peura TT, Davy M, Thompson JG, Mason HD, Rodgers RJ (2009) Phenotypes <strong>of</strong><br />

the ovarian follicular basal lamina predict developmental competence <strong>of</strong> oocytes. Human Reprod. 24, 936-944<br />

Irving-Rodgers HF, Harland ML, Sullivan TR and Rodgers RJ (2009) Studies <strong>of</strong> granulosa cells maturation in dominant<br />

and subordinate bovine follicles: Novel extracellular matrix focimatrix is co-ordinately regulated with cholesterol sidechain<br />

cleavage CYP11A1. Reproduction 137, 825-834.<br />

Key Words<br />

Extracellular matrix, ovary, follicle, corpus luteum.<br />

PROJECT: (Basic) Microarray analysis <strong>of</strong> the epithelialmesenchymal<br />

transition (EMT) <strong>of</strong> granulosa cells.<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Ray Rodgers<br />

<strong>Project</strong> Background<br />

The wall <strong>of</strong> the ovarian follicle is a stratified epithelium<br />

and at ovulation and after the release <strong>of</strong> the oocyte,<br />

the follicle wall remodels completely. At this time the<br />

follicular basal lamina is degraded, and cells from the<br />

surrounding stromal thecal layer migrate between<br />

the epithelial granulosa cells (Fig. 1). The follicle wall<br />

thus develops into the corpus luteum. This is<br />

accompanied by a substantial increase in<br />

progesterone synthesis via a process termed<br />

luteinisation and the cells derived from the granulosa<br />

cells are referred to as luteal cells.<br />

Luteinisation has all the features <strong>of</strong> an epithelialmesenchymal<br />

transformation. A key component <strong>of</strong><br />

EMT in vivo is the destruction <strong>of</strong> the epithelial basal<br />

lamina (Fig. 2). During this process the epithelial cells<br />

lose apical-basal polarity and instead develop the<br />

mesenchymal cell features necessary for migration.<br />

During embryogenesis and tissue repair or cellular<br />

reorganization, as well as in the metastasis <strong>of</strong><br />

carcinomas, epithelial cells can undergo a transition<br />

into mesenchymal cells that is referred to as<br />

epithelial-mesenchymal transition (EMT) (Fig. 2).<br />

Studies <strong>of</strong> EMT have largely been conducted using<br />

cells from simple epithelia. Importantly, this project<br />

will study the EMT <strong>of</strong> the stratified epithelium <strong>of</strong> the<br />

ovarian follicle. Techniques used will include<br />

microarray analysis comparing granulosa cells and<br />

luteal cells.<br />

20 |


PROJECT: (Basic) Stem Cells <strong>of</strong> Ovarian Follicles.<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Ray Rodgers<br />

<strong>Project</strong> Background<br />

Our laboratory has shown that a proportion <strong>of</strong><br />

granulosa cells isolated directly from antral follicles<br />

have a number <strong>of</strong> stem cell properties. These include<br />

the ability to divide under anchorage independent<br />

conditions [2], divide without contact inhibition [1]<br />

and express telomerase [3]. On this basis we<br />

proposed the model shown in the above figure [1].<br />

It assumes plutipotentiality into at least cumulus cells<br />

(specialized cells surrounding the oocyte) and mural<br />

granulosa cells. The pluripotency <strong>of</strong> granulosa stem<br />

cells has yet to be demonstrated, however in mice<br />

expressing a dominant-stable mutant <strong>of</strong> betacatenin,<br />

clonally derived granulosa cell tumours<br />

develop. These can be <strong>of</strong> granulosa, bone or neural<br />

phenotype [4], suggesting that tumours <strong>of</strong> granulosa<br />

cells are pluripoten. The goals <strong>of</strong> this project are to<br />

investigate the pluripotential <strong>of</strong> granulosa cells.<br />

PROJECT: (Basic) Thecal stem cells – Real or not?<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Ray Rodgers<br />

<strong>Project</strong> Background<br />

In a recent Proceedings <strong>of</strong> the National Academy <strong>of</strong><br />

<strong>Sciences</strong> article, ‘thecal stem cells’ [5] were reported<br />

in new born mice. However in this species follicles<br />

are still developing at birth.<br />

Whole ovaries were treated with collagenase and<br />

from this a heterogeneous mixture <strong>of</strong> cells was<br />

cultured and colonies <strong>of</strong> stem cells were identified in<br />

anchorage-independent culture. The authors<br />

identified expression <strong>of</strong> markers found in thecal cells,<br />

which in vivo are only expressed in the presence <strong>of</strong><br />

granulosa cells. However they did not examine the<br />

colonies to see if all cells expressed the markers.<br />

Therefore it is possible that the colonies were <strong>of</strong> mixed<br />

cell type, containing granulosa stem cells and some<br />

thecal cells. Certainly the colonies bore a sticking<br />

resemblance to granulosa stem cells [2].<br />

References<br />

Rodgers RJ, Lavranos TC, van Wezel IL, Irving-Rodgers<br />

HF. Development <strong>of</strong> the ovarian follicular epithelium.<br />

Mol Cell Endocrinol 1999; 151: 171-179.<br />

Lavranos TC, Rodgers HF, Bertoncello I, Rodgers RJ.<br />

Anchorage-independent culture <strong>of</strong> bovine granulosa<br />

cells: the effects <strong>of</strong> basic fibroblast growth factor and<br />

dibutyryl cAMP on cell division and differentiation.<br />

Exp Cell Res 1994; 211: 245-251.<br />

Lavranos TC, Mathis JM, Latham SE, Kalionis B, Shay<br />

JW, Rodgers RJ. Evidence for ovarian granulosa stem<br />

cells: telomerase activity and localization <strong>of</strong> the<br />

telomerase ribonucleic acid component in bovine<br />

ovarian follicles. Biol Reprod 1999; 61: 358-366.<br />

Boerboom D, White LD, Dalle S, Courty J, Richards JS.<br />

Dominant-stable beta-catenin expression causes cell<br />

fate alterations and Wnt signaling antagonist<br />

expression in a murine granulosa cell tumor model.<br />

Cancer Res 2006; 66: 1964-1973.<br />

Honda A, Hirose M, Hara K, Matoba S, Inoue K, Miki H,<br />

Hiura H, Kanatsu-Shinohara M, Kanai Y, Kono T,<br />

Shinohara T, Ogura A. Isolation, characterization, and<br />

in vitro and in vivo differentiation <strong>of</strong> putative thecal<br />

stem cells. Proc Natl Acad Sci U S A 2007; 104: 12389-<br />

12394.<br />

PROJECT: (Basic) Bioinformatics <strong>of</strong> Ovarian Follicle<br />

Development.<br />

R. O&G.<br />

Supervisor<br />

Pr<strong>of</strong>essor Ray Rodgers<br />

Bioinformatics is a newly emerging area <strong>of</strong> increasing<br />

importance in all areas <strong>of</strong> biology. Currently there<br />

are insufficient numbers <strong>of</strong> bioinformaticians to satisfy<br />

the need to fully exploit the explosion <strong>of</strong> data<br />

currently available, much less that to be produced in<br />

the future.<br />

Microarrays produce a wealth <strong>of</strong> data. We have<br />

conducted over 70 Affymetrix gene expression arrays<br />

each with over 20,000 probe sets which equates over<br />

1.5 million pieces <strong>of</strong> data from our experiments alone.<br />

We have arrays performed on RNA isolated different<br />

ovarian compartments (theca layers, granulosa cells,<br />

cumulus-oocyte complexes) from follicles at different<br />

stages <strong>of</strong> health and development, and from cells<br />

(theca and granulosa) in culture. There are<br />

numerous computer programs for basic analyses and<br />

we routinely use Partek and Ingenuity Pathway<br />

Analysis (IPA) and are currently conducting basic<br />

analysis <strong>of</strong> these arrays. We have made novel<br />

discoveries <strong>of</strong> follicle development, not hitherto<br />

apparent by previous hypothesis-driven research.<br />

Heat Map <strong>of</strong> gene expression comparing 14 follicles.<br />

21 |


This project will mine our data in greater detail for<br />

relationships between in vitro and in vivo data. It will<br />

compare our data with other data sets available<br />

publicly or collaboratively from ovaries <strong>of</strong> other<br />

species, from other organs and cell types and<br />

diseases identified by IPA. It will identify unique novel<br />

markers <strong>of</strong> cell types and behaviours in follicles.<br />

Key words<br />

Bioinformatics, microarray, computer analyses.<br />

PROJECT: (Basic) Ovarian fetal stroma and PCOS<br />

(polycystic ovary syndrome).<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Ray Rodgers<br />

Katja Hummitzsch<br />

<strong>Project</strong> Background<br />

Polycystic ovary syndrome (PCOS) is the commonest<br />

endocrine condition affecting an estimated 5-7% <strong>of</strong><br />

women <strong>of</strong> reproductive age in Western societies, and<br />

is a major burden on health costs. It is characterised<br />

by hyperandrogenaemia and hirsutism, chronic<br />

anovulation, and polycystic ovaries. Affected<br />

women are also at increased risk <strong>of</strong> anovulatory<br />

infertility, obesity, hyperlipidaemia, type II diabetes<br />

and possibly cardiovascular disease.<br />

Its causes are not well understood but it is thought<br />

that there is both a genetic predisposition and a fetal<br />

cause. The ovary also has excessive collagen and<br />

stroma, which in other tissues, is due to increased<br />

activity <strong>of</strong> transforming growth factor-β. In a recent<br />

study (1) we found that fibrillin 3, thought to regulate<br />

TGFβ activity in tissues and a gene that is familiarly<br />

linked to PCOS is expressed in fetal ovaries at a<br />

critical time when follicles are forming and the<br />

ovarian stroma is expanding.<br />

This project will examine cultured bovine fetal ovarian<br />

cells to determine which cells express fibrillin 3 and to<br />

examine how fibrillin 3 is regulated during gestation.<br />

This information is the critical next step in<br />

understanding the causes <strong>of</strong> PCOS, and hopefully its<br />

prevention.<br />

The project will involve primary cell culture from<br />

ovaries obtained from an abattoir and basic cell and<br />

molecular biology techniques.<br />

Reference<br />

Hatzirodos N, Bayne RA, Irving-Rodgers HF,<br />

Hummitzsch K, Sabatier L, Lee S, Bonner W, Gibson<br />

MA, Rainey WR, Carr BR, Mason HD, Reinhardt DP,<br />

Anderson RA, Rodgers RJ (2011) Linkage <strong>of</strong> regulators<br />

<strong>of</strong> TGFβ activity in the fetal ovary to polycystic ovary<br />

syndrome. FASEB Journal 25, 2256-2265.<br />

22 |


Early Development Group<br />

Contact Person<br />

Dr Mel McDowall<br />

Phone: 8313 8197<br />

melanie.mcdowall@adelaide.edu.au<br />

Supervisors<br />

Associate Pr<strong>of</strong>essor Jeremy Thompson<br />

Dr Robert Gilchrist<br />

Dr Mel McDowall<br />

Location: Medical School<br />

A major focus <strong>of</strong> our laboratory is the influence <strong>of</strong> the microenvironment surrounding the oocytes (eggs) and<br />

embryos on development outcomes, with particular focuses on metabolites (glucose, oxygen) and associated<br />

pathways and genes (for example Hypoxia Inducible Factor, HIF). The incidence <strong>of</strong> diet related obesity (such as high<br />

glucose diets) in Australia is rapidly increasing and is associated with higher susceptibility le to chronic diseases,<br />

decreased fertility, poor pregnancy outcomes and development <strong>of</strong> children born from obese mothers. Glucose is<br />

particularly important for oocyte development and alterations in the level <strong>of</strong> glucose the oocyte is exposed to prior<br />

to fertilisation can have a negative impact on the success <strong>of</strong> embryo development.<br />

Research performed within our group is relevant to human reproduction, human assisted reproduction and the<br />

livestock farming industries through a better understanding <strong>of</strong> the impact <strong>of</strong> the maternal environment on successful<br />

fertilisation and early embryonic development. Furthermore, our research aims to enhance the outcomes <strong>of</strong> both<br />

human and livestock in vitro embryo production by developing improved culture conditions.<br />

PROJECT: (Basic) Glucosamine supplementation<br />

during the peri-conception period and pregnancy<br />

outcomes.<br />

R. O&G.<br />

<strong>Project</strong> Background<br />

It is recognised that maternal health during<br />

pregnancy impacts the pre-natal development and<br />

long-term health <strong>of</strong> children. In particular, maternal<br />

hyperglycaemia (blood glucose levels greater than<br />

240-300 mg/dl, as a result <strong>of</strong> type I and II diabetes,<br />

poor diet and obesity) is related to reduced fertility<br />

and increased rates <strong>of</strong> miscarriage and congenital<br />

abnormalities.<br />

Recent developments suggest that the brief window<br />

prior to and during conception (peri-conceptional<br />

period), coinciding with oocytes (eggs) undergoing<br />

rapid changes to prepare for fertilisation, is critical for<br />

successful fertilisation, implantation, pregnancy and<br />

long-term health. In particular, maternal<br />

hyperglycaemia is related to compromised oocyte<br />

development, such as smaller and fewer oocytes<br />

and reduced ability to successfully develop after<br />

fertilisation. Hence, the levels <strong>of</strong> glucose the oocyte is<br />

exposed to prior to fertilisation can impact the<br />

establishment <strong>of</strong> pregnancy and possible the longterm<br />

health <strong>of</strong> resulting children.<br />

Our group has demonstrated that artificially<br />

stimulating a particular fuel sensing pathway using<br />

the hyperglycaemia mimetic glucosamine, results in<br />

poor oocyte and embryo quality. However, the<br />

mechanisms <strong>of</strong> how glucosamine is affecting fertility<br />

are unknown.<br />

The aim <strong>of</strong> this project is to characterise the influence<br />

<strong>of</strong> glucosamine administration in mice on<br />

reproductive organs (ovaries, uterus) and pregnancy<br />

outcomes (fetal development). Techniques include<br />

animal handling, tissue preparation and staining and<br />

morphology assessments.<br />

PROJECT: (Basic) Glucosamine supplementation and<br />

the oocyte.<br />

R. O&G.<br />

<strong>Project</strong> Backgound<br />

Glucosamine is a dietary supplement that is<br />

commonly used to treat joint problems. However,<br />

glucosamine supplementation during oocyte<br />

maturation results in poor embryo development postfertilisation<br />

by mimicking hyperglycaemic conditions.<br />

The aim <strong>of</strong> this project is to further investigate the<br />

mechanisms through which glucosamine<br />

supplementation is perturbing pig oocyte<br />

development. Techniques include tissue culture,<br />

staining and metabolism assays.<br />

23 |


PROJECT: (Basic) Why is there haemoglobin in<br />

cumulus cells?<br />

R. O&G.<br />

<strong>Project</strong> Backgound<br />

We have recently produced microarrays <strong>of</strong> mouse<br />

cumulus cells obtained from naturally ovulated<br />

cumulus-oocyte complexes (COCs) and in vitro<br />

matured COCs. A consistent finding is that mRNA for<br />

haemoglobin is produced by cumulus cells derived<br />

from in vivo matured COCs at high levels compared<br />

to in vitro matured COCs. Furthermore, this<br />

difference has been validated by RT-PCR.<br />

Haemoglobin production by cumulus cells has never<br />

previously been reported and is a unique<br />

observation. This begs the question “Why would<br />

cumulus cells require the production <strong>of</strong><br />

haemoglobin?” One candidate mechanism involves<br />

the regulation <strong>of</strong> cGMP and nitric oxide, both <strong>of</strong><br />

which are known to be involved in the ovulatory<br />

process. Haemoglobin is a known sequestering<br />

molecule for nitric oxide, so we have hypothesised<br />

that cumulus cells derived from in vivo ovulated<br />

COCs produce haemoglobin in response to the<br />

ovulatory process and this is lacking during in vitro<br />

maturation. This project aims to investigate the<br />

production <strong>of</strong> haemoglobin and determine its roles<br />

during follicle ovulation and oocyte maturation.<br />

Techniques will initially include RT-PCR,<br />

immunohistochemistry and western blotting.<br />

References<br />

Sutton ML, Gilchrist RB and Thompson JG (2003),<br />

Effects <strong>of</strong> in vivo and in vitro environments on<br />

metabolism <strong>of</strong> the cumulus-oocyte complex and its<br />

influence on oocyte developmental capacity, Hum<br />

Reprod Update, 9 (1): 35-48<br />

Sutton-McDowall ML, Gilchrist RB and<br />

Thompson JG (2004) Cumulus-expansion and glucose<br />

utilisation by bovine cumulus-oocyte complexes<br />

during in vitro maturation: the influence <strong>of</strong><br />

glucosamine and follicle stimulating hormone,<br />

Reproduction, 128 (3): 313-19<br />

Sutton-McDowall ML, Mitchell M, Cetica P,<br />

Dalvit G, Pantaleon M, Lane M, Gilchrist RB and<br />

Thompson JG (2006) Glucosamine supplementation<br />

during in vitro maturation inhibits subsequent oocyte<br />

developmental competence, Biol Reprod, 74: 881-88.<br />

Schelbach CJ, Kind KL, Lane M and Thompson<br />

JG (2010) Mechanisms contributing to the reduced<br />

developmental competence <strong>of</strong> glucosamineexposed<br />

mouse oocytes. Reprod Fertil Dev, 22 (5):<br />

771-9.<br />

Sutton-McDowall ML, Gilchrist RB and<br />

Thompson JG (2010) The pivotal role <strong>of</strong> glucose<br />

metabolism in determining oocyte developmental<br />

competence. Reproduction, 139(4):685-95.<br />

PROJECT: (Basic) Lipid oxidation in cattle/sheep<br />

oocytes. Unlocking an un-used energy source to<br />

increase developmental competence during in vitro<br />

maturation.<br />

R. O&G.<br />

Supervisors<br />

Dr Kylie Dunning<br />

Dr Melanie McDowall<br />

A/Pr<strong>of</strong> Jeremy Thompson<br />

<strong>Project</strong> Backgound<br />

Intra-oocyte ATP levels are correlated with oocyte<br />

developmental competence post-fertilisation.<br />

Oocytes have little capacity to produce ATP and are<br />

dependent on oxidative phosphorylation within<br />

mitochondria. Cattle, sheep and pig oocytes are<br />

characterised by significant intracellular stores <strong>of</strong><br />

lipid. It has been calculated that there is enough<br />

“stored energy” within these lipid stores to drive all<br />

the necessary ATP production for early development.<br />

However, when oocytes mature in vitro (or early<br />

embryos are cultured in vitro), exogenous<br />

carbohydrates and derivatives are essential for<br />

further development, suggesting the lipid stores are<br />

not utilised. Work conducted in the Ovarian Cell<br />

Biology Group has found that in the mouse, the<br />

addition <strong>of</strong> carnitine to oocyte maturation media<br />

significantly increases lipid metabolism and ATP<br />

production. Carnitine is a co-factor required for the<br />

transfer <strong>of</strong> lipids, in the form <strong>of</strong> fatty acids, from the<br />

cytosol, into mitochondria, where they undergoe �oxidation<br />

and producte ATP. This project will<br />

investigate the role <strong>of</strong> carnitine and lipid oxidation<br />

within cattle/sheep oocyte culture in vitro. Skills<br />

learnt will include oocyte maturation and embryo<br />

production in vitro as well as metabolic assays and<br />

fluorescence microscopy investigating mitochondrial<br />

function.<br />

24 |


Early Life Programming <strong>of</strong> <strong>Health</strong> and Disease<br />

Contact Persons<br />

Pr<strong>of</strong>essor Julie Owens<br />

Phone: 8313 4088<br />

julie.owens@adelaide.edu.au<br />

Dr Kathy Gatford<br />

Phone: 8313 4158<br />

kathy.gatford@adelaide.edu.au<br />

Supervisors<br />

Pr<strong>of</strong>essor Julie Owens<br />

Dr Kathy Gatford<br />

Pr<strong>of</strong>essor Claire Roberts<br />

Dr Miles De Blasio<br />

Ms Patricia Grant<br />

Emeritus Pr<strong>of</strong>essor Jeffrey Robinson<br />

Our research group is part <strong>of</strong> the Research Centre for the Early Origins <strong>of</strong> <strong>Health</strong> and Disease (EOHaD) and an<br />

international leader in the investigation <strong>of</strong> the intergenerational and perinatal origins <strong>of</strong> metabolic and<br />

cardiovascular health in postnatal life. We focus on how our health can be pr<strong>of</strong>oundly influenced by events in early<br />

life and possibly in previous generations, including diabetes, obesity and cardiovascular increasing risk <strong>of</strong> disease.<br />

Common exposures in early life that affect our later health include placental and fetal growth restriction, maternal<br />

vitamin supplementation and maternal obesity and diabetes.<br />

Our aims are to:<br />

• understand how these exposures interact with the genome and affect the epigenome to determine our later<br />

health<br />

• identify interventions to either prevent these conditions that program later health or to overcome or reverse<br />

such programming.<br />

Recent key discoveries include:<br />

• placental restriction<br />

o inducing diabetes in adult <strong>of</strong>fspring via impaired beta cell function and growth and insulin resistance<br />

o causing obesity in <strong>of</strong>fspring via altered appetite control<br />

• altering expression <strong>of</strong> novel small regulatory RNAs that target and repress expression <strong>of</strong> multiple mRNAs to<br />

affect major pathways and funtions<br />

• maternal folate deficiency or folic acid supplementation in changing the epigenome and physiology <strong>of</strong><br />

<strong>of</strong>fspring.<br />

Our research strategies include:<br />

• novel experimental models <strong>of</strong> common early life exposures in non human species<br />

• clinical research <strong>of</strong> human cohorts with exposure to common pregnancy complications<br />

• in vivo characterisation <strong>of</strong> insulin action, glucose and lipid control and other physiological functions<br />

• metabolite and hormone analysis by enzymatic analysis and immunoassay<br />

• quantitative morphmetric analysis <strong>of</strong> tissue structures underpinning function<br />

• molecular analysis <strong>of</strong> expression <strong>of</strong> regulatory RNAs and protein coding genes by quantitative PCR, microarray<br />

and western immunoblotting<br />

• analysis <strong>of</strong> epigenetic state <strong>of</strong> genes by bisulfite treatment <strong>of</strong> DNA and Sequenom or Pyrosequencing<br />

• bioinformatics, including pathway analysis <strong>of</strong> array data <strong>of</strong> microRNA and mRNA expression.<br />

We publish in the top discipline specific and general journals, are highly cited and regularly invite to speak at<br />

leading conferences. Our students publish their work, present at conferences and have gone on to a range <strong>of</strong><br />

careers as researchers, academics, health pr<strong>of</strong>essionals, in the biotechnology and pharmaceutical industry and<br />

management.<br />

25 |


PROJECT: (Basic) Can maternal dietary arginine<br />

supplementation increase placental function and<br />

fetal survival at birth?<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Julie Owens<br />

Dr Miles De Blasio<br />

<strong>Project</strong> Backgound<br />

The placenta acts as the lungs, gut and kidneys for<br />

the growing fetus before birth and influences how<br />

well it grows, develops and its survival. We have<br />

shown that increasing maternal dietary intake <strong>of</strong> the<br />

amino acid arginine throughout pregnancy may<br />

increase fetal growth, size at birth and survival. This<br />

project will investigate the mechanisms underlying<br />

this improvement, including by increasing production<br />

<strong>of</strong> the vasodilator nitric oxide and hence increasing<br />

placental blood flow, and development <strong>of</strong> blood<br />

vessels in the placenta. This may identify a new<br />

approach to restoring placental functional<br />

development in women and to enhancing<br />

reproductive efficiency in the pig industry.<br />

PROJECT: (Basic) Do small non-coding RNAs mediate<br />

onset <strong>of</strong> diabetes and obesity in <strong>of</strong>fspring <strong>of</strong> obese fat<br />

fed mothers?<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Julie Owens<br />

Dr Karen Kind<br />

Ms Patricia Grant<br />

<strong>Project</strong> Backgound<br />

Maternal obesity is common and increasing and<br />

results in a high birthweight and greater risk <strong>of</strong> obesity<br />

and diabetes in <strong>of</strong>fspring later in life. This may be<br />

partly responsible for the growing epidemic in<br />

obesity, including in children. Recently, we have<br />

shown that restricting fetal supply and growth, which<br />

also leads to diabetes in later life, appears to act in<br />

part by altering expression <strong>of</strong> microRNAs (miRs) in key<br />

functional tissues. A novel and major class <strong>of</strong> genes<br />

code for the miRs, which are small RNAs that can<br />

bind to sites in 3’UTRs <strong>of</strong> multiple target genes. The<br />

miRs can regulate many gene mRNA and/or protein<br />

products in a co-ordinated and powerful fashion. In<br />

this project we will determine for the first time using<br />

previously collected samples, if maternal obesity and<br />

oversupply o f the fetus before birth alters expression<br />

<strong>of</strong> microRNAs in the insulin target tissues, skeletal<br />

muscle and liver and if this relates to altered<br />

expression <strong>of</strong> insulin signalling pathways and<br />

metabolic genes. We will also see if plasma miRs are<br />

altered and reflect changes in these key tissues<br />

following maternal obesity. The outcomes will show if<br />

miRs are a key part <strong>of</strong> the mechanism by which<br />

maternal obesity affects metabolic health <strong>of</strong><br />

<strong>of</strong>fspring and if plasma miRs can be used as markers<br />

<strong>of</strong> this for future translation into human studies.<br />

PROJECT: (Basic) Does maternal folic acid<br />

supplementation alter plasma microRNAs in <strong>of</strong>fspring<br />

and are these predictive <strong>of</strong> later metabolic health?<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Julie Owens<br />

Dr Karen Kind<br />

Ms Patricia Grant<br />

Pr<strong>of</strong>essor Marie Dziadek<br />

<strong>Project</strong> Backgound<br />

We have recently shown in rodents that a common<br />

exposure, maternal folic acid supplementation<br />

(MFAS), alters metabolic control and insulin action in<br />

adult <strong>of</strong>fspring. Mandatory food fortification is being<br />

introduced in Australia as has already occurred<br />

overseas and will further increase exposure <strong>of</strong> the<br />

population before birth. We have shown that MFAS<br />

acts in part by altering expression <strong>of</strong> microRNAs<br />

(miRs) in liver <strong>of</strong> adult <strong>of</strong>fspring. The miRs can regulate<br />

many gene mRNA and/or protein products in a coordinated<br />

and powerful fashion. In this project we will<br />

determine if MFAS alters plasma concentrations <strong>of</strong><br />

miRs reflecting changed hepatic expression and<br />

release into blood. If proven, this would identify<br />

highly accessible potential biomarkers <strong>of</strong> MFAS<br />

programming <strong>of</strong> later diabetes for testing in humans.<br />

Plasma miRs will be analysed by RTPCR and<br />

microarray using previously collected samples from<br />

juvenile as well as adult <strong>of</strong>fspring <strong>of</strong> obese fat fed<br />

mothers.<br />

PROJECT: (Basic) Does maternal obesity alter<br />

epigenetic state and expression <strong>of</strong> key regulatory<br />

and functional genes in <strong>of</strong>fspring?<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Julie Owens<br />

Dr Karen Kind<br />

Ms Patricia Grant<br />

Pr<strong>of</strong>essor Marie Dziadek<br />

<strong>Project</strong> Backgound<br />

Maternal obesity is common and increasing and<br />

results in a high birthweight and greater risk <strong>of</strong> obesity<br />

and diabetes in <strong>of</strong>fspring later in life. This may be<br />

partly responsible for the growing epidemic in<br />

obesity, including in children. Maternal obesity<br />

exposes the developing fetus to high glucose and<br />

insulin abundance and other metabolic and<br />

endocrine changes when its epigenetic state is<br />

changing rapidly. This may alter DNA methylation, a<br />

major epigenetic mechanism that influences<br />

expression <strong>of</strong> genes. Once established, epigenetic<br />

patterns are heritable through cell division, and can<br />

26 |


persist long term. There is new evidence in humans<br />

that maternal obesity can change epigenetic state<br />

<strong>of</strong> some genes at birth in the new born, including that<br />

<strong>of</strong> PPARCG1A, an important regulator <strong>of</strong> many<br />

metabolic genes. This project will test for the first time<br />

using rodents, if maternal obesity alters epigenetic<br />

state and expression <strong>of</strong> PPARCG1A and other<br />

important genes in key metabolic tissues <strong>of</strong> <strong>of</strong>fspring<br />

later in life. DNA methylation in the promoter region<br />

<strong>of</strong> PPARCG1A and other genes and their expression<br />

in liver and skeletal muscle <strong>of</strong> <strong>of</strong>fspring <strong>of</strong> obese fat<br />

fed mothers will be analysed by pyrosequencing<br />

after DNA bisulfite treatment and by RTPCR. The<br />

outcomes will show if epigenetic modifications are a<br />

key part <strong>of</strong> the mechanism by which maternal<br />

obesity affects metabolic health <strong>of</strong> <strong>of</strong>fspring and if<br />

these can be used as markers <strong>of</strong> this for future<br />

translation into human studies.<br />

PROJECT: (Clinical)<br />

Is metformin superior to insulin in improving maternal<br />

and newborn metabolic and cardiovascular risk<br />

pr<strong>of</strong>ile in gestational diabetes?<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Julie Owens<br />

Dr Bill Hague<br />

<strong>Project</strong> Backgound<br />

Gestational diabetes is common and increasing in<br />

prevalence. It is associated with complications for<br />

the mother and baby and with an increased risk <strong>of</strong><br />

diabetes and associated cardiovascular disease in<br />

both the mother and the <strong>of</strong>fspring. Modification <strong>of</strong><br />

lifestyle and, if necessary because <strong>of</strong> maternal<br />

hyperglycaemia, treatment with insulin injections,<br />

can improve maternal and infant outcomes at birth.<br />

Recently, we have shown that oral metformin, which<br />

improves insulin sensitivity, is similarly effective and<br />

preferred by women. The aim <strong>of</strong> this project is to<br />

measure metabolic, hormonal and other markers <strong>of</strong><br />

diabetes and cardiovascular disease risk in maternal<br />

blood plasma during and after pregnancy and in<br />

new born cord blood plasma. These markers will<br />

include plasma glucose, free fatty acids, triglycerides,<br />

insulin, C-peptide, leptin, adiponectin and<br />

inflammatory cytokines. This will provide evidence as<br />

to the potential efficacy <strong>of</strong> metformin compared<br />

with insulin in improving circulating predictors <strong>of</strong> later<br />

metabolic and cardiovascular disease in the mother<br />

and her baby. This will provide the rationale for<br />

follow-up trials <strong>of</strong> the long term metabolic outcomes<br />

for the mother and child.<br />

PROJECT: (Basic) What are the effects <strong>of</strong> growth<br />

hormone on placental development and function?<br />

R. O&G.<br />

Supervisors<br />

Dr Kathy Gatford<br />

Pr<strong>of</strong>essor Julie Owens<br />

Pr<strong>of</strong>essor Claire Roberts<br />

<strong>Project</strong> Backgound<br />

Maternal growth hormone treatment can increase<br />

fetal growth, is being tested as a way to increase size<br />

at birth in animal industries, and may be a<br />

therapeutic approach for treating growth-restricted<br />

pregnancies in humans. Although we have and<br />

others have shown increased fetal growth in mothers<br />

given growth hormone injections, we do not know<br />

the relative importance <strong>of</strong> maternal metabolic and<br />

placental responses for increasing fetal growth. In this<br />

project, we will use archived porcine samples to<br />

investigate how maternal growth hormone affects<br />

placental structure and expression <strong>of</strong> the nutrient<br />

transporters that determine fetal nutrient supply and<br />

hence growth, using immunohistology and real-time<br />

PCR. This may identify a new approach to increasing<br />

placental functional development in women and in<br />

animal production, to increase size at birth including<br />

in growth-restricted pregnancies. This project will be<br />

jointly supervised by Dr Gatford, Pr<strong>of</strong> Owens and Pr<strong>of</strong><br />

Claire Roberts (Placental Biology lab).<br />

Students undertaking this project are also eligible for<br />

a Pork CRC <strong>Honours</strong> scholarship, valued at $10 000<br />

($5000 stipend plus $5000 operating).<br />

Application forms (to be completed by you and your<br />

supervisor and due October 31) are available at<br />

http://www.porkcrc.com.au/html/education.html.<br />

References<br />

M Dziadek. 2005 Genomic imprinting and epigenetic<br />

programming <strong>of</strong> fetal development: In: Perinatal<br />

Programming: Early Life Determinants <strong>of</strong> Adult <strong>Health</strong> and<br />

Disease (Eds DM Hodgson and C Coe). Taylor and Francis<br />

Medical <strong>Book</strong>s, London.<br />

Gatford KL, Mohammad SN, Harland ML, De Blasio MJ,<br />

Fowden AL, Robinson JS, Owens JA (2008) Impaired beta-cell<br />

function and inadequate compensatory increases in betacell<br />

mass after intrauterine growth restriction in sheep.<br />

Endocrinology.<br />

Oct;149(10):5118-27.<br />

Quigley SP, Kleemann DO, Walker SK, Speck PA,<br />

Rudiger SR, Nattrass GS, DeBlasio MJ, Owens JA (2008) Effect<br />

<strong>of</strong> variable long-term maternal feed allowance on the<br />

development <strong>of</strong> the ovine placenta and fetus. Placenta.<br />

Jun;29(6):539-48.<br />

Owens JA, Gatford KL, De Blasio MJ, Edwards LJ,<br />

McMillen IC, Fowden AL (2007) Restriction <strong>of</strong> placental<br />

growth in sheep impairs insulin secretion but not sensitivity<br />

before birth. J Physiol.<br />

Nov 1;584(Pt 3):935-49.<br />

Gatford KL, Boyce JM, Blackmore K, Smits RJ,<br />

Campbell RG, Owens PC (2004) Long-term, but not shortterm,<br />

treatment with somatotropin during pregnancy in<br />

underfed pigs increases the body size <strong>of</strong> progeny at birth. J<br />

Anim Sci. Jan;82(1):93-101<br />

27 |


PROJECT: (Basic) Epigenetic consequences <strong>of</strong><br />

placental restriction and dietary methyl<br />

supplementation.<br />

R. O&G.<br />

Supervisors<br />

Dr Kathy Gatford<br />

Dr Karen Kind<br />

Pr<strong>of</strong> Julie Owens<br />

<strong>Project</strong> Backgound<br />

Poor growth before birth is <strong>of</strong>ten caused by poor<br />

development and function <strong>of</strong> the placenta, which<br />

delivers nutrients from the mother to the growing<br />

fetus. The supply <strong>of</strong> particular nutrients, including<br />

methyl donors, is important for chemical<br />

modifications to DNA, which affect gene<br />

transcription and can persist into later life. These<br />

epigenetic changes are thought to contribute to<br />

long-term increased disease risks, such as diabetes<br />

and obesity, in the baby who grew poorly before<br />

birth. We have shown that surgical restriction <strong>of</strong><br />

placental growth and function (PR) produces similar<br />

long-term outcomes as poor growth before birth in<br />

humans, including impaired insulin secretion and<br />

diabetes. As part <strong>of</strong> a larger NHMRC-funded study,<br />

we are testing whether feeding the mother extra<br />

methyl donors in the diet can improve outcomes in<br />

the growth-restricted lamb. In this study, an <strong>Honours</strong><br />

student will investigate how PR affects circulating<br />

methyl donors levels in the neonate at birth, and<br />

whether this can be increased by feeding the mother<br />

with methyl donors in late pregnancy. We will also<br />

investigate the effects <strong>of</strong> PR and methyl donors on<br />

expression <strong>of</strong> epigenetically-regulated genes in white<br />

blood cells and circulating metabolites in plasma<br />

collected at birth, to assess whether the supply <strong>of</strong><br />

these nutrients in late pregnancy changes epigenetic<br />

and metabolic state at birth.<br />

The student will gain skills in animal handling including<br />

venepuncture, analysis <strong>of</strong> circulating metabolites<br />

including radioimmunoassay, measurement <strong>of</strong> gene<br />

expression by RT-PCR, data analysis, written and oral<br />

presentation skills.<br />

PROJECT: (Basic) Do male and females respond<br />

differently to restricted growth before birth?<br />

R. O&G.<br />

Supervisors<br />

A/Pr<strong>of</strong> Vicki Clifton<br />

Dr Kathy Gatford<br />

Dr Karen Kind<br />

Pr<strong>of</strong> Julie Owens<br />

<strong>Project</strong> Backgound<br />

Poor growth before birth is <strong>of</strong>ten caused by poor<br />

development and function <strong>of</strong> the placenta, which<br />

delivers nutrients from the mother to the growing<br />

fetus. Studies in humans, however, suggest that the<br />

response <strong>of</strong> male and female fetuses to a poor<br />

environment differs, so that female fetuses restrict<br />

their growth and are more likely to survive, while<br />

males continue to grow, but are then susceptible to<br />

any worsening <strong>of</strong> the environment. To test this<br />

concept, we will investigate the effects <strong>of</strong> restricted<br />

growth before birth and fetal sex on gene expression<br />

at birth in white blood cells collected at birth and in<br />

placental tissues. We will use our model <strong>of</strong> placental<br />

restriction in the sheep for this study, allowing this<br />

hypothesis to be directly tested in induced growth<br />

restriction and within a population <strong>of</strong> similar<br />

genotype.<br />

The student will gain skills in animal handling including<br />

venepuncture, tissue processing, measurement <strong>of</strong><br />

gene expression by array and RT-PCR, data analysis,<br />

written and oral presentation skills.<br />

PROJECT: (Basic) Immune function after placental<br />

restriction and dietary methyl supplementation.<br />

R. O&G.<br />

Supervisors<br />

Dr Kathy Gatford<br />

A/Pr<strong>of</strong> Vicki Clifton<br />

Dr Karen Kind<br />

Pr<strong>of</strong> Julie Owens<br />

<strong>Project</strong> Backgound<br />

Maternal methyl donor intake during pregnancy has<br />

been linked to increased incidence <strong>of</strong> asthma in<br />

children, suggesting impaired immune responses.<br />

Little is known about how growth before birth affects<br />

immune function into later life. As part <strong>of</strong> a larger<br />

NHMRC-funded study, we are testing whether<br />

feeding the mother extra methyl donors in the diet<br />

can improve metabolic outcomes, particularly for<br />

insulin action, in the growth-restricted lamb. An<br />

<strong>Honours</strong> student will be involved in testing circulating<br />

immune responses to injection <strong>of</strong> a stimulus<br />

(ovalbumin), to assess the effects <strong>of</strong> growth before<br />

birth, neonatal growth, and maternal methyl donors<br />

in late pregnancy, on immune function. We will use<br />

our model <strong>of</strong> placental restriction in the sheep for this<br />

study, allowing this hypothesis to be directly tested in<br />

induced growth restriction and within a population <strong>of</strong><br />

similar genotype.<br />

The student will gain skills in animal handling including<br />

venepuncture, immune challenge, analysis <strong>of</strong><br />

circulating immunoglobulins, data analysis, and<br />

written and oral presentation skills.<br />

28 |


PROJECT: (Basic) Behavioural effects <strong>of</strong> placental<br />

restriction, neonatal growth and dietary methyl<br />

supplementation.<br />

R. O&G.<br />

Supervisors<br />

Dr Susan Hazel<br />

Dr Karen Kind<br />

Dr Kathy Gatford<br />

<strong>Project</strong> Backgound<br />

Poor growth before birth (IUGR) is associated with<br />

decreased cognitive function in human children, as<br />

well as increased risk <strong>of</strong> metabolic disease in later life.<br />

These growth-restricted babies also characteristically<br />

have accelerated neonatal growth rates (neonatal<br />

catch-up growth). Rapid neonatal catch-up growth<br />

is independently associated with metabolic disease<br />

and obesity in later life, but positively predicts<br />

improved neurological development and survival.<br />

Using an ovine model <strong>of</strong> poor growth before birth,<br />

which produces similar adverse metabolic<br />

consequences as human IUGR, we wish to test<br />

whether neonatal interventions that improve insulin<br />

secretion, but which slow neonatal growth, have any<br />

impact on behaviour in early postnatal life. The<br />

<strong>Honours</strong> student will gain skills in animal handling<br />

including measures <strong>of</strong> behaviour and use <strong>of</strong><br />

behavioural analysis s<strong>of</strong>tware, data analysis, written<br />

and oral presentation skills.<br />

29 |


Early Origins <strong>of</strong> Cancer Supervisors<br />

Contact Person<br />

Dr Tina Bianco-Miotto<br />

Phone: 8313 6077<br />

tina.bianco@adelaide.edu.au<br />

Dr Tina Bianco-Miotto<br />

Pr<strong>of</strong>essor Julie Owens<br />

Location:<br />

Research Centre for Reproductive <strong>Health</strong>, Robinson Institute, School <strong>of</strong><br />

Paediatrics and Reproductive <strong>Health</strong><br />

Increasingly, the environment before birth as indicated by foetal growth and birth weight is implicated as a potential<br />

and influential risk factor for cancer. Thus, high birth weight, reflecting excess nutrition and overgrowth, is associated<br />

with an increased risk <strong>of</strong> cancer in many but not all studies. Perturbations <strong>of</strong> the prenatal environment, such as<br />

maternal under or overnutrition, malnutrition, obesity or exposure to endocrine disruptors, appear to alter the<br />

phenotype <strong>of</strong> <strong>of</strong>fspring long term, affecting their susceptibility to metabolic and cardiovascular disease and other<br />

conditions, including breast and prostate cancer. High birth weight is also predictive <strong>of</strong> later obesity, which may<br />

partly mediate the effects <strong>of</strong> excess and abnormal nutrition in utero on the risk <strong>of</strong> cancer. Obesity is increasingly<br />

implicated in cancer and <strong>of</strong> concern, maternal obesity and the associated high birth weight are increasingly<br />

common suggesting the potential for a growing contribution to the burden <strong>of</strong> disease in future. Our research<br />

objective is to determine if the early life environment, such as maternal obesity or folic acid supplementation,<br />

promotes excessive growth before birth and acts on the breast or prostate during development leading to an<br />

increased susceptibility <strong>of</strong> the individual to breast or prostate cancer in later life. We are also investigating whether<br />

epigenetic mechanisms are involved in the developmental origins <strong>of</strong> cancer.<br />

PROJECT: (Basic) Is maternal obesity associated with<br />

an increased risk <strong>of</strong> prostate or breast cancer in adult<br />

progeny?<br />

R. O&G.<br />

<strong>Project</strong> Background<br />

Using a rat model <strong>of</strong> maternal obesity we will<br />

investigate whether the male or female progeny <strong>of</strong><br />

mothers on a high fat diet are at an increased risk <strong>of</strong><br />

prostate or breast cancer. The rat prostates or breasts<br />

from maternal high fat and control diets will be<br />

examined for histological abnormalities. Key markers<br />

<strong>of</strong> cancer development, cell proliferation and<br />

apoptosis will be assessed by immunohistochemistry<br />

and real time quantitative PCR. To determine<br />

whether epigenetic mechanisms are altered<br />

between the two groups <strong>of</strong> rats, the expression <strong>of</strong><br />

epigenetic genes will be assessed by QPCR; global<br />

epigenetic modifications will be assessed by<br />

immunohistochemistry and high resolution melt<br />

curves; DNA methylation <strong>of</strong> key tumour suppressor<br />

genes and imprinted genes will be assessed by<br />

methylation specific PCR and pyrosequencing; and<br />

miRNA expression will also be assessed. A wellcharacterised<br />

mouse model <strong>of</strong> prostate or breast<br />

cancer will also be utilised to determine whether<br />

maternal obesity alters the time <strong>of</strong> onset <strong>of</strong> cancer in<br />

adult <strong>of</strong>fspring.<br />

30 |


Gamete and Embryo Biology Laboratory<br />

Contact Person<br />

Dr Michelle Lane<br />

Phone: 8313 8176<br />

michelle.lane@adelaide.edu.au<br />

PROJECT: (Basic) Making old eggs young.<br />

R. O&G.<br />

<strong>Project</strong> Backgound<br />

An increasing number <strong>of</strong> women seek IVF treatment<br />

as a result <strong>of</strong> declining fertility, associated with<br />

advanced maternal age. However, despite popular<br />

misconceptions that IVF can cure all infertility, IVF<br />

cannot cure aged eggs. It is currently this group <strong>of</strong><br />

patients who <strong>of</strong>ten do not reach their goal <strong>of</strong> a baby<br />

without repeated treatment with its emotional and<br />

financial toll. Mitochondrial function is the<br />

powerhouse <strong>of</strong> the cell and mitochondrial activity<br />

has been shown to reduce with age resulting in a<br />

significant reduction in egg and embryo viability. The<br />

aim <strong>of</strong> this study is to examine the ability <strong>of</strong> novel<br />

mitochondrial nutrients for their ability to rescue<br />

mitochondrial function in aged eggs and embryos to<br />

improve pregnancy rates.<br />

This study will investigate embryo development and<br />

viability after culture in different media formulations<br />

<strong>of</strong> mitochondrial nutrients. Techniques will involve<br />

general embryology, assessment <strong>of</strong> metabolism, epi-<br />

and confocal microscopy, general molecular<br />

methods such as q-PCR.<br />

Information from this study will assist in developing<br />

new media to improve IVF procedures.<br />

Supervisors<br />

Dr Michelle Lane<br />

Dr Deirdre Zander-Fox<br />

Dr Tod Fullston<br />

Dr Hassan Bakos<br />

PROJECT: (Basic) Interaction <strong>of</strong> male obesity and<br />

molecular function <strong>of</strong> human sperm on IVF outcomes.<br />

R. O&G.<br />

<strong>Project</strong> Backgound<br />

There is increasing awareness that male obesity has a<br />

negative impact on sperm quality. Male obesity is<br />

now known to be associated with reduced success<br />

rates after IVF treatment as well as increases in<br />

miscarriage and pregnancy loss. However, the<br />

mechanism for this impact on pregnancy health is<br />

largely unknown. The aim <strong>of</strong> this study is to examine<br />

the impact <strong>of</strong> paternal obesity at the time <strong>of</strong><br />

conception on the molecular structure <strong>of</strong> the sperm<br />

and how this impacts on fertilization, embryo<br />

development and pregnancy after IVF treatment.<br />

This study will investigate sperm motility, count,<br />

reactive oxygen species, methylation and<br />

acetylation <strong>of</strong> sperm in relation to fertilization, sperm<br />

binding and embryo development. Techniques will<br />

involve general andrology methods as well as<br />

immunohistochemistry, epi- and confocal<br />

microscopy, mitochondrial measurement and cell<br />

culture.<br />

31 |


Oocyte Biology Group<br />

Contact Persons<br />

Dr Robert Gilchrist<br />

Phone: 8313 8183<br />

robert.gilchrist@adelaide.edu.au<br />

Dr David Mottershead<br />

Phone: 8313 8201<br />

david.mottershead@adelaide.edu.au<br />

Supervisors<br />

Associate Pr<strong>of</strong>essor Jeremy Thompson<br />

Dr Robert Gilchrist<br />

Dr David Mottershead<br />

Location: Medical School<br />

The Oocyte Biology Group investigates fundamental physiological aspects <strong>of</strong> oocyte-granulosa cell interactions in<br />

an effort to improve understanding <strong>of</strong> mammalian oocyte biology and to better implement assisted reproductive<br />

technologies. Ovarian folliculogenesis consists <strong>of</strong> a series <strong>of</strong> ordered maturation and differentiation events <strong>of</strong> both<br />

the somatic and germ cell components that culminates in ovulation and the production <strong>of</strong> a mature oocyte that is<br />

able to support early embryogenesis. The immature oocyte and the follicular granulosa cells are in intimate contact<br />

and normal growth and development <strong>of</strong> the oocyte is entirely dependent on this association. Oocyte-secreted<br />

growth factors have significant effects on the somatic cell compartment <strong>of</strong> the ovarian follicle. We are actively<br />

studying two <strong>of</strong> these proteins, namely growth differentiation factor 9 (GDF9) and bone morphogenetic factor 15<br />

(BMP15), both <strong>of</strong> which appear to be essential for mammalian fertility. Our aim is to understand the functioning <strong>of</strong><br />

these proteins and the detailed basis <strong>of</strong> the somatic/germ cell (oocyte) interactions in the ovary in order to provide<br />

the optimal environment for early embryonic development.<br />

PROJECT: (Clinical/Basic) Development <strong>of</strong> a Novel<br />

Artificial Reproductive Technology.<br />

R. O&G.<br />

Supervisors<br />

Dr Robert Gilchrist<br />

Associate Pr<strong>of</strong>essor Jeremy Thompson<br />

<strong>Project</strong> Background<br />

Oocyte In Vitro Maturation (IVM) is an adjunct<br />

procedure to IVF (in vitro fertilisation) and is a<br />

platform technology for the treatment <strong>of</strong> human<br />

infertility, the generation <strong>of</strong> embryos for livestock<br />

biotech and in the generation <strong>of</strong> stem cells by<br />

cloning. We have developed a new approach to<br />

oocyte IVM that dramatically increases embryo yield<br />

and pregnancy rates. This discovery has significant<br />

practical and commercial implications (see below).<br />

Further research is required to understand the basic<br />

molecular mechanisms in the oocyte and the<br />

somatic cells associated with the oocyte, which<br />

underpins this technology and leads to such an<br />

improvement in oocyte quality. In addition,<br />

translational research is required to allow the<br />

generation <strong>of</strong> an efficacious, robust, and safe<br />

technique and product that can be used in clinical<br />

and commercial practice.<br />

Potential projects include:<br />

1) Characterisation <strong>of</strong> the molecular mechanisms<br />

in cumulus cells and oocytes regulating induced<br />

oocyte maturation and embryo developmental<br />

competence<br />

2) Validating the efficacy and safety <strong>of</strong> induced<br />

oocyte IVM to allow rapid translation to<br />

commercial application and clinical practice.<br />

We are developing this technology in close<br />

collaboration with industry and clinical partners. The<br />

discovery is protected by our patent and we have<br />

licensed application <strong>of</strong> this technology in humans to<br />

a medical device manufacturer. We are conducting<br />

the first pre-clinical trial <strong>of</strong> this technology using<br />

human oocytes in an overseas laboratory. We also<br />

have an in-house business unit; IVF Vet Solutions<br />

(ACN), which is set up to capture new intellectual<br />

property opportunities and to supply our new<br />

products to the animal embryology sector. Students<br />

working on these projects will be closely associated<br />

with these commercial ventures.<br />

References<br />

Gilchrist RB and Thompson JG. (2007) Oocyte<br />

maturation: Emerging concepts and technologies to<br />

32 |


improve developmental potential in vitro.<br />

Theriogenology. 67(1):6-15<br />

Gilchrist RB, Lane M, Thompson JG. (2008) Oocytesecreted<br />

factors: regulators <strong>of</strong> cumulus cell function<br />

and oocyte quality. Human Reproduction Update<br />

14(2):159-177<br />

PROJECT: (Basic) Understanding the Functioning <strong>of</strong><br />

Oocyte-derived Growth Factors.<br />

R. O&G.<br />

Supervisors<br />

Dr David Mottershead<br />

Dr Robert Gilchrist<br />

<strong>Project</strong> Background<br />

Oocytes produce proteins <strong>of</strong> great importance for<br />

their ability to successfully be fertilized and develop<br />

into a viable embryo. The transforming growth factor<br />

β (TGF-β) superfamily <strong>of</strong> proteins consists <strong>of</strong> some 40<br />

members in mammals, many <strong>of</strong> which are essential<br />

for normal development and maintenance <strong>of</strong> the<br />

adult organism. Growth differentiation factor 9<br />

(GDF9) and bone morphogenetic factor 15 (BMP15)<br />

are two members <strong>of</strong> this family <strong>of</strong> proteins which are<br />

produced by oocytes and are critical for female<br />

fertility. The proteins GDF9 and BMP15 have great<br />

potential for improving currently used infertility<br />

treatments, as demonstrated previously by our group<br />

(see Yeo et al., 2008). In fact Dr. Gilchrist has<br />

patented this technology and has industry support for<br />

our research efforts on GDF9/BMP15. To take such<br />

work to the clinic/industry setting we need well<br />

characterized, stable preparations <strong>of</strong> these proteins.<br />

These and closely related proteins also have great<br />

potential in the area <strong>of</strong> stem cell research, both for<br />

the maintenance <strong>of</strong> stem cells in a pluripotent state<br />

as well as for directed differentiation <strong>of</strong> stem cells<br />

toward a particular lineage.<br />

Potential projects include:<br />

1) Recombinant production and purification <strong>of</strong><br />

various mutants <strong>of</strong> human GDF9 and BMP15,<br />

potentially including mutating the proteolytic<br />

processing site or the phosphorylation /<br />

glycosylation sites, to determine the impact on<br />

function <strong>of</strong> such mutations.<br />

2) Use <strong>of</strong> a bacterially expressed recombinant<br />

GDF/BMP binding protein to create an affinity<br />

column to purify TGF-β family members from<br />

physiological fluids, such as follicular fluid or<br />

seminal plasma. The aim <strong>of</strong> this project is to<br />

determine in what forms do the proteins<br />

GDF9/BMP15 exist in such biological fluids, as<br />

such information could give insight into the<br />

mechanisms used in vivo to regulate the<br />

bioactivity <strong>of</strong> these proteins.<br />

Techniques used can include:<br />

• use <strong>of</strong> mammalian cell lines for recombinant<br />

protein production<br />

• bacterial expression <strong>of</strong> proteins<br />

• protein purification, especially affinity<br />

purification and HPLC/FPLC<br />

• molecular biology, plasmid preps, PCR,<br />

transfection <strong>of</strong> mammalian cells<br />

• growth factor assays, various types,<br />

especially transcriptional reporter assays<br />

References<br />

Mottershead DG, Ritter LJ, Gilchrist RB. (2012)<br />

Signalling pathways mediating specific synergistic<br />

interactions between GDF9 and BMP15. Mol Hum<br />

Reprod. 18(3):121-8.<br />

Pulkki MM*, Mottershead DG*, et al. (2012) A<br />

covalently dimerized recombinant human bone<br />

morphogenetic protein-15 variant identifies bone<br />

morphogenetic protein receptor type 1B as a key cell<br />

surface receptor on ovarian granulosa cells.<br />

Endocrinology. 153(3):1509-18.<br />

Pulkki MM, Myllymaa S, et al, Mottershead DG. (2011)<br />

The bioactivity <strong>of</strong> human bone morphogenetic<br />

protein-15 is sensitive to C-terminal modification:<br />

characterization <strong>of</strong> the purified untagged processed<br />

mature region. Mol Cell Endocrinol. 332(1-2):106-15.<br />

Mottershead DG, Watson AJ. (2009) Oocyte peptides<br />

as paracrine tools for ovarian stimulation and oocyte<br />

maturation. Mol Hum Reprod. 15(12):789-94.<br />

33 |


Placental Development Laboratory<br />

Contact Person<br />

Pr<strong>of</strong>essor Claire Roberts<br />

Phone: 8313 3118<br />

claire.roberts@adelaide.edu.au<br />

Location: Medical School North and Lyell McEwin Hospital<br />

Supervisors<br />

Pr<strong>of</strong>essor Claire Roberts<br />

Dr Denise Furness<br />

Pr<strong>of</strong>essor Gus Dekker<br />

Dr Amada Highet<br />

Dr Tina Bianco-Miotto<br />

Our group’s major current focus in research is on the development <strong>of</strong> the placenta and the vessels which supply it. A<br />

number <strong>of</strong> pathologies <strong>of</strong> pregnancy, including up to 50% <strong>of</strong> miscarriages, preeclampsia (hypertension and<br />

proteinuria in pregnancy), intrauterine growth restriction (IUGR), preterm labour, unexplained stillbirth and placental<br />

abruption are characterised by an impaired cytotrophoblast invasion and inadequate response <strong>of</strong> the uterine spiral<br />

arteries to undergo physiological transformation. Together these conditions affect more than one quarter <strong>of</strong><br />

pregnant women in developed societies.<br />

We are investigating the cellular and molecular mechanisms underlying the formation <strong>of</strong> vascular connections<br />

between the mother and fetus. The uterine arterioles which supply the placenta are invaded by placental<br />

cytotrophoblast cells and are dramatically remodelled to produce dilated compliant vessels which allow the 12 fold<br />

increase in blood flow to the uterus which is essential for normal placental, and hence fetal, growth. However, the<br />

mechanisms by which physiological transformation <strong>of</strong> these vessels occurs are poorly understood. We are currently<br />

delineating the cellular and molecular interactions between maternal decidual leukocytes, endothelium and fetal<br />

trophoblast during the normal transformation <strong>of</strong> the arterioles which supply the placenta. Our work focuses on the<br />

role <strong>of</strong> insulin-like growth factor II (IGF-II) in placental invasion and how a variety <strong>of</strong> molecules interact with IGF-II in<br />

early pregnancy. We have discovered that IGF-II is likely to promote invasion by its interaction with a variety <strong>of</strong> other<br />

factors that all bind the type 2 IGF receptor (IGF2R) forming a complex. Activation <strong>of</strong> transforming growth factor<br />

(TGF)- β1, a cytokine that inhibits invasion, occurs on this complex and so too does the activation <strong>of</strong> renin, an<br />

enzyme that takes part in blood pressure control.<br />

Sufficient understanding <strong>of</strong> how the placenta invades in early gestation is a pre-requisite to understanding what<br />

goes wrong in pregnancy complications associated with poor placental invasion and function. Currently the<br />

treatments we have are symptomatic and are not used until the pregnant woman already has the disease. Our<br />

animal experiments have shown that treating the pregnant mother in early to mid pregnancy with IGF-II improves<br />

placental differentiation, transport function and increases fetal weight near term. It is essential that treatment begins<br />

in early-mid pregnancy to be efficacious. However, who should we treat? We are developing tests to predict which<br />

women are likely to develop pregnancy complications before symptoms arise. Subsequently, antenatal care could<br />

be tailored to suit individual women.<br />

34 |


PROJECT: (Basic) Factors that regulate placental<br />

trophoblast invasion and angiogenesis.<br />

R. O&G.<br />

Supervisor<br />

Pr<strong>of</strong>essor Claire Roberts<br />

<strong>Project</strong> Background<br />

Angiogenesis is an essential feature <strong>of</strong> placental<br />

morphogenesis. We have shown that a number <strong>of</strong><br />

factors expressed in the placenta promote placental<br />

trophoblast differentiation and invasion and some <strong>of</strong><br />

these may also be involved in angiogenesis which<br />

uses common molecules to promote endothelial<br />

development. The molecular pathway that we have<br />

discovered regulates placental invasion may or may<br />

not include a signal transduction pathway from the<br />

IGF2 receptor complex. This pathway may also<br />

regulate angiogenesis. This project will utilise several<br />

human trophoblast and endothelial cell lines to<br />

demonstrate the molecular pathways involved in<br />

invasion and angiogenesis. The project will involve<br />

cell culture, RT-PCR, ELISAs, immuno-histochemistry,<br />

video image analyses and western blotting. The<br />

student will gain skills in a variety <strong>of</strong> experimental<br />

techniques, as well as data management and<br />

statistical analyses.<br />

PROJECT: (Basic) Molecular mechanisms in actions <strong>of</strong><br />

transforming growth factor β1 (TGFβ1) inhibition <strong>of</strong><br />

trophoblast invasion and induction <strong>of</strong> terminal<br />

differentiation.<br />

R. O&G.<br />

Supervisor<br />

Pr<strong>of</strong>essor Claire Roberts<br />

<strong>Project</strong> Background<br />

Although it is known that TGFβ1 inhibits trophoblast<br />

invasion and likely induces terminal differentiation the<br />

detailed molecular mechanisms by which this occurs<br />

are not understood. Trophoblast invasion is an<br />

important process in early pregnancy that is<br />

responsible for dramatic re-modelling <strong>of</strong> the uterine<br />

vasculature to permit the 12-fold increase in uterine<br />

blood flow that occurs in normal pregnancy. This<br />

process is influenced by the low oxygen environment<br />

in first trimester placenta. In the second half <strong>of</strong><br />

pregnancy placentation is characterised by terminal<br />

differentiation <strong>of</strong> trophoblasts with the development<br />

<strong>of</strong> a large highly attenuated surface area for<br />

exchange. This project will explore the molecular<br />

mechanisms by which TGFβ1 regulates placental<br />

differentiation and will utilise several human<br />

trophoblast cell lines and first trimester and term<br />

human placental samples to demonstrate the<br />

molecular pathways involved. The project will involve<br />

cell culture, RT-PCR, ELISAs, immuno-histochemistry,<br />

video image analyses and western blotting. The<br />

student will gain skills in a variety <strong>of</strong> experimental<br />

techniques, as well as data management and<br />

statistical analyses.<br />

PROJECT: (Clinical/basic) Associations between<br />

genome damage, DNA repair enzyme gene<br />

polymorphisms and B vitamin status.<br />

R. O&G.<br />

Supervisor<br />

Pr<strong>of</strong>essor Claire Roberts<br />

Dr Denise Furness<br />

Pr<strong>of</strong>essor Gus Dekker<br />

<strong>Project</strong> Background<br />

We have recently shown that genome damage in<br />

circulating lymphocytes in mid pregnancy is more<br />

common in women who subsequently develop<br />

pregnancy complications in which placental<br />

insufficiency is a feature. These include preeclampsia,<br />

pre-term birth and intrauterine growth restriction. This<br />

project will determine whether genome damage is<br />

associated with polymorphisms in genes that encode<br />

DNA repair enzymes and whether severity <strong>of</strong><br />

damage is influenced by B vitamin status and<br />

circulating homocysteine. If time permits, tunnel<br />

assays to quantify genome damage in sperm <strong>of</strong><br />

partners <strong>of</strong> women who develop pregnancy<br />

complications will be performed and the results<br />

compared with those from men who father normal<br />

pregnancies. The student will gain skills in a variety <strong>of</strong><br />

experimental techniques including cytokinesis block<br />

micronucleus cytome assay, PCR and HRM<br />

genotyping, as well as data management and<br />

statistical analyses. The student will interact with<br />

obstetricians and midwives as well as scientists.<br />

PROJECT: (Basic) Cytokine gene expression by<br />

placental trophoblast cells under hypoxic conditions.<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Claire Roberts<br />

Dr Amanda Highet<br />

<strong>Project</strong> Backgound<br />

Trophoblast cells form the outer layer <strong>of</strong> a blastocyst,<br />

and provide nutrients to the embryo and develop the<br />

epithelial part <strong>of</strong> the placenta. They are formed<br />

during the first stage <strong>of</strong> pregnancy in a low oxygen<br />

environment where they invade into the maternal<br />

decidua and spiral arteries, proliferate, differentiate<br />

and secrete cytokines. In preeclampsia the<br />

trophoblast cells fail to adequately invade as<br />

required to promote materno-placental blood flow.<br />

The placenta has a local renin-angiotensin system<br />

from which angiotensin-II and its degradation<br />

product angiotensin-IV are produced. Angiotensins-II<br />

and -IV both stimulate the expression <strong>of</strong> nuclear<br />

factor-kappaB, a transcription factor which regulates<br />

gene expression <strong>of</strong> inflammatory cytokines such as<br />

35 |


interleukin (IL)-6 which could modify extracellular<br />

matrix composition, stimulate growth factor secretion<br />

and promote cell invasion. IL-10 on the other hand is<br />

an inhibitor <strong>of</strong> nuclear factor-kappaB and may be an<br />

important regulator <strong>of</strong> trophoblast inflammatory<br />

activity. We have recently shown that polymorphisms<br />

in the IL-10 and IL-6 genes known to decrease IL-10<br />

expression and up-regulate IL-6 expression are more<br />

frequent in babies (and therefore placentas) from<br />

pregnancies complicated by PE.<br />

This honours project will use cell culture and real-time<br />

PCR gene expression assays to show that 1)<br />

trophoblast cells upregulate expression <strong>of</strong> the IL-6<br />

gene and downregulate IL-10 under low oxygen<br />

conditions and that this is further enhanced in the<br />

presence <strong>of</strong> exogenous angiotensin-II and -IV and 2)<br />

IL-6 gene expression is increased and IL-10 is<br />

decreased in placenta samples from preeclamptic<br />

pregnancies compared with normal term placenta.<br />

This research aims to acquire new knowledge about<br />

trophoblast cell activity in the low oxygen<br />

environment by testing our hypothesis that proinflammatory<br />

cytokines are up-regulated by<br />

angiotensin-II and -IV under low oxygen conditions.<br />

Understanding the role <strong>of</strong> cytokines in hypoxic<br />

placenta and how they affect trophoblast activity<br />

could provide insight into the pathways that become<br />

deregulated in preeclampsia.<br />

PROJECT: (Basic) Sex differences in placental gene<br />

expression and the X chromosome.<br />

R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong> Claire Roberts<br />

Dr Tina Bianco-Miotto<br />

<strong>Project</strong> Background<br />

We aim to demonstrate for the first time that X<br />

chromosome genes include many that are important<br />

in placental development and function and<br />

maternal adaptation to pregnancy, and that some<br />

<strong>of</strong> these escape X inactivation and are therefore<br />

differentially expressed by the male and female<br />

placenta. Most research on this to date has been<br />

conducted in mice which emerging evidence shows<br />

is unlike the human. Hence we expect our data will<br />

challenge established dogma. We expect our<br />

research will explain the different patterns <strong>of</strong> fetal<br />

growth observed in males and females before birth.<br />

This project will utilise our access to human placental<br />

tissues and skill and knowledge base in placental<br />

development, fetal growth and epigenetics. This<br />

project will involve gene expression analyses <strong>of</strong> X<br />

chromosome genes as well as epigenetic analysis <strong>of</strong><br />

differentially expressed genes.<br />

Other associated projects may be negotiated<br />

36 |


Pregnancy and Development Group<br />

Contact Person<br />

Associate Pr<strong>of</strong>essor Vicki Clifton<br />

Phone: 8313 8321<br />

vicki.clifton@adelaide.edu.au<br />

Supervisors (photograph left to right)<br />

Dr Annette Osei-Kumah<br />

Dr Michael Stark<br />

Associate Pr<strong>of</strong>essor Vicki Clifton<br />

Dr Nicolette Hodyl<br />

Location: Medical School<br />

If you are interested in why males and females are different and if you wonder about how babies survive when they<br />

deliver preterm or are exposed to a stressful pregnancy then come and work with us. Our group conducts human<br />

studies on pregnant women and their babies. We are interested in what factors influence the growth <strong>of</strong> the fetus in<br />

utero and how in utero events affect the survival <strong>of</strong> the neonate and their health and development. We find males<br />

and females are different in how they respond to stress during pregnancy and this difference changes their growth<br />

and survival. Our work focuses on the mechanisms that confer a difference between male and female babies.<br />

PROJECT: (Clinical / Basic)<br />

R. O&G.<br />

Supervisors<br />

Dr Nicki Hodyl<br />

Dr Michael Stark Assoc<br />

A/Pr<strong>of</strong> Vicki Clifton<br />

<strong>Project</strong> Backgound<br />

Although neonatal infections affect 50% <strong>of</strong> infants<br />

born prior to 32 weeks gestation, the factors that<br />

contribute to this disturbing statistic are largely<br />

unknown.<br />

Current treatment regimes and recent RCTs have<br />

failed to decrease preterm neonatal infection rates,<br />

indicating knowledge gaps in our understanding <strong>of</strong><br />

neonatal infection. Evidence supports an intrinsic<br />

immaturity <strong>of</strong> the preterm neonatal immune system<br />

(including deficits in B cell, T cell and innate immune<br />

function).<br />

This does not, however, explain sexual dimorphisms in<br />

the susceptibility to infectious and inflammatory<br />

conditions in the neonatal period and beyond, with a<br />

significant male disadvantage.<br />

Our animal data indicates in-utero events program<br />

neonatal immunity. We hypothesise that immunecompetence<br />

in preterm neonates is determined by<br />

sex, birth weight and timing <strong>of</strong> exposure to antenatal<br />

betamethasone as well as products <strong>of</strong> placental<br />

inflammation.<br />

Understanding the developmental pr<strong>of</strong>ile <strong>of</strong> the<br />

human innate immune response may lead to<br />

interventions that avoid the potential sequelae <strong>of</strong><br />

infections in the preterm neonate. Placenta and cord<br />

blood will be collected from preterm and term<br />

deliveries. Cord blood monocytes will be exposed to<br />

pathogens and cytokines, and intracellular pathway<br />

activation characterised and related to episodes <strong>of</strong><br />

neonatal infection.<br />

<strong>Project</strong>s may include:<br />

1. Determination <strong>of</strong> the impact <strong>of</strong> antenatal steroid<br />

exposure on innate immune cell signalling pathways<br />

in the placenta and neonatal immune cells<br />

2. Identification <strong>of</strong> sex-specific pathways in placenta<br />

and / or human vascular endothelial cell lines that<br />

contribute to aberrant innate immune responses in<br />

preterm neonates<br />

PROJECT: (Clinical / Basic)<br />

R. O&G.<br />

Supervisors<br />

Dr Nicki Hodyl<br />

Dr Michael Stark Assoc<br />

A/Pr<strong>of</strong> Vicki Clifton<br />

Improved survival rates following preterm birth have<br />

not been accompanied by equivalent reduction in<br />

the incidence <strong>of</strong> chronic lung disease (CLD), the<br />

major morbidity associated with prematurity. Despite<br />

the contribution <strong>of</strong> perinatal environmental factors in<br />

its pathogenesis, antenatal interventions have not<br />

decreased the incidence <strong>of</strong> CLD, while postnatal<br />

interventions may be too late to be effective.<br />

37 |


Perinatal inflammatory cytokine exposure and<br />

reactive oxygen species (ROS) significantly contribute<br />

to the pathogenesis <strong>of</strong> chronic lung disease (CLD).<br />

Recent attention has focused on novel therapies that<br />

may modulate inflammatory responses and regulate<br />

the activation <strong>of</strong> placental inflammatory pathways<br />

and the initiation <strong>of</strong> patho-physiologic processes<br />

associated with preterm birth.<br />

These studies will address the role <strong>of</strong> these modifiable<br />

pathways in placental inflammatory cytokine and<br />

ROS production and anti-oxidant defence ex-vivo<br />

and interrogate sex-specific differences in their<br />

regulatory function with respect to the pathogenesis<br />

<strong>of</strong> CLD.<br />

<strong>Project</strong>s may include:<br />

1. The characterisation placental oxidative stress and<br />

immune responses to inflammatory stimuli.<br />

2. Identification <strong>of</strong> sex-specific pathways that control<br />

placental anti-inflammatory/anti-oxidant effects in<br />

placental explant culture and / or human vascular<br />

endothelial cell lines.<br />

38 |


Reproductive Biotechnology Group<br />

Supervisors<br />

Associate Pr<strong>of</strong>essor Mark Nottle<br />

Dr Ivan Vassiliev<br />

Location: Medical School<br />

Contact Person<br />

A/Pr<strong>of</strong> Mark Nottle<br />

Phone: 8313 4087<br />

mark.nottle@adelaide.edu.au<br />

The Reproductive Biotechnology group has an international reputation in the general areas <strong>of</strong> reproductive biology<br />

and the development <strong>of</strong> associated technologies for biomedical and agricultural applications. In collaboration with<br />

a number <strong>of</strong> university, institute and hospital research groups in Australia as well as overseas, current research is<br />

focused on developing organ, tissue and cell replacement therapies.<br />

PROJECT: (Basic) Making embryos for Research.<br />

R. O&G.<br />

<strong>Project</strong> Backgound<br />

Our Laboratory uses in vitro produced (IVP) pig<br />

embryos for much <strong>of</strong> its research. This uses abattoir<br />

derived ovaries from which oocytes are removed,<br />

matured in culture, fertilised and then cultured up to<br />

six days. While IVP can produce embryos for research<br />

purposes these are <strong>of</strong> a lesser quality than those that<br />

would be found in the reproductive tract. The current<br />

project aims to improve one or more steps in this<br />

process so that we can ultimately produce embryos<br />

<strong>of</strong> the same quality. <strong>Project</strong> s are available in this<br />

general area and will provide students with research<br />

experience across a range <strong>of</strong> areas including<br />

molecular and cell biology and embryology.<br />

PROJECT: (Basic) Reducing early embryonic loss.<br />

R. O&G.<br />

<strong>Project</strong> Backgound<br />

Around 30% <strong>of</strong> embryos are lost early in pregnancy,<br />

with the reasons for this unknown. Our Lab is<br />

examining the hypothesis that the majority <strong>of</strong> these<br />

losses are due to differences in oocyte quality as a<br />

result <strong>of</strong> changes in nutrition , season, disease etc. In<br />

particular we are examining how these influence<br />

oocyte quality and embryo development. <strong>Project</strong> s<br />

are available in this general area and will provide<br />

students with research experience across a range <strong>of</strong><br />

areas including molecular and cell biology and<br />

embryology.<br />

PROJECT: (Basic) Saving endangered species.<br />

R. O&G.<br />

<strong>Project</strong> Backgound<br />

Somatic cell nuclear transfer or cloning a sit more<br />

commonly know has been proposed as a way <strong>of</strong><br />

saving endangered species. However this approach<br />

is limited because not enough oocytes are<br />

available from endangered females. The use <strong>of</strong><br />

oocytes from another species has been proposed<br />

as a way <strong>of</strong> overcoming this shortage. However so<br />

called interspecies nuclear transfer is still in its<br />

infancy . The current project aims improve the<br />

efficiency <strong>of</strong> interspecies nuclear transfer.<br />

<strong>Project</strong>s are available in this general area and will<br />

provide students with research experience across a<br />

range <strong>of</strong> areas including molecular and cell biology<br />

and embryology<br />

PROJECT: (Basic) Isolation and characterisation <strong>of</strong><br />

embryonic stem cells.<br />

R. O&G.<br />

<strong>Project</strong> Backgound<br />

As human stem cell research advances there is an<br />

increasing need for a large animal model for<br />

bridging the gap between mouse studies and<br />

clinical trials. We have developed a new method for<br />

isolating ESCs and are using this to develop the pig<br />

as a large animal model for stem cell research. This<br />

includes the isolation and characterisation <strong>of</strong><br />

embryonic stem cells from a range <strong>of</strong> sources<br />

including parthenogenetic embryos. <strong>Project</strong> s are<br />

available in this general area and will provide<br />

students with research experience in cell biology,<br />

embryology and stem cells .<br />

39 |


Reproductive Cancer Research Group<br />

Contact Person<br />

Dr Carmela Ricciardelli<br />

Phone: 8313 8255<br />

carmela.ricciardelli@adelaide.edu.au<br />

Supervisors<br />

Dr Carmela Ricciardelli<br />

Associate Pr<strong>of</strong>essor Martin Oehler<br />

Dr Darryl Russell<br />

Dr Alison Elder<br />

Location: Medical School<br />

Dr Ricciardelli a Cancer Cell Biologist; joined the Department in April 2005. Dr Ricciardelli was awarded her Ph D in<br />

1996 (Flinders <strong>University</strong>) and has undertaken postdoctoral studies in the Department <strong>of</strong> Surgery, Flinders Medical<br />

Centre, South Australia (1996-2001) and Dame Roma Mitchell Cancer Research Laboratories, Hanson Institute, South<br />

Australia (2002-2005). She was awarded a Senior Fellowship with Cancer Council <strong>of</strong> South Australia (2004-2006). In<br />

2007 she was the recipient <strong>of</strong> the Hilda Farmer Fellowship in <strong>Faculty</strong> <strong>of</strong> <strong>Health</strong> <strong>Sciences</strong>, <strong>University</strong> <strong>of</strong> Adelaide which<br />

has enabled her to build a Reproductive cancer research group in Discipline <strong>of</strong> Obstetrics and Gynaecology at<br />

<strong>University</strong> <strong>of</strong> Adelaide. Dr Ricciardelli’s current research focuses on further understanding the cross talk between<br />

cancer cells and the extracellular matrix and mechanisms whereby extracellular matrix proteins promote spread <strong>of</strong><br />

breast, prostate and ovarian cancers.<br />

PROJECT: (Clinical/basic) Identification <strong>of</strong> novel<br />

proteins involved in ovarian cancer spread.<br />

.R. O&G.<br />

Supervisors<br />

Dr Carmela Ricciardelli<br />

Associate Pr<strong>of</strong>essor Martin Oehler<br />

Background<br />

The implantation <strong>of</strong> cancer cells onto the peritoneal<br />

surfaces is one <strong>of</strong> the first crucial steps in ovarian<br />

cancer metastasis. However it remains unclear which<br />

factors promote this process. Our recent studies have<br />

examined the ovarian cancer – peritoneal<br />

mesothelial cell interaction by means <strong>of</strong> latest<br />

proteomics technology. Tumor-mesothelial cell<br />

interaction have been explored in vitro by coculturing<br />

human ovarian cancer and peritoneal<br />

mesothelial cells. This novel strategy may lead to the<br />

identification <strong>of</strong> novel proteins likely to be<br />

mechanistically involved in implantation and<br />

peritoneal metastasis <strong>of</strong> ovarian cancer.. Several<br />

protein bands that are either increased or decreased<br />

in co-culture experiments with ovarian and<br />

mesothelial cells have been excised and will be<br />

identified using MALDI-TOF/TOF mass spectrometry.<br />

This study will characterise the expression and<br />

localization <strong>of</strong> several proteins modulated by ovarian<br />

cancer cell-peritoneal interactions in human ovarian<br />

cancer tissues. Proteins whose expression are<br />

modulated in human ovarian cancer cells compared<br />

to non-cancer tissues will be further characterised in<br />

future studies using functional assays (adhesion,<br />

migration and invasion assays). The identification <strong>of</strong><br />

novel proteins which promote adhesion and invasion<br />

<strong>of</strong> ovarian cancer cells to peritoneal cells may serve<br />

as therapeutic targets to inhibit ovarian cancer<br />

metastasis. This project will utilise a broad range <strong>of</strong><br />

techniques including tissue culture, motility, adhesion<br />

and invasion assays.<br />

Aim 1. Characterise the expression <strong>of</strong> specific<br />

proteins modulated by ovarian and peritoneal coculture<br />

in human ovarian cancer tissues<br />

Aim 2. Identify novel proteins that modulate ovarian<br />

cancer cell adhesion, migration and invasion through<br />

mesothelial cells<br />

PROJECT: (Basic) Versican as a target to inhibit<br />

cancer metastasis.<br />

.R. O&G.<br />

Supervisors<br />

Dr Carmela Ricciardelli<br />

Dr Darryl Russell<br />

<strong>Project</strong> Background<br />

There is increasing evidence to suggest that<br />

extracellular matrix (ECM) components play an<br />

active role in tumour progression and metastasis.<br />

Proteoglycans are major components <strong>of</strong> the ECM<br />

and have been shown to regulate cell adhesion, cell<br />

signalling, apoptosis, migration and invasion.<br />

Increased expression <strong>of</strong> the chondroitin sulfate (CS)<br />

proteoglycan, versican in the peritumoral stromal<br />

matrix is associated with a poor outcome in many<br />

cancers, including breast and ovarian carcinoma.<br />

Although there is the accumulating in vivo evidence<br />

40 |


that versican is pivotal in promoting cancer cell<br />

metastasis in different cancer types, the means <strong>of</strong><br />

preventing actions <strong>of</strong> versican in carcinomas have<br />

not been explored. This study we will evaluate using<br />

in vitro and in vivo breast cancer models whether<br />

selective versican inhibition by versican siRNA, in<br />

addition to drugs known to inhibit versican synthesis;<br />

genistein, budesonide, formoterol and montelukast,<br />

can inhibit breast cancer invasive behaviour and<br />

block breast cancer metastasis. This project will utilise<br />

a broad range <strong>of</strong> techniques including using in vitro<br />

motility, adhesion and invasion assays and an in vivo<br />

animal model <strong>of</strong> breast cancer.<br />

Aim 1. To assess the ability <strong>of</strong> pharmacological<br />

agents to inhibit versican synthesis and block invasive<br />

tumour cell behaviour in vitro<br />

Aim 2. To determine whether versican synthesis<br />

inhibitors reduce metastasis in in vivo cancer models<br />

PROJECT: (Basic) Role <strong>of</strong> ADAMTS1 and ADAMTS4<br />

proteases in prostate cancer metastasis.<br />

.R. O&G.<br />

Supervisors<br />

Dr Darryl Russell<br />

Dr Carmela Ricciardelli<br />

<strong>Project</strong> Background<br />

Metastatic spread to the lymph node and bone is<br />

the most significant cause <strong>of</strong> relapse and mortality in<br />

prostate cancer patients. The molecular processes<br />

which mediate metastasis <strong>of</strong> cancers to the bone<br />

have been a strong focus <strong>of</strong> recent research. The<br />

Adamts (adamalysin-thrombospondin) proteases are<br />

a family <strong>of</strong> metalloproteinases involved in<br />

extracellular matrix (ECM) processing. Adamts1 and<br />

Adamts4 are major bone matrix remodelling<br />

proteases. Substrates include versican a prominant<br />

stromal matrix component associated with breast<br />

and prostate tumour progression. A recent report links<br />

Adamts1 to the bone metastatic gene signature. This<br />

project will focus on Adamts1 and 4 for which there<br />

growing evidence <strong>of</strong> a role in prostate cancer<br />

progression. This project will utilise a broad range <strong>of</strong><br />

techniques using including immunohistochemistry,<br />

motility, adhesion and invasion assays and an in vivo<br />

animal model <strong>of</strong> prostate cancer.<br />

Aim 1. To show that Adamts proteases promotes<br />

invasive migratory behaviour in prostate tumor cell<br />

lines in vitro.<br />

Aim 2. To elucidate the association between<br />

Adamts1 or Adamts4 protein levels and metastatic<br />

disease in human prostate cancer and an in vivo<br />

mouse model.<br />

PROJECT: (Basic) Investigation <strong>of</strong> a therapeutic target<br />

for ovarian cancer chemoresistance.<br />

.R. O&G.<br />

Supervisors<br />

Dr Carmela Ricciardelli<br />

Dr Alison Elder<br />

A/Pr<strong>of</strong> Martin Oehler<br />

<strong>Project</strong> Background<br />

Ovarian cancer is the most lethal gynaecological<br />

cancer and the development <strong>of</strong> chemoresistance<br />

results in significant mortality. Over 80% <strong>of</strong> patients<br />

treated via chemotherapy eventually relapse and<br />

become resistant to chemotherapy. A well<br />

established cause <strong>of</strong> chemoresistance involves the<br />

increased expression <strong>of</strong> ABC membrane transporter<br />

proteins, which act to decrease levels <strong>of</strong><br />

chemotherapy drugs within cells. Although effective<br />

in in vitro studies, clinical trials investigating ABC<br />

transporter inhibitors have failed to significantly<br />

improve patient survival, mainly due to focus on only<br />

a limited number <strong>of</strong> ABC transporters. Innovative<br />

treatment strategies to overcome chemoresistance<br />

are therefore urgently required. Our recent studies<br />

have linked chemoresistance with the production <strong>of</strong><br />

the extracellular matrix component hyaluronan (HA).<br />

HA plays an important role in promoting attachment<br />

<strong>of</strong> cancer cells to peritoneal cells via interactions with<br />

the HA receptor, CD44. We have shown that HA can<br />

increase the expression <strong>of</strong> ABC transporters in ovarian<br />

cancer cell lines expressing the HA receptor, CD44,<br />

and thereby induce resistance to the<br />

chemotherapeutic drug, carboplatin. This study will<br />

use samples prospectively collected between 2008<br />

and 2012 to determine, via ELISA assay, if serum HA<br />

levels at diagnosis are increased in patients who do<br />

not respond to first line chemotherapy, and whether<br />

it is predictive <strong>of</strong> recurrent or overall survival. Tissue<br />

sections from serous ovarian carcinoma patients will<br />

be immunostained with specific antibodies to various<br />

ABC transporter protein levels to determine whether<br />

ABC transporter expression correlates with response<br />

to chemotherapy and impacts on recurrence free<br />

and overall survival. HA inhibitors will also be used to<br />

determine whether they are able to significantly<br />

increase the cytotoxic effect <strong>of</strong> chemotherapeutic<br />

drugs on ovarian cancer cells, providing a strong<br />

rationale for combining HA inhibitors with<br />

chemotherapy drugs in future clinical studies. This<br />

project will utilise a broad range <strong>of</strong> techniques<br />

including enzymatic assays, immunohistochemistry,<br />

qPCR and in vitro and in vivo animal models <strong>of</strong><br />

ovarian cancer.<br />

Aim 1. Identify whether HA levels at diagnosis can<br />

predict therapeutic response and patient survival.<br />

Aim 2. Characterise whether ABC transporter<br />

expression can predict patient outcome.<br />

Aim 3. Determine whether the combination <strong>of</strong> HA<br />

inhibitors with chemotherapy drugs increases survival<br />

41 |


when compared to treatment with the<br />

chemotherapeutic drugs alone.<br />

Publications<br />

1. Ricciardelli, C., Brooks, J. H., Suwiwat, S.,<br />

Sakko, A. J., Mayne, K., Raymond, W. A.,<br />

Seshadri, R., LeBaron, R. G., and Horsfall, D.<br />

J. Regulation <strong>of</strong> stromal versican expression<br />

by breast cancer cells and importance to<br />

relapse-free survival in patients with nodenegative<br />

primary breast cancer. Clin<br />

Cancer Res, 8: 1054-1060, 2002.<br />

2. Sakko, A. J., Ricciardelli, C., Mayne, K.,<br />

Suwiwat, S., LeBaron, R. G., Marshall, V. R.,<br />

Tilley, W. D., and Horsfall, D. J. Modulation <strong>of</strong><br />

prostate cancer cell attachment to matrix<br />

by versican. Cancer Res, 63: 4786-4791,<br />

2003.<br />

3. Suwiwat, S., Ricciardelli, C., Tammi, R.,<br />

Tammi, M., Auvinen, P., Kosma, V. M.,<br />

LeBaron, R. G., Raymond, W. A., Tilley, W. D.,<br />

and Horsfall, D. J. Expression <strong>of</strong> extracellular<br />

matrix components versican, chondroitin<br />

sulfate, tenascin, and hyaluronan, and their<br />

association with disease outcome in nodenegative<br />

breast cancer. Clin Cancer Res,<br />

10: 2491-2498, 2004.<br />

4. Ricciardelli C & Rodgers RJ Matrix <strong>of</strong> ovarian<br />

tumours Seminars in Reproductive Medicine<br />

24:270-82, 2006<br />

5. Ricciardelli C, Russell D, Ween MP, Mayne K,<br />

Byers S, VR Marshall Tilley WD and Horsfall<br />

DJ. Formation <strong>of</strong> hyaluronan-and versicanrich<br />

pericellular matrix by prostate cancer<br />

cells promotes cell motility J Biol Chem:<br />

282:10814-10825, 2007.<br />

6. Sakko AJ, Butler MS, Byers S, Reinboth BJ,<br />

Stahl J, Kench JG, Horvath LG, Sutherland<br />

RL, Stricker PD, Henshall SM, Marshall VR,<br />

Tilley WD, Horsfall DJ & Ricciardelli C.<br />

Immunohistochemical level <strong>of</strong> unsulfated<br />

chondroitin disaccharides in the cancer<br />

stroma is an independent predictor <strong>of</strong><br />

prostate cancer relapse. Cancer<br />

Biomarkers, Epidemiology & Prevention, 17:<br />

2488-2497, 2008<br />

7. Ricciardelli C, Sakko AJ, Stahl J, Tilley WD,<br />

Marshall VR & Horsfall DJ. Prostatic<br />

chondroitin sulfate is increased in patients<br />

with metastatic disease but does not<br />

predict survival outcome. Prostate 69:761-9,<br />

2009<br />

8. Ricciardelli C, Sakko AJ, Ween MP, Russell<br />

DL & Horsfall DJ. The biological role and<br />

regulation <strong>of</strong> versican levels in cancer.<br />

Cancer and Metastasis Reviews 28:233-45,<br />

2009<br />

9. Ricciardelli C & Oehler MK. Diverse<br />

molecular pathways in ovarian cancer and<br />

their clinical significance. Maturitas 62:270-5,<br />

2009<br />

10. Ween MP, Lokman, NA, H<strong>of</strong>fmann P,<br />

Rodgers RJ, Ricciardelli C* & Oehler MK*.<br />

Transforming growth factor-beta induced<br />

protein secreted by peritoneal cells<br />

increases the metastatic potential <strong>of</strong><br />

ovarian cancer cells. International Journal<br />

Cancer, 128:1570-84, 2011, * co-directed<br />

11. Ween MP, Hummitzsch K, Rodgers RJ,<br />

Oehler MK & Ricciardelli C. Versican<br />

induces a pro-metastatic ovarian cancer<br />

cell behavior which can be inhibited by<br />

small hyaluronan oligosaccharides. Clinical<br />

and Experimental Metastasis, 28:113-25,<br />

2011<br />

12. Ricciardelli C, Frewin KM, Tan IA, Williams<br />

ED, Opeskin K, Pritchard MA, Ingman WV<br />

and Russell DL. The Adamts1 protease gene<br />

is required for mammary tumor growth and<br />

metastasis. American Journal <strong>of</strong> Pathology,<br />

179:3075-85, 2011<br />

42 |


Reproductive Endocrinology Group<br />

Supervisors<br />

Contact Person<br />

Louise Hull<br />

Phone: 0403 9933312<br />

louise.hull@adelaide.edu.au<br />

Dr Louise Hull<br />

Dr Jonathan McGuane<br />

Location: Medical School<br />

PROJECT: (Clinical or clinical/basic)<br />

.R. O&G.<br />

Endometriosis<br />

<strong>Project</strong> Background<br />

Endometriosis is another common condition in young<br />

women and its diagnosis and aetiology is uncertain.<br />

Dr Hull has pioneered new methods for diagnosis and<br />

treatment and uses cutting edge technology in her<br />

research group. Opportunity exists for medical<br />

students to combine patient interaction with high<br />

quality laboratory methods in her group.<br />

43 |


Reproductive Immunology Group<br />

Contact Person<br />

Pr<strong>of</strong>essor Sarah Robertson<br />

Phone: 8313 4094<br />

sarah.robertson@adelaide.edu.au<br />

Supervisors<br />

Pr<strong>of</strong>essor Sarah Robertson<br />

Dr David Sharkey<br />

Dr Anne Macpherson<br />

Pr<strong>of</strong>essor Claire Roberts<br />

Dr Kerri Diener<br />

Location: Medical School<br />

Sarah Robertson is a NHMRC Senior Research Fellow and Head <strong>of</strong> the Reproductive Immunology group in the<br />

Research Centre for Reproductive <strong>Health</strong>. Her research focus is early pregnancy, particularly the cytokine biology<br />

and immunology <strong>of</strong> the uterus and early embryo development. Her team is made up <strong>of</strong> 6 postdocs and 5<br />

postgraduate students, who work together to explore the importance <strong>of</strong> the peri-conceptual immune environment<br />

in optimal embryo implantation and placental development, and programming fetal development and health after<br />

birth.<br />

Research in the Reproductive Immunology group centres on three related themes:<br />

• The roles <strong>of</strong> cytokines and leukocytes in the events <strong>of</strong> embryo development during early pregnancy.<br />

• The impact <strong>of</strong> the maternal immune response on success and quality <strong>of</strong> embryo implantation and reproductive<br />

outcome.<br />

• Male seminal fluid signalling in the female reproductive tract and significance for reproductive events and<br />

immunity to sexually transmitted infection.<br />

We seek to unravel the immune and cytokine networks <strong>of</strong> early pregnancy to understand how maternal immune<br />

tolerance to pregnancy is established, and how failure in this process contributes to infertility, miscarriage and<br />

pathologies <strong>of</strong> pregnancy in women. Our work also has applications in animal breeding industries where early<br />

pregnancy loss is a significant constraint.<br />

Keywords<br />

Immunology, cytokines, uterus, embryo, pregnancy<br />

PROJECT: (Basic) T-Regulatory (Treg) cells and<br />

activiation <strong>of</strong> immune tolerance during early<br />

pregnancy.<br />

Theme title: Pregnancy immunology<br />

.R. O&G.<br />

Supervisor<br />

Pr<strong>of</strong>essor Sarah Robertson<br />

<strong>Project</strong> Background<br />

To allow embryo implantation and successful<br />

pregnancy, the maternal immune system must<br />

become ‘tolerant’ to paternal transplantation<br />

antigens. Treg cells are now implicated as key cells<br />

mediating maternal immune tolerance. Discoveries<br />

in our laboratory show that semen plays an important<br />

role in establishing functional tolerance to male<br />

transplantation antigens during early pregnancy. We<br />

now seek to investigate the role <strong>of</strong> seminal factors in<br />

activating and expanding Treg cells in preparation<br />

for embryo implantation. The aim <strong>of</strong> this project is to<br />

investigate in mice the molecular events involved in<br />

activating Treg cells after mating. In particular the<br />

importance <strong>of</strong> seminal plasma TGFbeta and the role<br />

<strong>of</strong> male MHC antigens in semen will be investigated.<br />

The project will employ cytokine null mutant mouse<br />

models, linked with state-<strong>of</strong>-the-art digital flow<br />

cytometry (FACS), and quantitative RT-PCR for the<br />

44 |


Treg fate-determining transcription factor Foxp3. The<br />

findings will help us understand the induction phase<br />

<strong>of</strong> the maternal immune response permitting<br />

successful pregnancy, and have broader relevance<br />

to the transmission <strong>of</strong> STDs and immune-mediated<br />

pathologies linked with infertility.<br />

Keywords<br />

Immunology / T regulatory cells / pregnancy<br />

PROJECT: (Clinical/Basic) Novel seminal plasma<br />

cytokines mediating male-female signalling at<br />

insemination.<br />

Theme title: Seminal fluid signalling<br />

.R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Sarah Robertson<br />

Dr David Sharkey<br />

<strong>Project</strong> Background<br />

Cytokines present in male seminal plasma play<br />

essential roles in ‘conditioning’ the female immune<br />

response for pregnancy, through activating immune<br />

tolerance to paternal transplantation antigens<br />

present on the conceptus. The active factors present<br />

in seminal plasma include members <strong>of</strong> the TGFβ<br />

family <strong>of</strong> cytokines. We have identified some <strong>of</strong><br />

these molecules but the full array <strong>of</strong> active factors<br />

and their synergistic interactions remain to be fully<br />

elucidated. The aim <strong>of</strong> this project is to identify new<br />

molecules present in seminal plasma that interact<br />

with female reproductive tract cells. The project will<br />

utilize in vitro models <strong>of</strong> male-female signalling,<br />

involving human cervical cell lines. Quantitative RT-<br />

PCR and cytokine immunoassay will be used to<br />

characterise cervical cell responsiveness to male<br />

factor regulation. Novel factors will be identified<br />

using factor-specific neutralising antibodies and<br />

quantified in an existing bank <strong>of</strong> human seminal<br />

plasma samples. We expect the results will provide<br />

new understanding <strong>of</strong> the factors mediating seminal<br />

signalling, and will assist in commercial development<br />

<strong>of</strong> novel diagnostic tools for infertility and subfertility in<br />

men.<br />

Keywords<br />

Seminal fluid / female reproductive tract / cytokines<br />

PROJECT: (Clinical/Basic) Cytokine regulation <strong>of</strong><br />

embryo development.<br />

Theme title: Embryo development<br />

.R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Sarah Robertson<br />

Dr Anne Macpherson<br />

<strong>Project</strong> Background<br />

The cytokine granulocyte-macrophage colonystimulating<br />

factor (GM-CSF) is produced in the<br />

oviduct and uterus during early pregnancy where it<br />

acts as a potent physiological regulator <strong>of</strong> human<br />

pre-implantation embryo development. GM-CSF<br />

added to embryo culture dramatically improves<br />

viability and developmental competence <strong>of</strong> human<br />

IVF embryos. Experiments in mouse and human have<br />

begun to unravel the mechanisms <strong>of</strong> action <strong>of</strong> GM-<br />

CSF. Microarray transcription pr<strong>of</strong>iling studies have<br />

revealed that in the absence <strong>of</strong> GM-CSF, apoptosis<br />

genes and several intracellular stress pathways may<br />

be activated. The aim <strong>of</strong> this project is to further<br />

investigate how GM-CSF acts to promote embryo cell<br />

survival, by in situ imaging <strong>of</strong> an array <strong>of</strong> proteins<br />

involved in the cellular stress response and in<br />

regulating the apoptosis cascade. The principal<br />

techniques will be in vitro embryo culture and<br />

confocal microscope analysis <strong>of</strong> immuno-labelled<br />

embryos, with potential for utilization <strong>of</strong> anti-sense<br />

mRNA strategies for investigating the effects <strong>of</strong><br />

perturbing GM-CSF signaling pathways in vitro. The<br />

experiments are expected to provide new<br />

information on the molecular pathways by which<br />

GM-CSF influences optimal embryo development.<br />

The research will assist in development <strong>of</strong> improved<br />

embryo culture processes in human IVF clinics.<br />

Keywords<br />

Embryo / cytokines / GM-CSF<br />

PROJECT: (Basic) TGFβ1 gene dosage effect on<br />

placental morphogenesis and fetal growth.<br />

Theme title: Pregnancy immunology<br />

.R. O&G.<br />

Supervisors<br />

Pr<strong>of</strong>essor Sarah Robertson<br />

Pr<strong>of</strong>essor Claire Roberts<br />

<strong>Project</strong> Background<br />

Transforming growth factor β1 (TGFβ1) is a key<br />

cytokine implicated in embryo implantation and<br />

development <strong>of</strong> the placenta. Female mice<br />

homozygous for a null mutation in the TGFβ1 gene<br />

are almost fully infertile, with impaired ovarian<br />

function and arrested pre-implantation embryo<br />

development. In heterozygote breeding pairs, the<br />

proportion <strong>of</strong> pups either homozygote or<br />

heterozygote for the null mutation is substantially<br />

fewer than the expected Mendelian frequency,<br />

indicating detrimental effects <strong>of</strong> reduced TGFβ1<br />

synthesis in the conceptus. The aim <strong>of</strong> this project is<br />

to determine whether the TGFβ1 gene dosage effect<br />

is mediated through impaired development <strong>of</strong> the<br />

placental tissue reflected in reduced capacity to<br />

support the developing fetus. The project will utilize<br />

TGFβ1 null mutant mice on immunodeficient and<br />

immune competent backgrounds. Placental tissue<br />

45 |


will be analysed by computer-assisted histological<br />

and morphometric analysis at key milestones in<br />

development, and quantitative RT-PCR based<br />

analysis <strong>of</strong> mRNA expression for placental genes<br />

implicated in development and function. Fetal tissue<br />

will be genotyped by diagnostic PCR and growth<br />

and survival rates <strong>of</strong> progeny <strong>of</strong> different genotypes<br />

will be determined. The potential contribution <strong>of</strong><br />

TGFβ1 in immune-mediated placental dysfunction will<br />

be analysed by assessing Treg cells, which are<br />

implicated in maternal immune tolerance <strong>of</strong> the<br />

conceptus. The results will provide a better<br />

understanding <strong>of</strong> the physiological role <strong>of</strong> TGFβ1 in<br />

establishing and maintaining pregnancy, and will<br />

inform upon the possible contribution <strong>of</strong> gene<br />

polymorphisms in TGFβ1, or other environmental<br />

perturbations in TGFβ1, in infertility and pathologies <strong>of</strong><br />

human pregnancy.<br />

Keywords<br />

Placenta / cytokines / TGFβ<br />

PROJECT: (Basic) Can macrophages aid embryo<br />

implantation for pregnancy success?<br />

.R. O&G.<br />

Supervisor<br />

Pr<strong>of</strong>essor Sarah Robertson<br />

Introduction to the laboratory<br />

To implant and establish the connections that are<br />

vital for further development, the early embryo must<br />

attach to and then breech the barrier posed by the<br />

epithelium <strong>of</strong> the maternal reproductive tract (Figure<br />

1).<br />

Infertility, miscarriage, and other pathologies <strong>of</strong><br />

pregnancy stemming from ‘shallow’ placental<br />

development are known to have their origins at the<br />

time <strong>of</strong> embryo implantation into the uterus.<br />

However the biological process <strong>of</strong> embryo<br />

implantation is not understood.<br />

While it is clear that precisely regulated changes in a<br />

wide array <strong>of</strong> parameters including glycosylated<br />

proteins are associated with achieving a receptive<br />

state for embryo implantation (‘window <strong>of</strong><br />

implantation’), how these changes occur remains to<br />

be clarified.<br />

Macrophages are particularly prominent amongst<br />

the large and dynamic population <strong>of</strong> inflammatory<br />

cells that traffic through the uterus and are able to<br />

secrete an array <strong>of</strong> potent regulatory molecules.<br />

Figure 1. Epithelial cells stained with lectin in mouse<br />

uterus (MJJ)<br />

Their recruitment, differentiation and state <strong>of</strong><br />

activation fluctuate over the course <strong>of</strong> the oestrous<br />

cycle and particularly during early pregnancy, under<br />

the direction <strong>of</strong> potent local determinants including<br />

cytokines and chemokines, and components <strong>of</strong> the<br />

extracellular matrix (ECM). Amongst these regulators<br />

<strong>of</strong> uterine macrophages, our interest has centred on<br />

the role <strong>of</strong> seminal fluid, which acts at the time <strong>of</strong><br />

insemination to elicit a rapid and dramatic influx <strong>of</strong><br />

macrophages and other activated inflammatory<br />

cells into the female reproductive tract (Figure 2).<br />

The success and quality <strong>of</strong> pregnancy is<br />

compromised if macrophage populations are<br />

disrupted in females by preventing exposure to<br />

seminal plasma. When the seminal vesicle is<br />

surgically removed from rodents implantation rates<br />

are reduced, and fetuses have a high incidence <strong>of</strong><br />

growth retardation secondary to changes in<br />

placental development. We have postulated that<br />

the inflammatory cells recruited during the response<br />

to seminal plasma exposure might impact on uterine<br />

receptivity, particularly the glycobiology <strong>of</strong> the<br />

epithelium associated with pregnancy success.<br />

The student will develop specialised knowledge <strong>of</strong><br />

the implantation process and early pregnancy. In<br />

addition, they will develop skills in real-time RT-PCR,<br />

immunohistochemistry and protein blotting.<br />

Hypothesis<br />

1. Successful embryo implantation is dependent on<br />

communication between uterine macrophages and<br />

luminal epithelial cells in the uterus during early<br />

pregnancy.<br />

2. Morphological and biochemical changes<br />

associated with epithelial cell receptivity, including<br />

their adhesive properties are regulated by paracrine<br />

and juxacrine signals <strong>of</strong> macrophage origin.<br />

3. Reduced abundance or altered functional<br />

phenotype in endometrial macrophages during the<br />

peri-implantation phase <strong>of</strong> pregnancy impairs<br />

embryo implantation and placental morphogenesis,<br />

with adverse consequences for fetal development<br />

and health after birth.<br />

46 |


PROJECT: (Basic) Viral Recognition within the female<br />

reproductive tract.<br />

.R. O&G.<br />

Macrophage<br />

Supervisor/contact person<br />

Dr Kerri Diener<br />

Phone 8313 3376<br />

kerri.diener@adelaide.edu.au<br />

est day 1 day 4 day 1 SV-<br />

Epithelium<br />

<strong>Project</strong> Background<br />

Reproduction in placental mammals represents an<br />

immunological paradox. This is because the<br />

ejaculates <strong>of</strong> males, and indeed the developing<br />

fetus, contain male-specific antigens which are<br />

recognised as foreign by the recipient female. Yet,<br />

the female does not respond destructively, nor is she<br />

immunologically primed to reject future mating<br />

events with the same male partner. However, this<br />

state <strong>of</strong> immunological tolerance that exists between<br />

the mother and her mate, and developing fetus,<br />

should not impinge on her capacity to respond<br />

appropriately to viral challenges, particularly since<br />

there is a substantial body <strong>of</strong> evidence to suggest<br />

that viral infections can compromise female fertility<br />

and reproductive outcomes, causing pregnancy<br />

complications including intrauterine growth restriction<br />

(IUGR), pre-eclampsia, fetal deformities, preterm<br />

labour and abortion, as well as postnatal morbidity<br />

such as cerebral palsy.<br />

A B C<br />

Figure 2. Macrophages in the uterus during early pregnancy and the effect <strong>of</strong> exposure to seminal<br />

fluid. Immunohistochemistry to detect macrophages is shown at estrus (A), on day 1 (B), and on<br />

day 4 after mating with a male. The post-mating inflammatory response is ablated on day 1 after<br />

mating with a male from which the seminal vesicle glands have been surgically removed (D).<br />

D<br />

In this project, established mouse models will be used<br />

to explore these issues, under the premise that it is the<br />

process <strong>of</strong> early immune recognition and subsequent<br />

response to viral challenges by the mother and/or<br />

the developing fetus which are a major contributing<br />

factor in causing these induced pathologies. A<br />

variety <strong>of</strong> genetically-modified mice made deficient<br />

for the expression <strong>of</strong> various early immune<br />

recognition receptors will be used to determine the<br />

mechanism behind viral recognition within the<br />

female reproductive tract.<br />

Students who have undertaken an honours project in<br />

the Reproductive Immunology Lab have continued<br />

on to complete PhDs, or work as research assistants.<br />

Previous honours students have presented their<br />

research at local ASMR conferences, and I have<br />

presented aspects <strong>of</strong> this work at national and<br />

international conferences.<br />

References<br />

Jones CJ, Aplin JD. Glycosylation at the<br />

fetomaternal interface: does the glycocode play a<br />

critical role in implantation? Glycoconj J.<br />

2009<br />

Apr;26(3):359-66. Epub 2008 Aug 3. Review.<br />

Aplin JD, Singh H. Bioinformatics and transcriptomics<br />

studies <strong>of</strong> early implantation.<br />

Ann N Y Acad Sci.<br />

2008 Apr;1127:116-20. Review<br />

47 |


Discipline <strong>of</strong> Paediatrics<br />

Women’s and Children’s Hospital<br />

Lyell McEwin Hospital<br />

Royal Adelaide Hospital<br />

“The Discipline <strong>of</strong> Paediatrics is located at the Women's<br />

and Children's Hospital, and includes the Vaccinology<br />

and Immunology Research Trials Unit. <strong>Honours</strong> projects in<br />

Paediatrics are also supervised by our multiple affiliates<br />

including members <strong>of</strong> other hospital departments and<br />

researchers in the Children's Research Centre. The<br />

Discipline has a wide range <strong>of</strong> clinical expertise and a<br />

strong commitment to clinical service in the Division <strong>of</strong><br />

Paediatric Medicine at the Children, Youth and Women's<br />

<strong>Health</strong> Service. Our clinical staff provide services in<br />

multiple fields, including diabetes, endocrinology,<br />

gastroenterology, pulmonary medicine, sleep medicine,<br />

immunology, rheumatology, allergy and child &<br />

adolescent psychiatry.”<br />

Pr<strong>of</strong>essor Jenny Couper<br />

Head <strong>of</strong> Discipline <strong>of</strong> Paediatrics<br />

48 |


Child Nutrition Research Centre (CNRC)<br />

Supervisors<br />

Pr<strong>of</strong>essor Maria Makrides<br />

Pr<strong>of</strong>essor Robert Gibson<br />

Dr Jo Zhou<br />

Dr Carmel Collins<br />

Location: Women’s and Children’s Hospital, Flinders Medical Centre<br />

and Foodplus Research Centre<br />

The overall aim <strong>of</strong> the Child Nutrition Research Centre is to optimise the nutritional intake <strong>of</strong> mothers and their infants,<br />

whether the infants are born preterm or at term. The rationale for our research is that improvements in nutritional<br />

status will optimise the growth, neurological development and immune system <strong>of</strong> all infants. Reducing susceptibility<br />

to allergy and associated diseases will have beneficial effects on infant quality <strong>of</strong> life as well as on whole-<strong>of</strong>-life<br />

health care costs.<br />

PROJECT: (Clinical) Neonatal Nutrition Research<br />

Priority Setting – partnership with parents and<br />

clinicians.<br />

Supervisors/Contact<br />

Dr Carmel Collins<br />

Carmel.Collins@health.sa.gov.au<br />

Pr<strong>of</strong> Maria Makrides<br />

maria.makrides@adelaide.edu.au<br />

<strong>Project</strong> Background<br />

The aim <strong>of</strong> the Neonatal Nutrition research priority<br />

setting partnership is to identify the unanswered<br />

questions around the nutritional management <strong>of</strong><br />

preterm infants that are important to parents and<br />

clinicians. The honours project will enable parents<br />

and clinicians caring for preterm infants to identify<br />

and prioritise what aspects <strong>of</strong> nutritional<br />

management <strong>of</strong> these vulnerable infants matters to<br />

them.<br />

Despite improved survival, preterm infants remain at<br />

high risk for long-term cognitive and educational<br />

problems. Their developmental quotient at school<br />

age is 11 points lower than term born children<br />

placing an extra burden on health and education<br />

resources. There is strong evidence that the failure to<br />

meet the nutritional needs <strong>of</strong> preterm infants is partly<br />

to blame for these developmental shortfalls. Setting<br />

research priorities <strong>of</strong> importance to those most<br />

closely involved with the care <strong>of</strong> preterm infants<br />

ensures research is grounded in clinical reality and<br />

addresses the concerns <strong>of</strong> those closest to these<br />

infants, the parents.<br />

Expected Outcomes<br />

Establish research priorities for the nutritional<br />

management <strong>of</strong> preterm infants.<br />

Reference<br />

Collins CT, Chua MC, Rajadurai VS, McPhee AJ, Miller LN,<br />

Gibson RA, Makrides M. Higher protein and energy intake<br />

is associated with increased weight gain in pre-term<br />

infants. J Paediatr Child <strong>Health</strong>. 2011 Mar;46(3):96-102.<br />

PROJECT: (Clinical) Innovative methods to maintain<br />

large cohorts in long-term studies.<br />

Supervisors/Contact<br />

Dr Carmel Collins<br />

Carmel.Collins@health.sa.gov.au<br />

Pr<strong>of</strong> Maria Makrides<br />

maria.makrides@adelaide.edu.au<br />

<strong>Project</strong> Background<br />

The CNRC is conducting follow up studies <strong>of</strong> women<br />

and children who participated in large scale dietary<br />

randomised controlled trials. Incomplete follow-up<br />

biases the results <strong>of</strong> a study, for e.g. when<br />

participants who are ‘lost to follow-up’ are different<br />

from those who complete follow-up or when there is<br />

differential dropout rates between study groups.<br />

Innovative strategies are needed to maintain families<br />

in the study across the years.<br />

Expected Outcomes<br />

Determine factors influencing continued<br />

participation in follow-up studies and develop<br />

strategies to support long-term involvement.<br />

49 |


PROJECT: (Basic) Measurement <strong>of</strong> energy metabolites<br />

in dried blood spots.<br />

Supervisors<br />

Pr<strong>of</strong> Robert Gibson<br />

Pr<strong>of</strong> Maria Makrides<br />

http://www.adelaide.edu.au/foodplus/<br />

http://www.wchri.com.au/<br />

Contact Person<br />

Pr<strong>of</strong> Robert Gibson<br />

robert.gibson@adelaide.edu.au.<br />

Please use the subject heading ‘<strong>Honours</strong> <strong>Project</strong>’.<br />

<strong>Project</strong> Background<br />

Assessing the nutritional status <strong>of</strong> preterm infants is <strong>of</strong><br />

vital importance to evaluating their overall health. In<br />

human trials, blood sample analysis is an integral part<br />

<strong>of</strong> assessing physiological and immune function in<br />

participants. This blood collection is invasive and<br />

analysis involves using volumes <strong>of</strong> blood, which are<br />

not possible to obtain from new born infants. We<br />

have developed a new system that is based on<br />

analysis <strong>of</strong> a single drop <strong>of</strong> capillary blood collected<br />

by a finger/heel prick on to a piece <strong>of</strong> filter paper<br />

and dried. In this project we will aim to develop and<br />

validate a method <strong>of</strong> assessing a range <strong>of</strong> nutrients<br />

that are important to good health in dried blood spot<br />

samples.<br />

This project will ideally suit an enthusiastic student<br />

who is interested in learning more about nutrition and<br />

the development <strong>of</strong> assays for detecting biomarkers<br />

for assessing nutrient status.<br />

Techniques/Skills Learnt: may include<br />

• Scientific writing & literature review<br />

• General laboratory techniques<br />

• Chemical extraction<br />

• Tandem mass spectrometry<br />

• High performance liquid chromatography<br />

Key References<br />

Jones DP, Park Y, Ziegler TR. Nutritional Metabolomics:<br />

Progress in Addressing Complexity in Diet and <strong>Health</strong>.<br />

Ann Rev Nutr. 2012 Apr 23. [Epub ahead <strong>of</strong> print]<br />

Skogstrand et.al. Effects <strong>of</strong> blood sample handling<br />

procedure on measurable inflammatory markers in<br />

plasma, serum, and dried blood spot samples. J Immunol<br />

Methods 336(2008) 78-84<br />

PROJECT: (Basic) Influence <strong>of</strong> maternal diet on the<br />

concentration <strong>of</strong> micronutrient elements in breast<br />

milk.<br />

Supervisors<br />

Dr Jo Zhou<br />

Pr<strong>of</strong> Robert Gibson<br />

Contact Person<br />

Pr<strong>of</strong> Robert Gibson<br />

robert.gibson@adelaide.edu.au.<br />

Please use the subject heading ‘<strong>Honours</strong> <strong>Project</strong>’.<br />

<strong>Project</strong> Background<br />

Micronutrients such as iron, zinc and selenium are<br />

important for many aspects <strong>of</strong> development, growth<br />

and metabolism. Breast milk is the sole source <strong>of</strong><br />

dietary micronutrients for exclusively breastfed<br />

infants. The aim <strong>of</strong> this study is to examine the<br />

influence <strong>of</strong> maternal diet during pregnancy and<br />

lactation on breast milk elemental micronutrient<br />

concentrations.<br />

Techniques/Skills Learnt<br />

• Scientific writing & literature review<br />

• Dietary intake assessment<br />

• General laboratory technique<br />

• Elemental analysis <strong>of</strong> breast milk with ICPMS<br />

Key References<br />

WHO (2001) Iron deficiency anaemia: assessment,<br />

prevention and control - A guide for programme<br />

managers.<br />

Siimes, et al. Breast milk iron--a declining<br />

concentration during the course <strong>of</strong> lactation. Acta<br />

Paediatr Scand 1979; 68:29<br />

PROJECT: (Basic) Iodine content <strong>of</strong> bread after<br />

mandatory iodine fortification.<br />

Supervisors<br />

Dr Jo Zhou<br />

Pr<strong>of</strong> Robert Gibson<br />

Contact Person<br />

Pr<strong>of</strong> Robert Gibson<br />

robert.gibson@adelaide.edu.au.<br />

Please use the subject heading ‘<strong>Honours</strong> <strong>Project</strong>’.<br />

<strong>Project</strong> Background<br />

Iodine is essential for thyroid hormone production,<br />

which in turn is important for normal growth and<br />

development. However, iodine deficiency has<br />

recently re-emerged in Australia. As a result, the<br />

government introduced mandatory iodine<br />

fortification by replacement <strong>of</strong> non-iodised salt with<br />

iodised salt in commercial bread making as a public<br />

health measure The aim <strong>of</strong> the study is to assess<br />

iodine content <strong>of</strong> different brands and types <strong>of</strong><br />

breads in the supermarket.<br />

Techniques/Skills Learnt<br />

• Scientific writing & literature review<br />

• General laboratory technique<br />

• Analysis <strong>of</strong> iodine content in foods using<br />

ICPMS<br />

Key References<br />

Food Standards Australia and New Zealand (2008)<br />

Proposal P1003 Mandatory iodine fortification for<br />

Australia assessment report.<br />

Australian Population <strong>Health</strong> Development Principal<br />

Committee (APHDPC) (2007) The Prevalence and<br />

Severity <strong>of</strong> Iodine Deficiency in Australia.<br />

50 |


PROJECT: (Basic) Measurement <strong>of</strong> vitamins A, D and E<br />

in dried blood spots.<br />

Supervisors<br />

Pr<strong>of</strong> Robert Gibson<br />

Dr Jo Zhou<br />

http://www.adelaide.edu.au/foodplus/<br />

Contact Person<br />

Pr<strong>of</strong> Robert Gibson<br />

robert.gibson@adelaide.edu.au.<br />

Please use the subject heading ‘<strong>Honours</strong> <strong>Project</strong>’.<br />

<strong>Project</strong> Background<br />

Assessing the nutritional status <strong>of</strong> humans and animals<br />

is <strong>of</strong> vital importance to evaluating overall health. In<br />

both animal and human trials, blood sample analysis<br />

is an integral part <strong>of</strong> assessing physiological and<br />

immune function in participants. This blood collection<br />

is invasive and analysis involves using volumes <strong>of</strong><br />

blood, which are not possible to obtain from some<br />

animals and new born infants. We have developed a<br />

new system that is based on analysis <strong>of</strong> a single drop<br />

<strong>of</strong> capillary blood collected by a finger/heel prick on<br />

to a piece <strong>of</strong> filter paper and dried. In this project we<br />

will aim to develop and validate a method <strong>of</strong><br />

assessing the fat soluble vitamins that are important<br />

to good health in dried blood spot samples.<br />

This project will ideally suit an enthusiastic student<br />

who is interested in learning more about nutrition and<br />

the development <strong>of</strong> assays for detecting biomarkers<br />

for assessing nutrient status.<br />

Techniques/Skills Learnt<br />

• Scientific writing and literature reviews<br />

• Basic laboratory techniques<br />

• Chemical extraction<br />

• Gas chromatography<br />

• High performance liquid chromatography<br />

Key References<br />

Santos J, Mendiola JA, Oliveira MB, Ibáñez E, Herrero M.<br />

Sequential determination <strong>of</strong> fat- and water-soluble<br />

vitamins in green leafy vegetables during storage. J<br />

Chromatogr A. 2012 May 3. [Epub ahead <strong>of</strong> print]<br />

Thomas JB, Sharpless KE, Yen JH, Rimmer CA.<br />

Determination <strong>of</strong> fat-soluble vitamins and carotenoids in<br />

standard reference material 3280<br />

multivitamin/multielement tablets by liquid<br />

chromatography with absorbance detection. J AOAC<br />

Int. 2011 May-Jun;94(3):815-22.<br />

Granado-Lorencio F, Herrero-Barbudo C, Blanco-Navarro<br />

I, Pérez-Sacristán B. Suitability <strong>of</strong> ultra-high performance<br />

liquid chromatography for the determination <strong>of</strong> fatsoluble<br />

nutritional status (vitamins A, E, D, and individual<br />

carotenoids). Anal Bioanal Chem. 2010 Jun;397(3):1389-<br />

93. Epub 2010 Apr 1.<br />

PROJECT: (Basic) Measurement <strong>of</strong> nutrients in breast<br />

milk.<br />

Supervisors<br />

Pr<strong>of</strong> Robert Gibson<br />

Dr Jo Zhou<br />

http://www.adelaide.edu.au/foodplus/<br />

Contact Person<br />

Pr<strong>of</strong> Robert Gibson<br />

robert.gibson@adelaide.edu.au.<br />

Please use the subject heading ‘<strong>Honours</strong> <strong>Project</strong>’.<br />

<strong>Project</strong> Background<br />

Assessing the nutritional status <strong>of</strong> humans and animals<br />

is <strong>of</strong> vital importance to evaluating overall health. In<br />

both animal and human studies, assessing the<br />

nutrient level in breast milk <strong>of</strong> dams/mothers is vital for<br />

an understanding <strong>of</strong> the nutrients that pups/babies<br />

receive. Currently there is no method to easily<br />

measure the nutrients in breast milk. We have<br />

developed a new system that is based on analysis <strong>of</strong><br />

a single drop <strong>of</strong> breast milk on to a piece <strong>of</strong> filter<br />

paper and dried. Sections <strong>of</strong> the dried milk spot can<br />

then be analysed by mass spectrometry. In this<br />

project we will aim to develop and validate a<br />

method <strong>of</strong> assessing a range <strong>of</strong> nutrients that are<br />

important to good health <strong>of</strong> infants.<br />

This project will ideally suit an enthusiastic student<br />

who is interested in learning more about nutrition and<br />

the development <strong>of</strong> assays for detecting biomarkers<br />

for assessing nutrient status.<br />

Techniques/Skills Learnt<br />

• Scientific writing & literature review<br />

• Dietary intake assessment<br />

• General laboratory techniques<br />

• Analysis <strong>of</strong> breast milk nutrient levels<br />

• Mass spectroscopy<br />

Key References<br />

Jones DP, Park Y, Ziegler TR. Nutritional Metabolomics:<br />

Progress in Addressing Complexity in Diet and <strong>Health</strong>.<br />

Ann Rev Nutr. 2012 Apr 23. [Epub ahead <strong>of</strong> print]<br />

Mäkelä J, Linderborg K, Niinikoski H, Yang B, Lagström H.<br />

Breast milk fatty acid composition differs between<br />

overweight and normal weight women: the STEPS Study.<br />

Eur J Nutr. 2012 May 26. [Epub ahead <strong>of</strong> print]<br />

Hoppu U, Isolauri E, Laakso P, Matomäki J, Laitinen K.<br />

Probiotics and dietary counselling targeting maternal<br />

dietary fat intake modifies breast milk fatty acids and<br />

cytokines. Eur J Nutr. 2012 Mar;51(2):211-9. Epub 2011<br />

May 31.<br />

Scholarships Available: Yes *<br />

* Scholarships are competitive and are awarded<br />

principally on academic merit.<br />

51 |


CNS Therapeutics Laboratory, Lysosomal Diseases Research Unit<br />

Supervisor/Contact Person<br />

Dr Kim Hemsley<br />

Phone: 8161 6740<br />

kim.hemsley@adelaide.edu.au<br />

Location: SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA<br />

The CNS Therapeutics Lab consists <strong>of</strong> 2 post-doctoral scientists, 2 scientists and 5 technical <strong>of</strong>ficers and 2 Hons<br />

students and is part <strong>of</strong> the Lysosomal Diseases Research Unit (LDRU). The LDRU’s research goal is ‘early diagnosis and<br />

treatment for all lysosomal storage disorders (LSD)’. A large, well-established and multidisciplinary team, the LDRU<br />

has been extremely successful at both basic science and translating this to successful clinical and commercial<br />

outcomes.<br />

LSD are inborn errors <strong>of</strong> metabolism in which there is a mutation in a gene encoding a lysosomal enzyme, resulting in<br />

low or no production <strong>of</strong> that lysosomal enzyme and subsequent accumulation <strong>of</strong> substrate within cells. Progressive<br />

neurodegeneration is seen in the majority <strong>of</strong> LSD and most <strong>of</strong> these disorders have no treatment at present, so<br />

affected children <strong>of</strong>ten die in their early teens.<br />

The CNS Therapeutics Lab primarily utilises a mouse model <strong>of</strong> a neurodegenerative LSD, known as MPS IIIA (1, 2).<br />

Other disease models are also being established. MPS IIIA is presently untreatable and so we are examining the<br />

cascade <strong>of</strong> neuropathological changes that occur within the MPS IIIA brain as a result <strong>of</strong> the accumulation <strong>of</strong><br />

substrate (3, 4). Gaining a better understanding <strong>of</strong> how the disorder manifests will permit optimisation <strong>of</strong> therapies in<br />

pre-clinical testing and potentially allow us to develop novel therapeutic approaches. Pre-clinical testing is being<br />

undertaken on the mice including, but not limited to, gene therapy and enzyme replacement therapy (5-8).<br />

<strong>Project</strong>: (Basic) Understanding and treating<br />

neuropathology in murine models <strong>of</strong> neurodegenerative<br />

lysosomal storage disorders.<br />

.R.<br />

<strong>Project</strong> Background<br />

The <strong>Honours</strong> projects on <strong>of</strong>fer in the lab will examine<br />

neuropathogenesis, and in some cases, treatment <strong>of</strong><br />

it, using small animal models <strong>of</strong> lysosomal storage<br />

disorders that primarily affect the brain. Treatments<br />

currently being examined in the lab include enzyme<br />

replacement, gene therapy and a variety <strong>of</strong> agents<br />

designed to act as co-therapies. Students involved in<br />

these studies will gain experience in a variety <strong>of</strong><br />

techniques which may include<br />

immunohistochemical, histochemical, molecular and<br />

biochemical assays, small animal handling, surgical<br />

techniques and behavioural assessments.<br />

References<br />

Bhaumik M, Muller VJ, Rozaklis T, Johnson L, Dobrenis<br />

K, Bhattacharyya R, Wurzelmann S, Finamore P,<br />

Hopwood JJ, Walkley SU, Stanley P. A mouse model<br />

for mucopolysaccharidosis type III A (Sanfilippo<br />

syndrome). Glycobiology. 1999 Dec;9(12):1389-96.<br />

Crawley AC, Gliddon BL, Auclair D, Brodie SL, Hirte C,<br />

King BM, Fuller M, Hemsley KM, Hopwood JJ.<br />

Characterization <strong>of</strong> a C57BL/6 congenic mouse strain<br />

<strong>of</strong> mucopolysaccharidosis type IIIA. Brain Res. 2006<br />

Aug 9;1104(1):1-17<br />

Savas PS, Hemsley KM, Hopwood JJ. Intracerebral<br />

injection <strong>of</strong> sulfamidase delays neuropathology in<br />

murine MPS-IIIA. Mol Genet Metab. 2004<br />

Aug;82(4):273-85.<br />

Hemsley KM, Beard H, King BM, Hopwood JJ. Effect <strong>of</strong><br />

high dose, repeated intra-CSF injection <strong>of</strong><br />

sulphamidase on neuropathology in MPS IIIA mice.<br />

Genes Brain Behav. 2008<br />

Lau AA, Hemsley KM, Meedeniya A, Hopwood JJ. In<br />

vitro characterization <strong>of</strong> genetically modified<br />

embryonic stem cells as a therapy for murine<br />

mucopolysaccharidosis type IIIA. Mol Genet Metab.<br />

2004 Feb;81(2):86-95.<br />

Fraldi A, Hemsley K, Crawley A, Lombardi A, Lau A,<br />

Sutherland L, Auricchio A, Ballabio A, Hopwood JJ.<br />

Functional correction <strong>of</strong> CNS lesions in an MPS-IIIA<br />

mouse model by intracerebral AAV-mediated<br />

delivery <strong>of</strong> sulfamidase and SUMF1 genes. Hum Mol<br />

Genet. 2007 Nov 15;16(22):2693-702.<br />

Hemsley KM, King B, Hopwood JJ. Injection <strong>of</strong><br />

recombinant human sulfamidase into the CSF via the<br />

cerebellomedullary cistern in MPS IIIA mice. Mol<br />

Genet Metab. 2007 Mar;90(3):313-28<br />

Hemsley KM, Luck AJ, Crawley AC, Hassiotis S, Beard<br />

H, King B, Rozek T, Rozaklis T, Fuller M, Hopwood JJ.<br />

Examination <strong>of</strong> intravenous and intra-CSF protein<br />

delivery for treatment <strong>of</strong> neurological disease. Eur J<br />

Neurosci. 2009 Mar;29(6):1197-214.<br />

52 |


Crani<strong>of</strong>acial Research Group<br />

Contact Person<br />

Associate Pr<strong>of</strong>essor Barry Powell<br />

Phone: 8161 7652<br />

barry.powell@adelaide.edu.au<br />

Supervisors<br />

Associate Pr<strong>of</strong>essor Barry Powell<br />

Associate Pr<strong>of</strong>essor Peter Anderson<br />

Locations: Women’s and Children’s Hospital, Women’s and Children’s <strong>Health</strong><br />

Research Institute<br />

Our research involves the study <strong>of</strong> genes involved in bone growth and craniosynostosis, a developmental disorder<br />

that affects skull growth and mental development in children (Anderson et al., 2006; Coussens et al., 2007; 2008). Our<br />

overarching goal is to understand gene actions in skull growth and to use that knowledge to develop novel<br />

therapeutics to improve treatment <strong>of</strong> bone disorders in collaboration with clinical colleagues. In the projects<br />

outlined below you will join a team working at the cutting edge <strong>of</strong> research, using sophisticated molecular and<br />

cellular approaches, cell culture and gene knockout animal models to study gene function. Your research could<br />

impact on the development <strong>of</strong> adjunctive treatments in the clinical management <strong>of</strong> craniosynostosis and other<br />

bone growth disorders and provide new insight into the functions <strong>of</strong> genes and how bone growth is regulated.<br />

PROJECT: (Basic) Application <strong>of</strong> recombinant proteins<br />

to modify abnormal bone growth.<br />

<strong>Project</strong> Background<br />

We are studying a genetic crani<strong>of</strong>acial abnormality<br />

(known as craniosynostosis) that affects skull growth<br />

by causing premature fusion <strong>of</strong> the skull plates. The<br />

only treatment is transcranial surgery, but where bony<br />

fusion re-occurs surgery is inadequate and novel<br />

therapies are needed. We have discovered that the<br />

proteoglycans, glypican 1 and glypican 3 (GPC1 and<br />

GPC3) regulate activity <strong>of</strong> the key bone growth<br />

factor, bone morphogenetic protein 2 (BMP2) in<br />

cranial cells.<br />

In this project we will mouse models to test if<br />

manipulation <strong>of</strong> BMP2 activity can prevent the reoccurrence<br />

<strong>of</strong> bone growth after surgery. This could<br />

be tested in tissue explants or in vivo. The effects on<br />

bone fusion, BMP2 activity and markers <strong>of</strong> bone<br />

growth can be assessed.<br />

Outcomes<br />

This research could lead to the development <strong>of</strong> novel<br />

therapeutic approaches to treat abnormal bone<br />

growth.<br />

PROJECT: (Basic) How do glypicans and bone<br />

morphogenetic proteins interact in cranial bone<br />

growth?<br />

<strong>Project</strong> Background<br />

The proteoglycans, GPC1 and GPC3, act as coreceptors<br />

for growth factors at the cell surface but<br />

they can also be released extracellulary to bind<br />

them. In this project you will investigate how GPC1<br />

and GPC3 regulate BMP2 in cranial suture cells.<br />

Building on data that GPC1 and GPC3 interact with<br />

BMP2 in vitro, this will be tested in vivo in this project.<br />

We can explore if interaction occurs at the cell<br />

surface or after the proteins have been released from<br />

the cell. The fate <strong>of</strong> GPC-BMP2 complexes can be<br />

explored. Are they internalised and degraded, or<br />

recycled? Regions responsible for interaction could<br />

be mapped using mutant proteins.<br />

Outcomes<br />

This research will provide insight into how glypicans<br />

and BMPs interact and can therefore have impact in<br />

the broader fields <strong>of</strong> bone research and<br />

proteoglycan biology.<br />

PROJECT: (Basic) What does retinol binding protein do<br />

in skull growth?<br />

<strong>Project</strong> Background<br />

Retinol binding protein (RBP) is well known as a<br />

transport protein for retinol in serum and has recently<br />

been implicated in metabolic syndrome. However,<br />

our research shows that it may have other<br />

unexpected functions. We have discovered that RBP<br />

is expressed by skull bone-forming cells and is<br />

dramatically down-regulated in the congenital<br />

condition <strong>of</strong> craniosynostosis (Coussens et al., 2007)<br />

suggesting that it has a role in skull bone formation.<br />

Our recent data analysing RBP and its receptor<br />

reveal remarkable subcellular locations and<br />

movement within cells.<br />

In this project we aim to determine what RBP and its<br />

receptor do in skull bone-forming cells.<br />

Outcomes<br />

This research will provide insight into the function <strong>of</strong><br />

RBP and its receptor in skull bone growth.<br />

53 |


Techniques that will be used in the projects include<br />

human cell culture, animal studies, micro-computed<br />

tomography, gene transfection, real-time RT-PCR,<br />

luciferase assay, mineralization assay, proliferation<br />

assay, western blot, immunohistochemistry and<br />

confocal microscopy.<br />

References<br />

Anderson PJ,<br />

DJ Netherway, P Self and DJ David.<br />

Scanning electron microscope and Micro CT<br />

evaluation <strong>of</strong> cranial sutures in health and disease.<br />

Journal <strong>of</strong> Crani<strong>of</strong>acial Surgery 17, 909-919, 2006.<br />

Coussens AK, CR Wilkinson, IP Hughes, CP Morris, A<br />

van Daal, PJ Anderson and BC Powell.<br />

2007.<br />

Unravelling the molecular control <strong>of</strong> calvarial suture<br />

morphogenesis in children with craniosynostosis. BMC<br />

Genomics. 8:doi: 10.1186/1471-2164-8-458.<br />

Coussens AK, CR Wilkinson, IP Hughes, BC Powell, PJ<br />

Anderson and A van Daal. 2008. Identification <strong>of</strong><br />

genes differentially expressed by prematurely fused<br />

human sutures using a novel in vivo-in vitro approach.<br />

Differentiation. 76:531-545.<br />

Donato R, SA Wood, I Saunders, B Gundsambuu, KY<br />

Mak, CA Abbott and BC Powell.<br />

2009. Regulation <strong>of</strong><br />

epithelial apical junctions and barrier function by<br />

Galpha13, Biochim Biophys Acta - Molecular Cell<br />

Research 1793, 1228-1235.<br />

These projects are eligible for the WCHRI <strong>Honours</strong><br />

Scholarship <strong>of</strong> $2,000. See the website for further<br />

details and how to apply<br />

http://www.wchri.com.au/Scholarships.htm.<br />

54 |


Lysosomal Biology, Lysosomal Diseases Research Unit<br />

Supervisor/Contact Person<br />

Dr Maria Fuller<br />

Phone: 8161 6741<br />

maria.fuller@adelaide.edu.au<br />

Location: SA Pathology, Women’s and Children’s Hospital<br />

The Lysosomal Diseases Research Unit is highly regarded by its peers internationally for its seminal scientific<br />

contributions to the area <strong>of</strong> lysosomal research. Its research has resulted in FDA approval <strong>of</strong> first-ever treatments for<br />

two lysosomal storage disorders. The Unit consists <strong>of</strong> about 35 research staff and students working on a range <strong>of</strong><br />

projects directed at understanding, improving diagnostic capabilities and treatment for a group <strong>of</strong> inherited<br />

metabolic diseases, known as lysosomal storage disorders. The Lysosomal Biology section <strong>of</strong> the Unit is currently<br />

investigating the role <strong>of</strong> the lysosome with respect to lipid metabolism in conditions outside the spectrum <strong>of</strong> classical<br />

lysosomal storage disorders, such as diabetes, metabolic syndrome, obesity, and Alzheimer’s and Parkinson’s<br />

diseases. We utilise state-<strong>of</strong>-the-art technology, including electrospray ionisation-tandem mass spectrometry, and<br />

lipid analytical techniques developed in our group. This technology provides a powerful platform to the<br />

investigation <strong>of</strong> metabolic disease. In February this year the group acquired a Waters Synapt High Definition mass<br />

spectrometer – the first one <strong>of</strong> its kind in Australia – capable <strong>of</strong> metabolomics, proteomics and MALDI imaging. This<br />

instrument is unique in <strong>of</strong>fering an additional dimension to the analysis <strong>of</strong> compounds by using ion mobility<br />

separation, which allows compounds to be differentiated by size and shape.<br />

The following projects will give students broad scientific skill in cell models <strong>of</strong> disease, cell biology and the latest mass<br />

spectrometry technology in a highly challenging and stimulating academic environment. The student will also have<br />

ample support from other staff within the group who are experts in the methodology used (pictured above).<br />

Students will have the opportunity to participate in regular scientific discussions and meetings within the Unit.<br />

PROJECT: (Basic) Lipid disturbances in Neimann Pick<br />

C disease.<br />

<strong>Project</strong> Background<br />

Niemann-Pick C (NPC) disease is a hereditary<br />

cholesterol storage disorder resulting in severe<br />

neurovisceral pathology for which there is no cure.<br />

Many <strong>of</strong> the known risk factors <strong>of</strong> other diseases such<br />

as Alzheimer’s disease are also associated with<br />

cholesterol metabolism. This project proposes a<br />

“lipidomics” approach to identify the type <strong>of</strong> lipid<br />

alterations that occur secondarily to impaired<br />

cholesterol homeostasis using NPC patient cultured<br />

skin fibroblasts as a cell model. Sub-cellular<br />

fractionation techniques will also be employed to<br />

determine the location <strong>of</strong> these lipid alterations within<br />

the cell. The second part <strong>of</strong> this project will evaluate<br />

the possibility <strong>of</strong> normalising these lipid alterations by<br />

supplementing the culture media with particular fatty<br />

acids and drugs <strong>of</strong> lipid metabolism, as a potential<br />

therapeutic strategy for diseases <strong>of</strong> cholesterol<br />

metabolism.<br />

The overall scientific objective <strong>of</strong> this project is to<br />

identify lipid alterations that may be treated with diet<br />

and/or conventional drugs, providing a treatment for<br />

many individuals with diseases <strong>of</strong> cholesterol<br />

metabolism, including Alzheimer’s and NPC. The<br />

student will gain experience in cell culture and a<br />

variety <strong>of</strong> cell biology techniques including sub-<br />

cellular fractionation, as well as learning aspects <strong>of</strong><br />

mass spectrometry.<br />

PROJECT: (Basic) Sphingolipids in diabetes and<br />

Gaucher disease.<br />

<strong>Project</strong> Background<br />

The membranes <strong>of</strong> mammalian cells are composed<br />

<strong>of</strong> an ordered array <strong>of</strong> lipids, and in many instances<br />

chemical or functional changes in these membranes<br />

are central to the pathogenesis <strong>of</strong> disease. We have<br />

shown that membrane lipids are altered in lysosomal<br />

storage disorders, and recent studies in the<br />

glycosphingolipid storage disorder, Gaucher disease,<br />

associate such lipid alterations with insulin resistance.<br />

It is now obvious that lysosomal storage disorders and<br />

insulin resistance diabetes share disturbances in<br />

sphingolipid metabolism, and because sphingolipids<br />

predominate in membrane rafts, the mechanistic link<br />

may be mediated by rafts. Rafts are specialised<br />

membrane microdomains that facilitate interactions<br />

between the protein and lipid components <strong>of</strong><br />

signalling pathways. This project proposes to<br />

determine the lipid composition <strong>of</strong> membrane rafts in<br />

cell models <strong>of</strong> Gaucher disease and insulin resistant<br />

diabetes to unravel common nodes <strong>of</strong> interactions<br />

between these two pathogenic mechanisms. For<br />

effective long term-treatment <strong>of</strong> these conditions,<br />

understanding the pathogenic mechanisms will be<br />

imperative.<br />

55 |


The student will gain broad experience in cell culture<br />

with both primary and gene knockdown<br />

immortalised cell lines. The student will gain<br />

experience in Western blot analysis, membrane<br />

microdomain isolation and various aspects <strong>of</strong> mass<br />

spectrometry.<br />

Publications<br />

Hein LK, Meikle PJ, Hopwood JJ, Fuller M. (2007)<br />

Secondary sphingolipid accumulation in a cell model<br />

<strong>of</strong> Gaucher disease. Mol Genet Metab 92, 336-45.<br />

Meikle PJ, Whitfield PD, Rozaklis T, Blacklock D,<br />

Duplock S, Elstein D, Zimran A, Mengel E, Cannell P,<br />

Hopwood JJ, Fuller M. (2008) Plasma lipids are altered<br />

in Gaucher disease: Biochemical markers to evaluate<br />

therapeutic intervention. Blood Cells Mol Dis 40, 420-<br />

7.<br />

Meikle PJ, Duplock S, Blacklock D, Whitfield PD,<br />

Macintosh GL, Hopwood JJ, Fuller M. (2008) Effect <strong>of</strong><br />

lysosomal storage on bis(monoacylglycero)<br />

phosphate. Biochem J 411, 71-8.<br />

Fuller M, Rozaklis T, Lovejoy M, Zarrinkalam K,<br />

Hopwood JJ, Meikle PJ. (2008) Glucosylceramide<br />

accumulation is not confined to the lysosome in<br />

fibroblasts from patients with Gaucher disease. Mol<br />

Genet Metab 93, 437-43.<br />

Hein LK, Duplock S, Hopwood JJ, Fuller M. (2008) Lipid<br />

composition <strong>of</strong> microdomains is altered in a cell<br />

model <strong>of</strong> Gaucher disease. J Lipid Res 49, 1725-34.<br />

56 |


Matrix Biology Unit<br />

Location: Women’s and Children’s Hospital<br />

Supervisor/Contact Persons<br />

Dr Sharon Byers<br />

Phone: 8161 7093<br />

sharon.byers@adelaide.edu.au<br />

Dr Ainslie Derrick-Roberts<br />

Phone: 8161 6373<br />

ainslie.derrickroberts@adelaide.edu.au<br />

Our research interest lies in the regulation <strong>of</strong> a group <strong>of</strong> complex carbohydrates called glycosaminoglycans (gags).<br />

Gags are found in all tissues where they define tissue structure and modify tissue growth through the regulation <strong>of</strong><br />

growth factor activity. They are constantly synthesised and degraded as part <strong>of</strong> normal development. Defects in<br />

degradation lead to the mucopolysaccharidosis (MPS) group <strong>of</strong> inherited metabolic disorders. Our laboratory is<br />

focused on the characterisation <strong>of</strong> disease progression and the development <strong>of</strong> safe and effective therapies for<br />

MPS children and played a key role in the commercialization <strong>of</strong> enzyme replacement therapy. The laboratory is<br />

situated in the department <strong>of</strong> Genetics and Molecular Pathology, SA Pathology (WCH site). The department is the<br />

Australasian centre for the diagnosis and treatment <strong>of</strong> children with MPS.<br />

The multiple organs systems affected by the accumulation <strong>of</strong> undegraded gag in MPS presents a challenge to the<br />

clinical management <strong>of</strong> affected children. While somatic tissues such as liver and kidney are amenable to<br />

replacement therapies, the brain and the cartilage component <strong>of</strong> the musculoskeletal system are untreatable.<br />

Students will join ongoing projects in one <strong>of</strong> the following areas:-<br />

Key Words:<br />

genetic disease, childhood disorders, therapy, murine models <strong>of</strong> disease, behavioural tests, bone growth<br />

PROJECT: (Basic) Targeting CNS pathology in MPS<br />

disorders.<br />

<strong>Project</strong> Background<br />

CNS pathology that manifests as a severe,<br />

progressive loss <strong>of</strong> cognitive function is a major<br />

symptom <strong>of</strong> 7 <strong>of</strong> the 11 MPS types and is currently<br />

untreatable. The aim <strong>of</strong> this project is to find a<br />

minimally invasive approach to treat CNS pathology<br />

in MPS disorders. Gene therapy replacement <strong>of</strong> the<br />

missing gene has shown promising results in mouse<br />

models (Anson et al., 2011), however has a limited<br />

effect on the CNS due to the presence <strong>of</strong> the bloodbrain<br />

barrier. The aim <strong>of</strong> this project is to improve<br />

blood brain barrier transport using a modified<br />

enzyme to enhance receptor mediated transport<br />

across the blood-brain barrier. By combining gene<br />

therapy to increase residual enzyme levels with small<br />

molecule SDT (Derrick-Roberts et al., 2012) to<br />

decrease gag levels is another approach to improve<br />

the outcome <strong>of</strong> CNS disease in MPS disease also<br />

under investigation.<br />

Students undertaking this project will learn<br />

fundamental research techniques applicable to a<br />

wide range <strong>of</strong> research areas. These include small<br />

animal handling, behavioural testing (eg water maze<br />

tests), enzyme assay, electrophoresis and<br />

carbohydrate biochemistry. Equipment to be used<br />

will include: microscopes, centrifuges, electrophoresis<br />

equipment, fluorimeter, mass spectrometer and<br />

laminar flow/biohazard hoods.<br />

References<br />

Roberts ALK, Rees MH, Klebe S, Fletcher JM and Byers<br />

S. (2007): Improvement in behaviour after substrate<br />

deprivation therapy with rhodamine B in a mouse<br />

model <strong>of</strong> MPS IIIA. Molecular Genetics and<br />

Metabolism, 92: 115-121.<br />

Anson DS, McIntyre C, Byers S. (2011): Therapies for<br />

neurological disease in the mucopolysaccharidoses.<br />

Curr Gene Ther. 11: 132-43.<br />

Derrick-Roberts ALK, Ly M, Marais W and Byers S.<br />

(2012). Rhodamine B and 2-acetamido-1,3,6-tri-Oacetyl-4-deoxy-4-fluoro-D-glucopyranose<br />

(F-GlcNAc)<br />

inhibit chondroitin/dermatan and keratan sulphate<br />

synthesis by different mechanisms in bovine<br />

chondrocytes. Mol. Genet. Metab. 106: 214-20.<br />

PROJECT: (Basic) Understanding skeletal disease in<br />

MPS patients.<br />

<strong>Project</strong> Background<br />

Direct injection <strong>of</strong> enzyme into the joint space<br />

increases the localized concentration <strong>of</strong> enzyme<br />

replacement therapy (ERT) and improves joint<br />

outcome (Auclair et al 2006, 2007). We have recently<br />

shown that the same improvement can be achieved<br />

57 |


y a gene therapy approach to transduce joint cells<br />

(Byers et al 2009). In this project students will join an<br />

ongoing project to investigate the efficacy <strong>of</strong><br />

systemic gene therapy alone or in combination with<br />

intra-articular gene therapy to prevent the<br />

development <strong>of</strong> both bone and joint pathology.<br />

Short stature is a feature <strong>of</strong> 6 <strong>of</strong> the 11 MPS disorders<br />

which leads to problems with mobility. Therapy<br />

options are limited and ERT available for some MPS<br />

disorders does not increase bone length in affected<br />

children even though it has a positive effect on other<br />

symptoms. This suggests that we need a better<br />

understanding bone growth and by isolating distinct<br />

regions <strong>of</strong> the growth plate responsible for bone<br />

lengthening we can identify key markers involved in<br />

long bone growth which can be new targets to<br />

improve short stature.<br />

Students undertaking this project will learn<br />

fundamental research techniques applicable to a<br />

wide range <strong>of</strong> research areas. These include small<br />

animal handling, bone histomorphometric<br />

techniques, micro CT, laser capture microdissection,<br />

real-time PCR, electrophoresis and carbohydrate<br />

biochemistry. Equipment to be used will include:<br />

microscopes, image analysis equipment, centrifuges,<br />

electrophoresis equipment, fluorimeter and laminar<br />

flow/biohazard hoods.<br />

References<br />

Macsai CE, Derrick-Roberts AL, et al. (2012): Skeletal<br />

response to lentiviral mediated gene therapy in a<br />

mouse model <strong>of</strong> MPS VII. Mol Genet Metab. 2012<br />

Jun;106: 202-13<br />

Byers S, Rothe M, et al. (2009). Lentiviral mediated<br />

correction <strong>of</strong> MPS VI cells and gene transfer to<br />

skeletal tissues. Molecular Genetics and Metabolism,<br />

97: 102-108.<br />

Auclair D, Hein LK, et al. (2006): Intra-articular enzyme<br />

administration for joint disease in feline<br />

mucopolysaccharidosis VI: Enzyme dose and interval.<br />

Pediatric Research, 59: 538-542.<br />

Auclair D, Hopwood JJ, et al. (2007): Intra-articular<br />

enzyme administration for joint disease in feline<br />

mucopolysaccharidosis VI: Long-term therapy.<br />

Molecular Genetics and Metabolism, 91: 352-61.<br />

58 |


Molecular Immunology<br />

Cheryl Brown<br />

Phone: 8161 6729<br />

cheryl.brown@adelaide.edu.au<br />

Supervisors/ Contact Persons<br />

Associate Pr<strong>of</strong>essor Simon Barry<br />

Phone: 8161 6562<br />

simon.barry@adelaide.edu.au<br />

Tim Sadlon<br />

Phone: 8161 6729<br />

timothy.sadlon@adelaide.edu.au<br />

Location: Women’s and Children’s Hospital<br />

My lab is interested in how a healthy immune system balances being ready to react by swiftly fighting <strong>of</strong>f<br />

pathogens, while maintaining tolerance to harmless challenges such as food and body tissues. The cellular immune<br />

repertoire in humans is broad, but we are focussed on a T cell subset that is shaped along with the immune system<br />

from birth. These cells are known as regulatory T cells, and they are accepted as the policemen <strong>of</strong> the immune<br />

system. There is increasing evidence that in a wide number <strong>of</strong> disease states including autoimmune diseases such as<br />

Type 1 diabetes and Multiple Sclerosis, these cells fail to regulate the immune system, and allow inappropriate<br />

destruction <strong>of</strong> tissues that are essential for life. In order to understand how this breaks down in disease one must first<br />

understand what is the basis <strong>of</strong> a healthy Treg. To do this we are focussed on human cells and we use a number <strong>of</strong><br />

state <strong>of</strong> the art gene discovery tools such as microarrays to identify and then confirm the key genes in Treg function.<br />

As Treg play a role in autoimmune disease, cancer and transplantation tolerance, our research findings have a wide<br />

clinical application. The projects below are from parts <strong>of</strong> this overall research program. We currently have 11<br />

members <strong>of</strong> the research group, which provides lab and academic support for students.<br />

PROJECT: (Basic or Clinical/basic) Molecular<br />

identification <strong>of</strong> Regulatory T cells.<br />

.R.<br />

<strong>Project</strong> Background<br />

The recent identification <strong>of</strong> regulatory T cells (Tregs)<br />

as a key mediator <strong>of</strong> central and peripheral<br />

tolerance has led to an increase in our<br />

understanding <strong>of</strong> the cellular mechanisms by which<br />

humans maintains the healthy state. This has been<br />

confirmed by the identification <strong>of</strong> a transcription<br />

factor named FOXP3 in both mouse and human<br />

Tregs, which is proven to be essential for formation<br />

and function <strong>of</strong> the committed T cell subset that has<br />

regulatory capacity. There is however, very little<br />

known about the molecular basis <strong>of</strong> this process. This<br />

project aims to identify the genes directly regulated<br />

by FOXP3 and to determine their role in the<br />

regulatory phenotype. We have used a number <strong>of</strong><br />

direct and indirect molecular approaches such as<br />

Chromatin Immunoprecipitation and microarray<br />

analysis to pr<strong>of</strong>ile genes regulated by FOXP3 (Fig 1),<br />

and we will validate their role in regulatory function<br />

by direct assays and by over expression or gene<br />

ablation studies.<br />

Fig 1: Comparative microarray analysis <strong>of</strong> CD4<br />

CD25+ natural Treg to identify the key genes<br />

required for function.<br />

59 |


The candidate genes identified in this approach may<br />

lead to therapeutic approaches for intervention in<br />

the function <strong>of</strong> regulatory cells, and will also have<br />

application for diagnostic analysis <strong>of</strong> regulatory cell<br />

function. Analysis <strong>of</strong> these and other Tcells from<br />

disease samples eg IBD (Fig 2) reveals the clinical<br />

defect in numbers <strong>of</strong> Treg.<br />

Fig 2: Comparative analysis <strong>of</strong> CD4 CD25+ natural Treg<br />

and Th17 in IBD patients shows a defect in the ratio <strong>of</strong><br />

these cells.<br />

Techniques/skills/approach<br />

Molecular biology, RT PCR, microarray data analysis,<br />

tissue culture lentiviral production, gene silencing and<br />

over expression, functional assay.<br />

References<br />

1. Bresatz S Sadlon T, Millard D, Zola H, Barry SC.<br />

Purification and characterization <strong>of</strong> CD4+ CD25+<br />

regulatory cells from Cord and peripheral blood. J<br />

Immunuol Methods 2007;327:53-62 [Epub ahead <strong>of</strong><br />

print]. Impact Factor = 2.5, Journal rank in field 46/114<br />

2. Zola H, Swart B, Banham A, Barry SC, Beare A, Bensussan<br />

A, Boumsell L, D Buckley C, Buhring HJ, Clark G, Engel P,<br />

Fox D, Jin BQ, Macardle PJ, Malavasi F, Mason D,<br />

Stockinger H, Yang X. CD molecules 2006--human cell<br />

differentiation molecules. J Immunol Methods<br />

2007;319(1-2):1-5. Epub 2006 Dec 4. Impact Factor = 2.5,<br />

Journal rank in field 46/114<br />

PROJECT: (Basic) Cord Blood Stem cell differentiation<br />

in to regulatory T cells.<br />

.R.<br />

<strong>Project</strong> Background<br />

The clinical application <strong>of</strong> regulatory T cells is<br />

significantly hampered by the limited cell numbers<br />

that can be obtained from either cord or adult<br />

blood. Attempts to expand these purified Treg ex vivo<br />

have shown some promise, but there is some<br />

evidence that after extended culture ex vivo these<br />

cells lose their suppressive capacity. An alternative<br />

approach is to generate large numbers <strong>of</strong> T cells de<br />

novo from stem cells since these cells have the<br />

capacity to differentiate into all cells <strong>of</strong> the<br />

haemopoietic system. We have established an ex<br />

vivo differentiation assay that can expand cord<br />

blood stem cells and induce their differentiation<br />

along the lymphoid pathway using a co-culture<br />

system giving notch signals via the Notch ligand<br />

Delta like 1 ( Fig 3). In this system we robustly observe<br />

5-600 fold expansion <strong>of</strong> cell numbers and the<br />

generation <strong>of</strong> T cell subsets as defined by CD4/CD8<br />

staining.<br />

Fig 3. Cord blood stem cells on feeder cells<br />

showing formation <strong>of</strong> CD4 CD25+ subsets with<br />

similar characteristics to natural Treg<br />

Techniques/skills/approach<br />

Molecular biology, RT PCR, tissue culture, lentiviral<br />

production, gene silencing and over expression,<br />

functional assay with cord blood stem cells.<br />

References<br />

1. Hutton JF, Gargett T, Sadlon TJ, Bresatz S, Brown CY, Zola<br />

H, Shannon MF, D'Andrea RJ, Barry SC: Development <strong>of</strong><br />

CD4 + CD25 + FOXP3 + Regulatory T cells from Cord Blood<br />

Haemopoietic Progenitor Cells. Journal <strong>of</strong> Leukocyte<br />

Biology in press ePub Dec 22 08. Impact Factor = 4.3,<br />

Journal rank in field 50/156<br />

PROJECT: (Basic) Lentiviral vectors for gene delivery<br />

and gene ablation.<br />

.R.<br />

<strong>Project</strong> Background<br />

Manipulation <strong>of</strong> primary cells has a key limitation in<br />

that these cells are refractory to standard<br />

transfection protocols. Also, since they are <strong>of</strong>ten <strong>of</strong><br />

low mitotic index, they are only infected at low<br />

efficiency by murine retroviruses (RV), as these viruses<br />

require cell division for integration. The recent<br />

development <strong>of</strong> HIV1 based lentivectors (LV)<br />

provides an attractive option for gene delivery into T<br />

cell and stem cell populations, as these viruses carry<br />

the necessary cis elements to facilitate nuclear<br />

transport and integration in the absence <strong>of</strong> cell<br />

division. We have developed a suite <strong>of</strong> lentiviral<br />

vectors for stable gene delivery into primary cells<br />

both for gene therapy and gene discovery<br />

applications, and more recently for gene ablation<br />

using RNA interference (Fig 4). This technology relies<br />

on the expression <strong>of</strong> a short hairpin RNA structure that<br />

is complimentary to the gene target, and that is<br />

processed by cellular machinery to generate an RNA<br />

oligo that can bind to and target the mRNA for<br />

destruction.<br />

60 |


Fig 4. Lentiviral delivery <strong>of</strong> shRNAi showing abalation <strong>of</strong><br />

FoxP3 expression that is inducible with doxicyclin and is<br />

reversible.<br />

Techniques/skills/approach<br />

Molecular biology, RT PCR, Tissue culture, lentiviral<br />

production, gene silencing and over expression,<br />

functional assay, T cell assays.<br />

References<br />

1. Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A,<br />

Farshid G, Vadas MA, Khew-Goodall Y and Goodall GJ:<br />

The microRNA-200 family and miR-205 regulate<br />

epithelial-mesenchymal transition by targeting the Ecadherin<br />

repressors, ZEB1 and SIP1. Nature Cell Biology<br />

2008 10 (5)593-601 Impact Factor F = 18.1, [28] J Journal<br />

rank in field 5/156<br />

2. Barry SC,Coates PT. Retroviral escape using DC - that's<br />

what Friends are for. Blood 2007 110(12)3819-3820<br />

Impact Factor = 10.1, Journal rank in field 2/59<br />

3. Drabsch Y, Hugo H, Zhang R, Dowhan DH, Miao YR,<br />

Gewirtz AM, Barry, SC, Ramsay RG and Gonda TJ.<br />

Mechanism <strong>of</strong> and Requirement for Estrogen-Regulated<br />

MYB Expression in Estrogen Receptor-Positive Breast<br />

Cancer Cells. PNAS 2007;104(34):13762-7. Impact Factor<br />

= 10.9, Journal rank in field 3/45<br />

4. Brzezinski M, Yanay O, Waldron L, Barry SC and Osborne<br />

WRA. G-CSF-lentivirus provided long-term elevated<br />

neutrophil counts in rats and sequential EPO-lentivirus<br />

administration produced increased hematocrits in both<br />

lentivirus treated and naïve rats. J Gene Medicine<br />

2007;9(7):571-8. Impact Factor = 4.8, Journal rank in<br />

field 8/75<br />

5. Barry SC Brezinzki M, Yanay O, Seppen, J Osborne WRA.<br />

Sustained elevation <strong>of</strong> neutrophils in rats induced by<br />

lentivirus-mediated G-CSF delivery. J Gene medicine<br />

2005;7(12):1510-6. Impact Factor = 4.8, Journal rank in<br />

field 8/75<br />

PROJECT: (Basic/clinical basic) Is there a role for<br />

FOXP3 in Breast Cancer?<br />

.R.<br />

<strong>Project</strong> Background<br />

There is very recent evidence that FOXP3 may also<br />

play a role in breast cancer as a new tumour<br />

suppressor. This means that its expression prevents the<br />

transformation leading to cancer, and in cells that<br />

have transformed it is no longer correctly expressed.<br />

As we have discovered a great many <strong>of</strong> the genes<br />

that are directly regulated by FOXP3 in Tregs, it is likely<br />

that some <strong>of</strong> these are also required for prevention <strong>of</strong><br />

breast cancer. We have now confirmed that FOXP3 is<br />

expressed in healthy breast epithelial cells, and are<br />

testing a number <strong>of</strong> genes for their role in either the<br />

loss <strong>of</strong> normal FOXP3 expression, or the progression to<br />

cancer. This includes a number <strong>of</strong> micro RNAs that<br />

are controlled by FOXP3 (quantitated by RT PCR in<br />

Fig 5), one <strong>of</strong> which (mir21) has been confirmed to<br />

play a role on breast cancer. We aim to demonstrate<br />

that correction <strong>of</strong> the FOXP3 expression defect can<br />

reverse these transformation causing genes and<br />

restore a healthy state.<br />

mir7<br />

mir101<br />

mir155<br />

mir19b<br />

mir21<br />

mir146a<br />

0.01 0.10 1.00 10.00<br />

Log Fold Change in Expression<br />

BT+Fp3 2<br />

BT+Fp3 1<br />

Fig 5: When FOXP3 is over expressed using a lentivirus in<br />

a breast cancer cell line, we observe increased<br />

expression <strong>of</strong> target micro RNAs.<br />

Techniques/skills/approach<br />

Molecular biology, RT PCR, microarray data analysis,<br />

tissue culture, lentiviral production, gene silencing<br />

and over expression, functional assay in breast<br />

cancer cells.<br />

References<br />

1. Zuo, T., et al., FOXP3 is a novel transcriptional repressor<br />

for the breast cancer oncogene SKP2. The Journal <strong>of</strong><br />

Clinical Investigation, 2007. 117(12): p. 3765-3773.<br />

2. Zuo, T., et al., FOXP3 is an x-linked breast cancer<br />

suppressor gene and an important repressor <strong>of</strong> the HER-<br />

2/ErbB2 oncogene. Cell, 2007. 129: p. 1275-1286.<br />

Collaborators<br />

Shaun McColl- Micro and Immunol, Adelaide<br />

Jenny Couper-Diabetes Endocrinology, Adeliade<br />

Greg Goodall– IMVS, Adelaide<br />

Tom Gonda – CICR, Brisbane<br />

Frances Shannon – JCSMR, ANU Canberra<br />

Heddy Zola – WCHRI, Adelaide<br />

What happens to <strong>Honours</strong> students from my lab?:<br />

2007 Kai Mak: 1st Class Hons. 2yrs as Research<br />

associate, starting a Ph.D in 2011.<br />

2008 Tessa Gargett: 1st Class Hons. Awarded the<br />

Derek Rowley prize for best in year. Currently a<br />

Research Associate in the Barry lab.<br />

Steve Pederson: 1st<br />

class Hons. Awarded APA<br />

(PhD scholarship) 2yr PhD in the Barry Lab.<br />

2009 Natasha McInnes: 1st<br />

class Hons. Awarded<br />

APA (PhD scholarship) 1yr PhD in the Barry lab.<br />

Danika Hill: 1st Class Hons, Awarded Derek<br />

Rowley prize for best in year. Currently a<br />

Research Associate in the Barry lab.<br />

<strong>Honours</strong> students are encouraged to continue on to<br />

PhD studies in my group.<br />

61 |


Molecular Neurogenetics<br />

Contact Person<br />

Dr Cheryl Shoubridge<br />

Phone: 8161 8105<br />

cheryl.shoubridge@adelaide.edu.au<br />

Supervisors<br />

Dr Cheryl Shoubridge<br />

Dr Kristie Lee<br />

Location: Women’s and Children’s Hospital<br />

Identification <strong>of</strong> genes and understanding <strong>of</strong> molecular mechanisms leading to intellectual disabilities, autisms and<br />

some epilepsies represents a challenge <strong>of</strong> significant medical importance. Our research seeks to further our<br />

understanding <strong>of</strong> human brain function through the identification <strong>of</strong> genes and characterisation <strong>of</strong> their naturally<br />

occurring mutations implicated in various disorders <strong>of</strong> the brain. The Molecular Neurogenetics laboratory focuses on<br />

identifying the molecular mechanisms and functional impact <strong>of</strong> mutations in genes causing X-linked intellectual<br />

disability (XLID).<br />

The key areas <strong>of</strong> research include utilizing primary neuronal cell culture models to investigate the functional impact<br />

<strong>of</strong> patient mutations in genes involved in X-linked Intellectual disability, in particular the ARX and IQSEC2 genes. We<br />

have animal models to investigate functional impact <strong>of</strong> the two most frequent expanded polyalanine tract<br />

mutations in the ARX gene. Our ongoing work aims to establish the molecular mechanisms <strong>of</strong> disease associated<br />

with a range <strong>of</strong> expanded polyalanine tract mutations in ARX to begin to understand how these mutations underpin<br />

the intellectual disability with and without a broad spectrum <strong>of</strong> associated clinical symptoms in affected patients,<br />

including epilepsy. We are also interested in more fundamental aspects <strong>of</strong> ARX biology and are currently<br />

investigating how phosphorylation may regulate the function <strong>of</strong> the ARX homeodomain transcription factor.<br />

<strong>Honours</strong> students are encouraged to remain with our research team to undertake a Ph.D.<br />

PROJECT (Basic): Do expanded polyalanine tract<br />

mutations in ARX disrupt neuronal migration?<br />

<strong>Project</strong> Background<br />

When the function <strong>of</strong> the Aristaless related<br />

homeobox gene (ARX) is lost due to severe<br />

mutations, patients are afflicted with catastrophic<br />

brain malformations such as smooth brain<br />

(lissencephaly) or hydrocephaly. Disruptions to<br />

normal neuronal migration underlie these conditions.<br />

Understanding the role <strong>of</strong> ARX in the cell fate and<br />

migration <strong>of</strong> neurons during the development <strong>of</strong> the<br />

brain remains a key goal. A less severe but more<br />

common mutation in ARX lead to expanded<br />

polyalanine tract (polyA expansions) with these<br />

patients displaying intellectual disability <strong>of</strong>ten in<br />

conjunction with seizures and other clinical features.<br />

We have mice that model the two most common<br />

human polyA expansions providing us an excellent<br />

tool to interrogate the role <strong>of</strong> ARX in neuronal<br />

development. The goal <strong>of</strong> this project would be to<br />

establish the effect <strong>of</strong> expanded polyalanine tract<br />

mutations on neuronal migration. Students would<br />

generate neurosphere cultures from the embryonic<br />

brains <strong>of</strong> wild-type and mutant mice. These<br />

neurospheres can then be manipulated in culture to<br />

assess aspects <strong>of</strong> neuronal migration and establish<br />

changes in the types and maturity <strong>of</strong> cells arising<br />

from the sphere. The findings will be validated in the<br />

brains <strong>of</strong> the mutant mice. The research techniques<br />

the student will be using include small animal<br />

handling and micro-dissection techniques,<br />

generation <strong>of</strong> primary neurospheres in culture,<br />

immun<strong>of</strong>lorescent techniques to identify different cell<br />

types in culture and a broad range <strong>of</strong> molecular<br />

techniques including real time PCR, RNA and protein<br />

extraction and electrophoresis.<br />

PROJECT (Basic): Understanding the molecular<br />

pathogenesis <strong>of</strong> polyalanine expansions in mice<br />

modelling intellectual disability and epilepsy.<br />

<strong>Project</strong> Background<br />

Intellectual disability (ID) is a disorder whereby<br />

affected individuals display an intelligent quotient<br />

≤70. It is estimated that 1-3% population suffer from ID,<br />

with which mutations in genes at the X chromosome<br />

account for up to 30% <strong>of</strong> the cases that have a<br />

genetic basis. The X chromosome gene Aristaless<br />

related homeobox gene (ARX) encodes a<br />

transcription factor that encompasses four<br />

polyalanine (polyA) tracts, and is one <strong>of</strong> the most<br />

frequently mutated ID genes. Mutations leading to<br />

extended polyalanine tract lengths (polyA<br />

expansions) are the most common mutations <strong>of</strong> ARX.<br />

Human patients with ARX polyA expansions all display<br />

intellectual disability with varying severity, while some<br />

also suffer from epilepsy. We have mice that model<br />

62 |


the two most common human polyA expansions, and<br />

our research to date has found that polyA mutations<br />

lead to a loss <strong>of</strong> ARX protein abundance during<br />

embryonic development. We are now interested in<br />

understanding the molecular basis that underlies ARX<br />

protein reduction, as well as elucidating downstream<br />

transcription targets that are differentially regulated<br />

as a result <strong>of</strong> the polyA expansions. The fundamental<br />

research techniques the student will be using include<br />

small animal handling and micro-dissection<br />

techniques, generation <strong>of</strong> primary neuronal cell<br />

culture, a broad range <strong>of</strong> molecular techniques<br />

including real time PCR, RNA and protein extraction<br />

and electrophoresis.<br />

References<br />

Shoubridge C,<br />

Fullston T and Gecz J. ARX spectrum<br />

disorders; making inroads in to the molecular<br />

pathology. Human Mutation 31(7) 2010.<br />

Kitamura, K., Itou, Y., Yanazawa, M., Ohsawa, M.,<br />

Suzuki-Migishima, R., Umeki, Y., Hohjoh, H.,<br />

Yanagawa, Y., Shinba, T., Itoh, M. et al. (2009) Three<br />

human ARX mutations cause the lissencephaly-like<br />

and mental retardation with epilepsy-like pleiotropic<br />

phenotypes in mice. Hum Mol Genet, 18, 3708-24.<br />

Shoubridge C<br />

, Tan MH Seiboth G and Gecz J.<br />

Missense mutations in the homeodomain <strong>of</strong> ARX<br />

abolish binding to DNA and cause a loss <strong>of</strong><br />

transcriptional repression activity. Human Molecular<br />

Genetics, 2012 Apr 1;21(7):1639-47.<br />

63 |


Nutritional Immunology<br />

Contact Persons<br />

Imme Penttila<br />

Phone : 8161 7072<br />

irmeli.penttila@adelaide.edu.au<br />

Beverly Muhlhausler<br />

Phone: 8313 0848<br />

beverly.muhlhausler@adelaide.edu.au<br />

Supervisors<br />

A/Pr<strong>of</strong> Imme Penttila<br />

Dr Beverly Mulhausler<br />

Pr<strong>of</strong> Robert Gibson<br />

Location: Women’s and Children’s <strong>Health</strong> Research<br />

Institute<br />

Biomarkers such as cytokines and eicosanoids are potent mediators that can act together and play a critical role in<br />

inflammation and other disease states. In some clinical trials, blood sample analysis is an integral part <strong>of</strong> assessing<br />

physiological and immune function in participants. This blood collection and analysis involves using volumes <strong>of</strong><br />

blood, which are not possible to obtain from new born infants. New born infants however undergo screening<br />

programs based on analysis <strong>of</strong> a single sample <strong>of</strong> capillary blood collected by heel prick between 3 and 14 days <strong>of</strong><br />

age. In this project we will aim to develop and validate a method <strong>of</strong> assessing cytokines and other markers in dried<br />

blood spot samples.<br />

In summary, this project will ideally suit an enthusiastic student who is interested in learning more more about<br />

nutritional intervention and the development <strong>of</strong> immuno-based assays for detecting biomarkers from associated<br />

clinical trials.<br />

PROJECT: (Basic) Measurement <strong>of</strong> immune markers in<br />

dried blood spots by immunoassay.<br />

WCHRI Scholarship available:<br />

Contact: 81617443<br />

Website http://www.wchri.com.au/<br />

<strong>Project</strong> Background<br />

Biomarkers such as cytokines and eicosanoids are<br />

potent mediators that can act together and play a<br />

critical role in inflammation and other disease states.<br />

In some clinical trials, blood sample analysis is an<br />

integral part <strong>of</strong> assessing physiological and immune<br />

function in participants. This blood collection and<br />

analysis involves using volumes <strong>of</strong> blood, which are<br />

not possible to obtain from new born infants. New<br />

born infants however undergo screening programs<br />

based on analysis <strong>of</strong> a single sample <strong>of</strong> capillary<br />

blood collected by heel prick between 3 and 14<br />

days <strong>of</strong> age. In this project we will aim to develop<br />

and validate a method <strong>of</strong> assessing cytokines and<br />

other markers in dried blood spot samples.<br />

In summary, this project will ideally suit an enthusiastic<br />

student who is interested in learning more about<br />

nutritional intervention and the development <strong>of</strong><br />

immuno-based assays for detecting biomarkers from<br />

associated clinical trials.<br />

Reference<br />

Skogstrand et.al. Effects <strong>of</strong> blood sample handling<br />

procedure on measurable inflammatory markers in<br />

plasma, serum, and dried blood spot samples. J<br />

Immunol Methods 336(2008) 78-84<br />

64 |


Paediatrics and Child <strong>Health</strong>, Flinders Medical Centre<br />

Supervisor/Contact Persons<br />

Dr Dani-Louise Dixon<br />

Phone: 8204 5494<br />

dani.dixon@flinders.edu.au<br />

Pr<strong>of</strong> Kevin Forsyth<br />

Phone: 8204 5259<br />

kevin.forsyth@flinders.edu.au<br />

Major Interests<br />

• Child <strong>Health</strong><br />

• Strategic planning in health care delivery for children<br />

• Paediatric Immunology and Paediatric Infectious Diseases<br />

• Research in the immunopathology <strong>of</strong> inflammation, particularly viral induced respiratory disease in infants<br />

• Immunisation<br />

• Medical Education<br />

• Curriculum reform in the teaching <strong>of</strong> Child <strong>Health</strong> at both undergraduate and postgraduate levels<br />

• Use <strong>of</strong> Information Technology in teaching and learning<br />

PROJECT: (Clinical) Decreasing neutrophil-induced<br />

lung damage in respiratory syncytial virus (RSV)<br />

infection: a means to ameliorate infant bronchiolitis.<br />

<strong>Project</strong> Background<br />

Bronchiolitis is the most common severe respiratory<br />

tract illness in infants and remains a major cause <strong>of</strong><br />

infant hospitalisation as currently there is no<br />

treatment. Strong evidence suggests that the virus<br />

which causes bronchiolitis, RSV, induces host immune<br />

cells (neutrophils) to damage the lung, increasing<br />

disease severity as well as leading to the<br />

development <strong>of</strong> asthma in up to 50% <strong>of</strong> patients.<br />

Recently our lab demonstrated the ability <strong>of</strong> a<br />

protein, feG, to prevent and treat such lung<br />

damage.<br />

We aim to test the therapeutic potential <strong>of</strong> feG in<br />

decreasing lung damage caused by RSV, utilising a<br />

range <strong>of</strong> techniques which may include in vitro (cell<br />

culture), animal models, and/or clinical (human)<br />

aspects.<br />

The project will be conducted in the Lung Injury<br />

Laboratory within the discipline <strong>of</strong> Critical Care<br />

Medicine. The lab has expertise in fields as diverse as<br />

immunology, pathology, anatomy, histology,<br />

physiology, and molecular biology and undertakes<br />

research that crosses all <strong>of</strong> these disciplines, in both<br />

laboratory based and clinical projects. The<br />

Laboratory has an excellent international reputation<br />

and an outstanding record <strong>of</strong> obtaining funding from<br />

government and non-government sources resulting in<br />

regular publication in top international journals.<br />

65 |


Public <strong>Health</strong> Research Unit<br />

Supervisors<br />

Associate Pr<strong>of</strong>essor Peter Baghurst<br />

Associate Pr<strong>of</strong>essor Maree O’Keefe<br />

Jacqueline Stephens<br />

Location: Women’s and Children’s Hospital<br />

Contact Person<br />

Jacqueline Stephens<br />

Phone: 8161 7790<br />

jacqueline.stephens@adelaide.edu.au<br />

The Public <strong>Health</strong> Research Unit (PHRU) plays a strong support role in the planning and provision <strong>of</strong> efficient, effective<br />

and appropriate health services within the Child, Youth and Women’s <strong>Health</strong> Service. Staff <strong>of</strong> the PHRU are affiliated<br />

with the Children’s Research Centre and <strong>Health</strong>y Development Adelaide, <strong>University</strong> <strong>of</strong> Adelaide. The PHRU aims to<br />

achieve better health outcomes for consumers and their communities, with a strong focus on epidemiology, social<br />

epidemiology and public health.<br />

PROJECT: (Clinical) What influences adolescents to<br />

decide to have surgical intervention for tonsillitis?<br />

<strong>Project</strong> Background<br />

The removal <strong>of</strong> tonsils is a very common surgical<br />

procedure. While most tonsillectomies occur in very<br />

young children, a considerable number <strong>of</strong><br />

tonsillectomies are also performed in adolescents.<br />

While the research literature discusses the frequency,<br />

severity and duration <strong>of</strong> tonsillitis during the years<br />

preceding tonsillectomy in adolescents, little is known<br />

about the adolescent’s role in agreeing to the<br />

procedure, which is not without significant<br />

postoperative morbidity. Anecdotal experience has<br />

shown that adolescents present for consideration <strong>of</strong><br />

tonsillectomy because <strong>of</strong> the impact it is having on<br />

their education, sport and social life. A better<br />

understanding <strong>of</strong> the motivations for wanting surgical<br />

intervention, the expectations <strong>of</strong> the clinical<br />

encounter, and the experience <strong>of</strong> surgery may help<br />

improve our management <strong>of</strong> this under-studied<br />

group.<br />

The research will include interviews with adolescents<br />

and parents/guardians to gain an understanding <strong>of</strong><br />

what influences the decision to elect for surgical<br />

intervention for tonsillitis. The study will also aim to<br />

understand how adolescents assess their tonsillar<br />

health and how much influence they have in the<br />

decision to have surgical intervention.<br />

Training<br />

The student will be trained in literature reviews,<br />

interviewing and interview design, recruitment, data<br />

entry, data analysis and manuscript preparation.<br />

66 |


Vaccine Safety Research Group<br />

Supervisor/Contact Person<br />

Dr Mike Gold<br />

Phone: 8161 7266<br />

michael.gold@adelaide.edu.au<br />

Location: Women’s and Children’s Hospital<br />

Despite the public health benefit <strong>of</strong> vaccination, concern about the safety <strong>of</strong> vaccines may be the most important<br />

issue to affect immunisation coverage and the sustainability <strong>of</strong> an immunisation program. There is a limited capacity<br />

to evaluate all aspects <strong>of</strong> vaccine safety before vaccines are approved for use. This is because the safety<br />

evaluation <strong>of</strong> new vaccines takes place during clinical trials, which usually include thousands <strong>of</strong> healthy subjects<br />

(which is an adequate number for the purposes <strong>of</strong> detecting common adverse events), and involves a short period<br />

<strong>of</strong> post-vaccination monitoring. Unfortunately, this process does not identify adverse events which are rare,<br />

delayed, occur with specific vaccine combinations or in a sub-group <strong>of</strong> children with co-existing disorders.<br />

Passive surveillance <strong>of</strong> adverse events following immunisation (AEFI) is used as the primary mechanism for safety<br />

surveillance following vaccine licensure; however, this also has limitations which include underreporting and the<br />

inability to establish a casual relationship between immunisation and a specific adverse event. In Australia, other<br />

mechanisms <strong>of</strong> surveillance are currently in use; known as enhanced passive surveillance and active sentinel<br />

surveillance. Given the limitation <strong>of</strong> resources, it is important to determine the most effective mechanism <strong>of</strong><br />

surveillance for an AEFI.<br />

The vaccine safety research group is a multi-disciplinary group, funded by the Australian Research Council (ARC) to<br />

investigate the role <strong>of</strong> data linkage to assess vaccine safety in Australia. The project involves an investigation <strong>of</strong> the<br />

technical feasibility and privacy issues required for data linkage. One <strong>of</strong> the aims <strong>of</strong> the project will be to compare<br />

the effectiveness <strong>of</strong> surveillance, using data linkage, with other methods <strong>of</strong> surveillance currently being used in<br />

Australia. This honours project will assess the performance <strong>of</strong> three methods <strong>of</strong> surveillance for three sentinel<br />

conditions which may occur after immunisation.<br />

PROJECT: (Clinical/Basic) A comparative analysis <strong>of</strong><br />

the surveillance <strong>of</strong> intussusceptions, seizures and<br />

anaphylaxis following immunisation in Australia –<br />

2007 to 2009.<br />

<strong>Project</strong> Background<br />

Intussusception, seizures, and anaphylaxis are all<br />

important adverse events for surveillance following<br />

immunisation. In Australia, health providers are asked<br />

to report these events to the Adverse Drug Reactions<br />

Committee (ADRAC) <strong>of</strong> the Therapeutic Goods<br />

Administration (TGA). However, these events have<br />

also been under surveillance using enhanced passive<br />

surveillance and active sentinel surveillance. The<br />

Australian Paediatric Surveillance Unit (APSU)<br />

conducts enhanced passive surveillance by<br />

requesting monthly AEFI reports from paediatricians.<br />

Active sentinel surveillance is conducted through a<br />

Commonwealth initiative known as Paediatric<br />

Enhanced Disease Surveillance (PAEDS). This scheme<br />

funds part-time specialist nurses in four tertiary care<br />

hospitals in Australia (SA, NSW, Vic, and WA) to<br />

identify children with an AEFI. Detailed clinical data<br />

and biological samples are collected and additional<br />

information on risk factor and vaccination history is<br />

sought from parents. The aim <strong>of</strong> the project will be to<br />

evaluate the effectiveness <strong>of</strong> these three surveillance<br />

methods by comparing data, which has already<br />

been collected. The skills required will be an interest<br />

in one or more <strong>of</strong> the following areas: public health,<br />

pharmacovigilance, paediatrics or immunisation.<br />

The techniques will involve analysis <strong>of</strong> AEFI reports<br />

and comparative data analysis. The student will be<br />

supported by the group who have skills in statistical<br />

analysis <strong>of</strong> data, and vaccine safety.<br />

References<br />

JML Brotherton, MS Gold, AS Kemp, PB McIntyre, MA<br />

Burgess, S Campbell-Lloyd, on behalf <strong>of</strong> the New South Wales<br />

<strong>Health</strong> HPV Adverse Events Panel CMAJ. 2008 Sep<br />

9;179(6):525-33.<br />

Monitoring vaccine safety: a critical component <strong>of</strong><br />

every immunization programme Brotherton J Gold M. Med J<br />

Australia 2008: 189(5):243-244<br />

Human papillomavirus vaccine hypersensitivity<br />

reactions – a retrospective cohort study in Australia<br />

Liew WK, Nigel Crawford N, Tang M, Buttery J, Royle J, Gold<br />

M, Zeigler C, Quinn P, Elia S, Choo S. BMJ 2008: 337;a2642<br />

Guidelines for collection, analysis and presentation <strong>of</strong><br />

vaccine safety data in pre- and post-licensure clinical<br />

studies. Bonhoeffer J, Bentsi-Enchill A, Chen RT, Fisher MC,<br />

Gold MS et al ; The Brighton Collaboration Methods Working<br />

Group. Vaccine. 2009 Apr 6;27(16):2282-2288.<br />

Guidelines for collection, analysis and presentation <strong>of</strong><br />

vaccine safety data in surveillance systems. Bonhoeffer J,<br />

Bentsi-Enchill A, Chen RT, Fisher MC, Gold MS, et al The<br />

Brighton Collaboration Methods Working Group. Vaccine.<br />

2009 Apr 6;27(16):2289-2297. Epub 2008 Dec 4<br />

67 |


Vaccinology and Immunology Research Trials Unit<br />

Supervisor/Contact Person (Far left)<br />

Dr Helen Marshall<br />

Phone: 8161 8115<br />

helen.marshall@adelaide.edu.au<br />

Location: Women’s and Children’s Hospital<br />

The Vaccinology and Immunology Research Trials Unit, is a multidisciplinary immunisation research group at the<br />

Women’s and Children’s Hospital, affiliated with the <strong>University</strong> <strong>of</strong> Adelaide and part <strong>of</strong> the Children’s Research<br />

Centre, <strong>University</strong> <strong>of</strong> Adelaide. The centre’s membership includes internationally renowned experts in paediatrics,<br />

immunology and public health. VIRTU’s research program includes clinical trials in investigational vaccines,<br />

infectious disease epidemiology, social epidemiology, immunology and public health.<br />

Current research programs include:<br />

1. Investigational vaccines; meningococcal B vaccine, (pre) pandemic flu vaccines, influenza vaccine, pertussis<br />

immunisation at birth, Respiratory Syncytial Virus (RSV) and Parainfluenza Virus vaccine (PIV3)<br />

2. Infectious disease epidemiology; severity <strong>of</strong> pertussis (whooping cough) infection in hospitalised children, RSV<br />

epidemiology and severity <strong>of</strong> disease in hospitalised children, rotavirus vaccine effectiveness<br />

3. Immunisation <strong>of</strong> children at special risk; Human Papillomavirus vaccine<br />

4. Social epidemiology; Pandemic influenza community preparedness, HPV school immunisation program; why<br />

are girls opting out?, and enhanced surveillance <strong>of</strong> paediatric conditions <strong>of</strong> public health importance<br />

Clinical/Public <strong>Health</strong> Relevance<br />

Conducting trials in investigational vaccines provides local Australian data for licensing authorities. Studies <strong>of</strong><br />

investigational vaccines conducted at our centre provide Australian data for regulatory authorities such as the<br />

Therapeutic Goods Administration (TGA) and Food and Drug Administration, (FDA, US). It is an advantage for both<br />

vaccine companies and the TGA to have safety and immunogenicity data based on the Australian population<br />

when licensing <strong>of</strong> a new vaccine is being considered. Other significant contributions include studies in new<br />

combination vaccines. This allows a reduction in the number <strong>of</strong> needles that need to be administered to infants<br />

and children making vaccination more acceptable (and more cost-effective with less time spent in the clinic) for all<br />

involved. Many <strong>of</strong> the new vaccine combinations trialled by our unit are now licensed (or have been filed for<br />

licensing) for use in Australian children including a combination measles, mumps, rubella, varicella vaccine. Studies<br />

in vaccine safety identify adverse reactions associated with vaccination and provide data that support changes to<br />

the vaccination schedule to reduce the adverse effects associated with immunisation. Studies in epidemiology <strong>of</strong><br />

disease provide data used to determine whether development <strong>of</strong> a vaccine to prevent an infection is feasible and<br />

guides the scheduling <strong>of</strong> the vaccine in the National Immunisation Program. Our group also conducts research into<br />

community attitudes and acceptance <strong>of</strong> new vaccines to encourage engagement <strong>of</strong> the community in<br />

immunisation policy decisions. All studies have resulted in a publication or recent preparation <strong>of</strong> a manuscript for<br />

publication.<br />

PROJECT: (Clinical) Are there differences in severity <strong>of</strong><br />

illness before and after introduction <strong>of</strong> the infant<br />

rotavirus vaccination program in children who are<br />

admitted to hospital with gastroenteritis?<br />

<strong>Project</strong> Backgound<br />

The introduction <strong>of</strong> rotavirus vaccination for infants at<br />

2, 4 and 6 months <strong>of</strong> age in July 2007 has had a<br />

significant impact on the number <strong>of</strong> children<br />

hospitalised with gastroenteritis in South Australia.<br />

Recent research from several states in Australia has<br />

indicated that the reduction in severe rotavirus<br />

infections is greater than 80% following the<br />

introduction <strong>of</strong> the vaccination program. Another<br />

important question remaining is whether there has<br />

been any impact on the severity <strong>of</strong> illness for children<br />

who still present to hospital with gastroenteritis<br />

following the introduction <strong>of</strong> the vaccination<br />

program. This project aims to review hospitalisations<br />

occurring in children before and after the<br />

68 |


introduction <strong>of</strong> the rotavirus vaccination program<br />

and to assess severity using two internationally<br />

recognised severity scoring systems.<br />

PROJECT: (Clinical) Can presenting symptoms in<br />

children who present to emergency departments with<br />

pertussis (whooping cough) infection, predict severity<br />

<strong>of</strong> disease?<br />

<strong>Project</strong> Backgound<br />

Although pertussis immunisation was introduced in<br />

Australia 50 years ago, initially as a whole cell<br />

pertussis vaccine and subsequently as an acellular<br />

vaccine, pertussis remains a poorly controlled<br />

infection with deaths still occurring in young infants.<br />

In Australia, the majority <strong>of</strong> severe pertussis occurs<br />

under 6 months <strong>of</strong> age, despite high immunisation<br />

coverage in children over 6 months <strong>of</strong> age.<br />

Unimmunised infants are most likely to develop<br />

severe disease requiring hospitalisation, frequently<br />

complicated by apnoea, seizures, encephalopathy<br />

or death. Approximately 400 infants are hospitalised<br />

each year in Australia, the majority being < 6 months<br />

<strong>of</strong> age. Eighteen deaths due to pertussis occurred in<br />

Australia during 1993 - 2004, all but two <strong>of</strong> these<br />

children were less than 6 months <strong>of</strong> age. Five deaths<br />

in infants < 6 months occurred during the current<br />

2009-2010 epidemic in Australia.<br />

We have recently developed a severity scoring<br />

system for pertussis and this project will aim to add<br />

further data to support our understanding <strong>of</strong> pertussis<br />

disease in Australia with a specific aim <strong>of</strong> establishing<br />

whether clinical signs and symptoms at presentation<br />

can predict the likelihood <strong>of</strong> severe disease requiring<br />

more intensive management and care. The ability to<br />

predict which children are likely to require intensive<br />

care and management early has the potential to<br />

improve practice and outcomes for vulnerable<br />

children.<br />

Training<br />

The student will be trained in literature reviews,<br />

human research ethics, data entry and analysis and<br />

manuscript preparation.<br />

Recent Publications<br />

Clarke M, Davidson G, Gold M and Marshall H.<br />

Decline in Gastroenteritis Admissions in South Australia<br />

in both vaccinated and unvaccinated children post<br />

introduction <strong>of</strong> rotavirus vaccine. Vaccine. 2011 Jun<br />

24;29(29-30):4663-7.<br />

Quinn P, Gold M, Royle J, Buttery J, Richmond<br />

P, McIntyre P, Wood N, Lee SS, Marshall H.<br />

Recurrence <strong>of</strong> extensive injection site reactions<br />

following DTPa or dTpa vaccine in children 4-6 years<br />

old. Vaccine. 2011 Jun 6;29(25):4230-7.<br />

Marshall H, Tooher R, Collins J, Mensah F,<br />

Braunack-Mayer A, Street J, Ryan P. Awareness,<br />

anxiety, compliance: Community perceptions and<br />

response to the threat and reality <strong>of</strong> an influenza<br />

pandemic. Am J Infection Control. In Press<br />

Khandaker G, Marshall H, Peadon E, Zurynski Y,<br />

Burgner D, Buttery J, Gold M, Nissen M, Elliott E, Booy<br />

R. Congenital and neonatal varicella: Impact <strong>of</strong> the<br />

national varicella vaccination program in Australia.<br />

Arch Dis Child. 2011 May;96(5):453-6.<br />

Booy R, Richmond P, Nolan T, McVernon J,<br />

Marshall H, Nissen M, Reynolds G, Ziegler JB, Heron L,<br />

Lambert S, Caubet M, Mesaros N, Boutriau D.<br />

Immediate and longer term immunogenicity <strong>of</strong> a<br />

single dose <strong>of</strong> the combined haemophilus influenzae<br />

type B-Neisseria meningitidis serogroup C-tetanus<br />

toxoid conjugate vaccine in primed toddlers 12 to 18<br />

months <strong>of</strong> age. Pediatr Infect Dis J. 2011<br />

Apr;30(4):340-2.<br />

Nolan T, Richmond P, Marshall H, McVernon J,<br />

Alexander K, Mesaros N, Aris E, Miller J, Poolman J,<br />

Boutriau D. Immunogenicity and safety <strong>of</strong><br />

vaccination with a combined Haemophilus influenza<br />

type b - Neiseria meningitidis serogroups C and Y –<br />

tetanus toxoid candidate vaccine (HibMenCY-TT).<br />

Pediatr Infect Dis J. 2011 Mar;30(3):190-6.<br />

Marshall H, Nolan T, Diez-Domingo J, Rombo L,<br />

Sokal E, Mares J, Casanovas J, Kuriyakose S, Leyssen<br />

M, J Jacquet. Long-term (5 year) antibody<br />

persistence following two and three dose regimens <strong>of</strong><br />

a combined hepatitis A and B vaccine in children<br />

aged 1 – 11 years. Vaccine. 2010 Jun 17;28(27):4411-<br />

5.<br />

Nolan T, McVernon J, Skeljo M, Richmond P,<br />

Wadia U, Lambert S, Nissen M, Marshall H, Booy R,<br />

Heron L, Hartel G, Lai M, Basser R, Gittleson C,<br />

Greenberg M. Immunogenicity <strong>of</strong> a monovalent 2009<br />

influenza A (H1N1) vaccine in infants and children – a<br />

randomized trial. Journal <strong>of</strong> the American Medical<br />

Association, 2010;303(01):37-46.<br />

69 |


Respiratory and Sleep Medicine<br />

Supervisor/Contact Person<br />

Dr David Parsons<br />

Phone: 8161 7004<br />

david.parsons@adelaide.edu.au<br />

Our group is developing an effective treatment and potentially a life-long correction for the airway disease <strong>of</strong> Cystic<br />

Fibrosis (CF). This lung disease is the primary cause <strong>of</strong> worsening health problems in young people with CF, greatly<br />

affecting their quality <strong>of</strong> life and is the overwhelming cause <strong>of</strong> early death. We have been supported by grants from<br />

the NH&MRC, the USA CF Foundation, CF Australia and South Australian research funding bodies where we use a<br />

lentivirus-based gene delivery vector to study how to produce safe and effective gene delivery into airway cells.<br />

We use reporter genes such as LacZ and luciferase, to prepare for studies with the therapeutic gene CFTR.<br />

Our work has been focussed across three animal models: mice, sheep and marmosets. Our approach especially<br />

targets airway stem cells in vivo, and in some mouse studies this approach has already produced up to lifetime gene<br />

expression after a single dose event. The field has lacked a simple way to measure the success in airways and lungs,<br />

whether for humans or animal models. To overcome that limitation we have developmed new synchrotron X-ray<br />

imaging techniques with Monash <strong>University</strong> colleagues. Synchrotron Xrays allow non-invasive viewing <strong>of</strong> airway<br />

surfaces in live mice, at resolutions close to that <strong>of</strong> normal light microscopes. We are also now able to examine<br />

mouse lung airflows in 3-D. With further development we expect these techniques will create new options for rapid<br />

and accurate assessment <strong>of</strong> potential therapies (such as gene-correction) for their effectiveness in improving airway<br />

health in live and intact animal models.<br />

Our research is closely connected with the clinical needs <strong>of</strong> CF children, and most <strong>of</strong> our studies are carried out at<br />

the Women’s & Children’s hospital campus in Adelaide. The synchrotron X-ray studies are performed in Japan, and<br />

in 2010 parallel studies began with the assistance <strong>of</strong> Monash <strong>University</strong> collaborators at the new Australian<br />

Synchrotron in Melbourne. Finally, we have new state-<strong>of</strong>-the-art research laboratories in 2012! Through the<br />

enthusiastic initial work <strong>of</strong> the Cure4CF Foundation Ltd (www.cure4cf.org), and substantial completion funding<br />

provided by the WCH Foundation, the Alan Scott CF research laboratory is now in place in the Gilbert Building at the<br />

WCH<br />

PROJECT (Basic): Airway gene transfer for cystic<br />

fibrosis: converting fluid-based lentiviral gene vector<br />

delivery to aerosol delivery.<br />

.R.<br />

This study is designed to translate our well established<br />

and effective protocols for bolus-delivery <strong>of</strong> the<br />

lentivirus vector in airway into one that can be<br />

provided in an aerosol form. Aerosols are considered<br />

to be the most practical way to deliver lung gene<br />

therapies in the future.<br />

Preliminary data from our group indicates that the<br />

viability <strong>of</strong> the vector can be largely preserved with<br />

the incorporation <strong>of</strong> specific supporting protein<br />

agents in the viral vector carrier fluid. Using a<br />

clinically-applicable electronic aerosolisation device<br />

(Aerogen) the study will be designed to define the<br />

parameters <strong>of</strong> vector delivery to maintain viability for<br />

in-vitro assessment.<br />

Once those parameters have been defined aerosol<br />

delivery will be tested in the nasal and lung airways<br />

<strong>of</strong> laboratory mice. The outcome sought in this<br />

<strong>Honours</strong> year is to define an effective aerosol-based<br />

delivery using in-vitro methods and apply it in-vivo to<br />

laboratory mouse airways.<br />

PROJECT (Basic): Enhancing airway gene transfer for<br />

cystic fibrosis treatment: effects <strong>of</strong> agents repurposed<br />

from current use in clinical care.<br />

.R.<br />

Agents used clinically to clear CF airways <strong>of</strong> mucus<br />

and cellular debris may be helpful in improving initial<br />

gene transfer into airway surface epithelial cells, by<br />

enabling better access by the gene vector to the<br />

defective epithelial cells lining the affected airways.<br />

Initial studies will utilise airway-specific cell cultures to<br />

screen a range <strong>of</strong> these potentially useful<br />

compounds, with lead candidates moving to<br />

preliminary testing using our established gene transfer<br />

methods in the airways <strong>of</strong> laboratory mice.<br />

The outcome desired is to achieve more efficient<br />

levels <strong>of</strong> airway gene expression with reporter-genes,<br />

to improve the future opportunities for development<br />

<strong>of</strong> therapeutically active and effective airway gene<br />

expression using the corrective CFTR gene.<br />

70 |


Index<br />

Breast Biology and Cancer, 16<br />

Child Nutrition Research Centre (CNRC), 49<br />

Circadian Physiology Group, 18<br />

CNS Therapeutics Laboratory, Lysosomal Diseases Research Unit, 52<br />

Crani<strong>of</strong>acial Research Group, 53<br />

Developmental Biology <strong>of</strong> the Ovary, 20<br />

Early Development Group, 23<br />

Early Life Programming <strong>of</strong> <strong>Health</strong> and Disease, 25<br />

Early Origins <strong>of</strong> Cancer, 30<br />

Gamete and Embryo Biology Laboratory, 31<br />

Lysosomal Biology, Lysosomal Diseases Research Unit, 55<br />

Matrix Biology Unit, 57<br />

Molecular Immunology, 59<br />

Molecular Neurogenetics, 62<br />

Nutritional Immunology, 64<br />

Oocyte Biology Group, 32<br />

Paediatric Trials Unit, 68<br />

Paediatrics and Child <strong>Health</strong>, 65<br />

Placental Development Laboratory, 34<br />

Pregnancy and Development Group, 37<br />

Public <strong>Health</strong> Research Unit, 66<br />

Reproductive Biotechnology Group, 39<br />

Reproductive Cancer Research Group, 40<br />

Reproductive endocrinology and reproductive medicine group, 43<br />

Reproductive Immunology Group, 44<br />

Respiratory and Sleep Medicine, 70<br />

The Australian Research Centre for <strong>Health</strong> <strong>of</strong> Women and Babies, 12<br />

Vaccine Safety Research Group, 67<br />

71 |


School <strong>of</strong> Paediatrics and Reproductive <strong>Health</strong><br />

The <strong>University</strong> <strong>of</strong> Adelaide<br />

Telephone + 61 8 8313 5100<br />

Facsimile + 61 8313 4099<br />

Web http://health.adelaide.edu.au/school_paedrepro/honours

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!