13.07.2013 Views

Current Honours Projects - Faculty of Medicine, Nursing and Health ...

Current Honours Projects - Faculty of Medicine, Nursing and Health ...

Current Honours Projects - Faculty of Medicine, Nursing and Health ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

<strong>Honours</strong> in Pharmacology<br />

2013<br />

www.monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

Welcome to the Pharmacology Department!<br />

The <strong>Honours</strong> year represents a new adventure, very different to your undergraduate<br />

experience, in which you will have the opportunity to undertake a research project,<br />

communicate your science to colleagues <strong>and</strong> peers <strong>and</strong> learn to critically evaluate scientific<br />

concepts <strong>and</strong> literature.<br />

Your supervisor(s) will be there to guide <strong>and</strong> advise you along this research journey.<br />

At the very least, you are expected to bring with you the following skills set, in no particular<br />

order:<br />

-enthusiasm<br />

-an enquiring mind<br />

-respect & humility<br />

-determination & persistence<br />

-a sense <strong>of</strong> humour<br />

-a collegial spirit<br />

-patience<br />

This booklet provides information about the research projects on <strong>of</strong>fer in the Department <strong>of</strong><br />

Pharmacology <strong>and</strong> we encourage you to identify the areas <strong>of</strong> research in which you are most<br />

interested, contact potential supervisors <strong>and</strong> discuss the projects with them.<br />

The course convenors, A/Pr<strong>of</strong> Grant Drummond <strong>and</strong> Dr Barb Kemp-Harper, can advise on<br />

projects, guide you through the application process <strong>and</strong> help with any queries you may have.<br />

We look forward to welcoming you the Department <strong>of</strong> Pharmacology in 2013 <strong>and</strong> wish you<br />

all the best for a rewarding <strong>and</strong> exciting year <strong>of</strong> research.<br />

Good luck!<br />

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

Head, Department <strong>of</strong> Pharmacology<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

Department <strong>of</strong> Pharmacology <strong>Honours</strong> Convenors<br />

Dr Barb Kemp-Harper A/Pr<strong>of</strong> Grant Drummond<br />

Email: Barbara.Kemp@monash.edu Email: Grant.Drummond@monash.edu<br />

Phone: 9905 4674 Phone: 9905 4869<br />

Pharmacology <strong>Honours</strong>: Pre-requisites<br />

BSc / BBNSc / BBiotech BMS<br />

Pre-requisites A Distinction average (>70)<br />

in 24 points at 3 rd year level,<br />

including at least 18 points<br />

in 3 rd year PHA units<br />

A Distinction average (>70)<br />

in 24 points at 3 rd year<br />

level, including 12 points in<br />

3 rd year core BMS units<br />

<strong>and</strong> 12 points in other 3 rd<br />

year units<br />

Application closing date 19 th November, 2012 16 th November, 2012<br />

Application form www.sci.monash.edu.au/undergra<br />

d/honours/apply.html<br />

www.med.monash.edu/biomed/<br />

honours/<br />

Commencement date 25 th February, 2013 25 th February, 2013<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

Honour <strong>Projects</strong> 2013 – On campus projects<br />

LABS / SUPERVISOR(S)<br />

Clinical & Experimental Toxicology<br />

Group<br />

Andis Graudins<br />

Education-based projects<br />

Eva Patak, Liz Davis<br />

Fibrosis Group<br />

Chrishan Samuel, Simon Royce<br />

Integrative Cardiovascular<br />

Pharmacology Group<br />

Tracey Gaspari, Rob Widdop<br />

Claudia McCarthy, Antony Vinh, Rob<br />

Widdop<br />

Antony Vinh, Rob Widdop<br />

Neuropharmacology Group<br />

Richard Loiacono, Siew Yeen Chai<br />

(Physiology)<br />

Vascular Biology &<br />

Immunopharmacology Group<br />

Brad Broughton, Chris Sobey<br />

Grant Drummond, Rebecca Lim<br />

Barb Kemp-Harper<br />

Barb Kemp-Harper, Grant Drummond<br />

Helena Kim, Chris Sobey<br />

Alyson Miller<br />

Stavros Selemidis<br />

PROJECT TITLE<br />

• An assessment <strong>of</strong> fructose-1,6-diphosphate alone <strong>and</strong> in<br />

concert with other inotropic agents on inotrope sparing<br />

in the treatment <strong>of</strong> beta-receptor antagonist poisoning<br />

• Evaluation <strong>of</strong> web-based pharmacology resources<br />

• Investigating novel anti-fibrotic therapies<br />

• Cardio-protective effects <strong>of</strong> incretin hormones: potential<br />

beyond glycaemic control?<br />

• Mechanisms <strong>of</strong> angiotensin type-2 receptor-mediated<br />

neuroprotection during stroke<br />

• Role <strong>of</strong> T cells during the development <strong>of</strong> high blood<br />

pressure<br />

• Modulation <strong>of</strong> neuronal oxytocin – effects on social<br />

behaviour<br />

• Are the adverse effects <strong>of</strong> hormone replacement therapy<br />

on stroke outcome mediated by G protein-coupled<br />

estrogen receptor signalling?<br />

• Stem cell therapy for the treatment <strong>of</strong> high blood<br />

pressure<br />

• Exploring the vasoprotective actions <strong>of</strong> novel HNO<br />

donors<br />

• Exploring novel therapeutic strategies to prevent<br />

atherosclerotic plaque rupture<br />

• Exploring the role <strong>of</strong> endothelial NF-KB on ischemic<br />

stroke outcome<br />

• Investigating mechanisms linking hypertension <strong>and</strong><br />

cognitive impairment<br />

• Novel pharmacological targets for Influenza A virus<br />

induced lung inflammation<br />

• Underst<strong>and</strong>ing the biology <strong>of</strong> reactive oxygen species in<br />

cancer<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

Honour <strong>Projects</strong> 2013 – Off campus projects<br />

LABS/SUPERVISOR(S)<br />

Australian Centre for Blood<br />

Diseases<br />

Justin Hamilton, Grant Drummond<br />

Baker IDI Heart & Diabetes Institute<br />

Jaye Chin-Dusting, Jennifer Irvine,<br />

Rob Widdop<br />

Ge<strong>of</strong>f Head, Pamela Davern<br />

Ge<strong>of</strong>f Head, Pamela Davern,<br />

S<strong>and</strong>ra Burke<br />

Ge<strong>of</strong>f Head, Pamela Davern, Joon Lim<br />

Rebecca Ritchie, Barb Kemp-Harper<br />

Rebecca Ritchie<br />

Drug Discovery Biology Theme<br />

Monash Institute <strong>of</strong> Pharmaceutical<br />

Sciences<br />

Michelle Halls, Roger Summers<br />

Rob Lane, Arthur Christopoulos<br />

Katie Leach, Laura Lopez, Arthur<br />

Christopoulos<br />

Katie Leach, Arthur Christopoulos<br />

Patricia Rueda, Arthur Christopoulos,<br />

Chris Langmead<br />

Celine Valant, Lauren May<br />

Celine Valant, Meritxel Canals<br />

Celine Valant, Arthur Christopoulos<br />

PROJECT TITLE<br />

• Defining the function <strong>of</strong> human platelet thrombin<br />

preceptor, PAR4, in arterial thrombosis<br />

• What turns you on? Using a novel RNAi-based<br />

approach to determine platelet activation signals in<br />

thrombosis<br />

• The role <strong>of</strong> AT2R in macrophage polarisation<br />

• Effects <strong>of</strong> positive allosteric modulator <strong>of</strong> GABAA<br />

receptors on hypertension <strong>and</strong> stress<br />

• Role <strong>of</strong> brain pathways in chronic stress<br />

• Which parts <strong>of</strong> the brain are affected in obesity related<br />

hypertension?<br />

• Nitroxyl, a relative <strong>of</strong> NO, is a naturally-occurring<br />

cardioprotective molecule<br />

• New strategies to rescue diabetes induced cardiac<br />

dysfunction<br />

• Targeting anti-inflammatory protein annexin-A1 for<br />

protection from myocardial infarction (heart attack)<br />

• Compartmentalisation <strong>of</strong> relaxin-stimulated signalling in<br />

single cells<br />

• Molecular mechanisms <strong>of</strong> a novel allosteric modulator <strong>of</strong><br />

the D2-like dopamine receptors<br />

• Probing an allosteric binding site in the human CASR<br />

• Allosteric pharmacoregulation <strong>of</strong> a headless CASR<br />

• Investigation <strong>of</strong> muscarinic M4 receptor function using<br />

label free impedance technology<br />

• Investigating the signalling cascade <strong>of</strong> novel adenosine<br />

receptor lig<strong>and</strong>s at the adenosine A2B receptor (A2B-<br />

AR)<br />

• Investigation <strong>of</strong> an absolutely conserved pocket at the<br />

muscarinic acetylcholine M1 receptor (M1 MACHR)<br />

• Investigation <strong>of</strong> an absolutely conserved pocket at the<br />

muscarinic acetylcholine M2 receptor (M2 MACHR)<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

AN ASSESSMENT OF FRUCTOSE-1,6-DIPHOSPHATE ALONE<br />

AND IN CONCERT WITH OTHER INOTROPIC AGENTS ON<br />

INOTROPE SPARING IN THE TREATMENT OF<br />

BETA-RECEPTOR ANTAGONIST POISONING<br />

Supervisors: Pr<strong>of</strong>essor Andis Graudins<br />

Location: Clinical <strong>and</strong> Experimental Toxicology Laboratory<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

Heart failure <strong>and</strong> shock resulting from drug poisoning may be a difficult treatment<br />

challenge in the clinical setting. Severe poisoning with beta-receptor antagonists such as<br />

propranolol may result in hypotension (low blood pressure) that may not respond<br />

adequately to st<strong>and</strong>ard treatment measures such as infusion <strong>of</strong> adrenaline or other<br />

inotropic therapies. Treatment <strong>of</strong>ten results in the administration <strong>of</strong> multiple medications<br />

in an attempt to maintain blood pressure. In this stressed state, the myocardium is less<br />

effective in its ability to produce energy substrates (such as ATP) from glucose <strong>and</strong> free<br />

fatty acids.<br />

Fructose-1-6,diphosphate (FDP) is endogenously produced in human cells as an<br />

intermediary metabolite <strong>of</strong> the glycolytic pathway. Its production requires the<br />

expenditure <strong>of</strong> 2 molecules <strong>of</strong> ATP for each molecule <strong>of</strong> glucose. The administration <strong>of</strong><br />

exogenous FDP improves heart function in a number <strong>of</strong> models <strong>of</strong> shock. This is the result<br />

<strong>of</strong> improved energy utilization, ATP sparing, improved enzyme activity as well as a number<br />

<strong>of</strong> other mechanisms.<br />

Pilot data from our laboratory has shown that IV administration <strong>of</strong> FDP to rodents<br />

poisoned with propranolol resulted in an improvement in blood pressure <strong>and</strong> survival. This<br />

was significantly better than that seen in control animals treated either with glucose or<br />

adrenaline infusion. We do not know whether co-administration <strong>of</strong> FDP with other<br />

inotropes such as adrenaline infusion or high-dose insulin infusion results in increased<br />

improvement in heart function. Also, co-administration <strong>of</strong> FDP with other agents may<br />

result in inotrope sparing, thereby reducing the doses <strong>of</strong> these agents required to improve<br />

shock. Positive results will help design subsequent clinical trials <strong>of</strong> FDP in poisoning.<br />

Project aims: The aim <strong>of</strong> this honours project is to assess the effects <strong>of</strong> FDP infusion on<br />

inotrope sparing, blood pressure, cardiac output <strong>and</strong> survival in a rodent model <strong>of</strong> severe<br />

propranolol poisoning.<br />

Techniques: Anaesthetised rodents will require surgery to be instrumented to record<br />

blood pressure, cardiac output <strong>and</strong> electrocardiographic trace. Intravenous canulae will be<br />

inserted to infuse propranolol <strong>and</strong> inotropic treatments. Monitoring <strong>and</strong> data collection<br />

using PowerLab hardware <strong>and</strong> s<strong>of</strong>tware.<br />

Contacts:<br />

Pr<strong>of</strong>essor Andis Graudins<br />

Southern Clinical School <strong>and</strong> Department <strong>of</strong> Pharmacology, Monash University<br />

Phone: 95943193, Rm E140; Andis.Graudins@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

EVALUATION OF WEB-BASED PHARMACOLOGY RESOURCES<br />

Supervisors: Dr Elizabeth Davis & Dr Eva Patak<br />

Location: Pharmacology Education Research Initative<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

Our group aims to gain a better underst<strong>and</strong>ing <strong>of</strong> factors that influence the engagement<br />

<strong>of</strong> students with their learning <strong>of</strong> pharmacology <strong>and</strong> the development <strong>of</strong> resources <strong>and</strong><br />

curricula to support students in their learning, particularly within the science <strong>and</strong> medical<br />

courses. This group is currently involved in several ongoing projects <strong>and</strong> prospective<br />

students could become involved in aspects <strong>of</strong> these.<br />

Project aim:<br />

The aim <strong>of</strong> one honours project proposal is to gain a greater underst<strong>and</strong>ing <strong>of</strong> the web<br />

resources that undergraduate science students use to supplement their learning <strong>of</strong><br />

pharmacology. Student use <strong>of</strong> information sources will be assessed <strong>and</strong> popular electronic<br />

resources will be evaluated for accuracy, reliability <strong>and</strong> consistency <strong>of</strong> information.<br />

Academic <strong>and</strong> student attitudes towards the use <strong>of</strong> web resources compared to<br />

instructor-provided lecture material <strong>and</strong> recommended textbooks will also be assessed.<br />

The results <strong>of</strong> the project will inform the future development <strong>of</strong> teaching resources.<br />

This project could also encompass the development <strong>of</strong> practical / tutorial material – in<br />

particular, how we can develop practical sessions that address learning outcomes <strong>and</strong><br />

maintain student engagement.<br />

Techniques:<br />

In addition to literature searching <strong>and</strong> reviews to identify what is known <strong>of</strong> student<br />

attitudes to learning, research techniques that will be used to complete this project, will<br />

include the design, development, implementation <strong>and</strong> analysis <strong>of</strong> academic/student surveys<br />

<strong>and</strong> focus group interviews along with monitoring <strong>of</strong> the use <strong>of</strong> lecture recordings <strong>and</strong><br />

Moodle sites.<br />

Contacts:<br />

Dr Elizabeth Davis<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 5755, Rm E123<br />

Elizabeth.Davis@monash.edu<br />

Dr Eva Patak<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 5783, Rm E117<br />

Eva.Patak@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

INVESTIGATING NOVEL ANTI-FIBROTIC THERAPIES<br />

Supervisors: Dr Chrishan Samuel & Dr Simon Royce<br />

Location: Fibrosis Laboratory (Rms E101/E102)<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

Fibrosis is defined as the hardening <strong>and</strong>/or scarring <strong>of</strong> various organs including the<br />

heart, kidney <strong>and</strong> lung; which usually arises from abnormal wound healing to tissue injury,<br />

resulting in an excessive deposition <strong>of</strong> extracellular matrix components, primarily<br />

collagen. The eventual replacement <strong>of</strong> normal tissue with scar tissue leads to organ<br />

stiffness <strong>and</strong> ultimately, organ failure. Despite a number <strong>of</strong> available treatments for<br />

patients with various heart/kidney/lung diseases, patients receiving these therapies still<br />

progress to end-stage organ failure due to the inability <strong>of</strong> these treatments to directly<br />

target the build-up <strong>of</strong> fibrosis. Hence, novel <strong>and</strong> more direct anti-fibrotic therapies are<br />

still required to be established.<br />

Aims:<br />

The Fibrosis Lab aims to identify novel anti-fibrotic therapies (relaxin, stem cells, trefoil<br />

factor 2; <strong>and</strong> combinations <strong>of</strong> these) that will more effectively prevent/reverse fibrosis<br />

progression. Additionally, by underst<strong>and</strong>ing the mechanisms <strong>of</strong> action <strong>of</strong> these potential<br />

therapies <strong>of</strong> future, we aim to delineate new targets that can be utilized to enhance their<br />

therapeutic potential <strong>and</strong> abrogation <strong>of</strong> organ scarring.<br />

<strong>Projects</strong>:<br />

1. Signal transduction studies (in models <strong>of</strong> heart / kidney / lung disease)<br />

2. Head-to-head <strong>and</strong> combination therapy efficacy studies<br />

3. The influence <strong>of</strong> ageing <strong>and</strong> gender on fibrosis<br />

4. Development <strong>of</strong> new approaches to target airway remodeling in asthma<br />

Techniques:<br />

Depending on the project involved, animal/cell culture models <strong>of</strong> (heart/kidney/lung<br />

disease), blood pressure <strong>and</strong> functional measurements, matrix biology, protein<br />

biochemistry, molecular biology <strong>and</strong>/or histological techniques will be utilized.<br />

Contacts:<br />

Dr Chrishan Samuel <strong>and</strong> Dr Simon Royce<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9902 0152 / 9905 0913<br />

chrishan.samuel@monash.edu<br />

simon.royce@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

CARDIO-PROTECTIVE EFFECTS OF INCRETIN HORMONES:<br />

POTENTIAL BEYOND GLYCAEMIC CONTROL?<br />

Supervisors: Dr Tracey Gaspari & Pr<strong>of</strong> Robert Widdop<br />

Location: Cardiovascular Integrative Group<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Collaborator: Dr Anthony Dear<br />

Deputy Director, Eastern Clinical Research Unit, Monash<br />

University<br />

Background:<br />

Our research focuses on underst<strong>and</strong>ing the pathophysiological processes involved in the<br />

development <strong>of</strong> cardiovascular disease, one <strong>of</strong> the major causes <strong>of</strong> illness <strong>and</strong> death in<br />

Australia <strong>and</strong> worldwide.<br />

Project aim:<br />

Two classes <strong>of</strong> diabetic drugs targeting the glucagon-like peptide-1 (GLP-1) pathway are<br />

used clinically to treat Type 2 Diabetes Mellitus. These are GLP-1 receptor agonists (e.g.,<br />

Liraglutide) <strong>and</strong> dipeptidyl peptidase-4 (DPP-4) inhibitors (prolong ½ life <strong>of</strong> endogenous<br />

GLP-1). In collaboration with Dr Dear we aim to have projects that investigate the<br />

vascular <strong>and</strong> cardio-protective effect <strong>of</strong> these classes <strong>of</strong> drugs in a number <strong>of</strong><br />

cardiovascular disease states, including atherosclerosis, vascular injury (neointimal<br />

hyperplasia) <strong>and</strong> hypertension.<br />

Techniques:<br />

<strong>Projects</strong> could incorporate a range <strong>of</strong> methodologies including animal dietary <strong>and</strong><br />

pharmacological treatments, surgical techniques for vascular injury models,<br />

immunohistochemistry, biochemical measures (western blot, markers <strong>of</strong> cellular<br />

proliferation <strong>and</strong> hypertrophy), histological <strong>and</strong> morphological analyses (such as fibrosis,<br />

intimal to medial measurements).<br />

Contacts:<br />

Dr Tracey Gaspari <strong>and</strong> Pr<strong>of</strong> Rob Widdop<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 4762, Rm E119<br />

Tracey.Gaspari@monash.edu<br />

Robert.Widdop@monash.edu<br />

Figure 1: Representative images <strong>of</strong> crosssectional<br />

lesion development in aortic arch <strong>of</strong><br />

ApoE KO mice. Liraglutide treatment shows<br />

decreased intima (I) development. Intima (I) <strong>and</strong><br />

media (M) areas are identified by dotted line.<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

MECHANISMS OF ANGIOTENSIN TYPE-2 RECEPTOR-<br />

MEDIATED NEUROPROTECTION DURING STROKE<br />

Supervisors: Dr Claudia McCarthy, Dr Antony Vinh & Pr<strong>of</strong> Rob Widdop<br />

Location: Integrated Cardiovascular Pharmacology Laboratory<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton.<br />

Background:<br />

Stroke is the third largest cause <strong>of</strong> death <strong>and</strong> the major cause <strong>of</strong> disability in<br />

industrialised countries. Every year, approximately 15-20 million people worldwide, will<br />

suffer a new or recurrent stroke. Despite this, relatively few therapies are available<br />

for the treatment <strong>of</strong> stroke.<br />

The renin angiotensin system (RAS) is a hormonal cascade which has multiple roles<br />

throughout the body, including the control <strong>of</strong> the cardiovascular system, renal <strong>and</strong><br />

adrenal function governing fluid <strong>and</strong> electrolyte balance <strong>and</strong> arterial pressure. We<br />

recently demonstrated that a particular subtype <strong>of</strong> angiotensin receptor, the<br />

angiotensin II type 2 receptor (AT2 R), is neuroprotective during stroke <strong>and</strong> has been<br />

associated with anti-inflammatory effects nerve regeneration following nerve injury.<br />

Project aims: This study will elucidate mechanics <strong>of</strong> AT2R mediated neuroprotection<br />

specifically looking at its interaction with the immune system in determining stroke<br />

outcome.<br />

Techniques: An animal model <strong>of</strong> stroke will be implemented in conjunction with<br />

behavioural tests to assess stroke severity <strong>and</strong> any symptomatic relief resulting from<br />

treatment. Several physiological parameters will be monitored throughout the<br />

experimental protocol, including blood pressure. Specific organs will be isolated <strong>and</strong><br />

preserved to measure stroke-induced changes using histology. In addition, several<br />

techniques, including immunohistochemistry <strong>and</strong> flow cytometry, will identify the<br />

specific neuronal <strong>and</strong> immune cells associated with stroke <strong>and</strong> AT2R function.<br />

Contacts:<br />

Dr Claudia McCarthy <strong>and</strong> Dr Antony Vinh<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 0095, Rm E119<br />

Claudia.mccarthy@monash.edu<br />

Antony.vinh@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

ROLE OF T CELLS DURING THE DEVELOPMENT OF HIGH<br />

BLOOD PRESSURE<br />

Supervisors: Dr Antony Vinh & Pr<strong>of</strong> Rob Widdop<br />

Location: Integrative Cardiovascular Pharmacology<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

Inflammation <strong>and</strong> immunity continues to be a topical area <strong>of</strong> hypertension research. The<br />

adaptive immune system <strong>and</strong> particularly T cells have recently been implicated in the<br />

development <strong>of</strong> hypertension. Interestingly, there are several similarities between<br />

hypertension <strong>and</strong> autoimmune disorders, <strong>and</strong> <strong>of</strong> particular interest is local infiltration<br />

<strong>of</strong> T cells. Like synovial joints <strong>of</strong> arthritic patients, during experimental hypertension<br />

there is significantly increased T cell infiltration into non-lymphoid organs such as the<br />

vasculature, brain <strong>and</strong> kidneys, which are vital organs that regulate blood pressure.<br />

While it is hypothesized that these tissue-infiltrating T cells promote local<br />

inflammation, their precise function remains speculative <strong>and</strong> undefined. Moreover, the<br />

timing <strong>of</strong> the immune response during hypertension is unstudied.<br />

Project aims:<br />

The aim <strong>of</strong> this honours project is to (1) investigate the function <strong>of</strong> infiltrating T cells<br />

during hypertension, <strong>and</strong> through a series time-course experiments, (2) pinpoint the<br />

sequence <strong>of</strong> immune-related events leading to activation <strong>of</strong> the immune system during<br />

hypertension. This study will identify when <strong>and</strong> how T cells <strong>and</strong> the immune system<br />

contribute to elevating blood pressure, <strong>and</strong> may lead to novel drug targets to treat this<br />

common disorder.<br />

Techniques:<br />

It is anticipated that this will involve the isolation <strong>and</strong> sorting (magnetic cell isolation<br />

kits <strong>and</strong> flow cytometry) immune cells from various organs from animal models <strong>of</strong><br />

hypertension. In addition, assays to detect T cell derived-cytokines/chemokines<br />

(cytometric bead array) <strong>and</strong> reactive oxygen species generation (chemiluminescence)<br />

will also be utilized.<br />

Contacts:<br />

Dr Antony Vinh <strong>and</strong> Pr<strong>of</strong> Rob Widdop<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9902 4844, Rm E33<br />

Antony.Vinh@monash.edu<br />

Robert.Widdop@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

MODULATION OF NEURONAL OXYTOCIN – EFFECTS ON<br />

SOCIAL BEHAVIOUR<br />

Supervisors: Dr Richard Loiacono & Dr Siew Yeen Chai<br />

Location: Neuropharmacology Lab & Neurophysiology Team<br />

Departments <strong>of</strong> Pharmacology & Physiology<br />

Monash University, Clayton<br />

Background:<br />

Metallo-peptidases cleave amino acids from either the N- <strong>and</strong> C-termini <strong>of</strong> peptide<br />

hormones to either generate or degrade biologically active peptides. These enzymes play<br />

important roles in the body <strong>and</strong> alterations in their activities can impact on a diverse<br />

range <strong>of</strong> physiological processes in both healthy <strong>and</strong> diseased states. This project is<br />

focussed on one such enzyme known as insulin-regulated aminopeptidase (IRAP) or<br />

oxytocinase. Oxytocin is known as the social hormone, regulating complex social<br />

behaviours including promoting trust, pair-bonding <strong>and</strong> has been explored as potential<br />

therapy for social behaviour impairments observed with autism spectrum disorders.<br />

Aim:<br />

This project will investigate the behavioural phenotypes in the oxytocinase deficient<br />

mice to determine if regulation <strong>of</strong> endogenous oxytocin levels will affect social<br />

behaviour.<br />

Techniques:<br />

Behavioural testing, cell culture, immunohistochemistry<br />

Contacts:<br />

Dr Richard Loiacono & Dr Siew Yeen Chai<br />

Departments <strong>of</strong> Pharmacology & Physiology<br />

Monash University<br />

Phone: Richard 9905 4859 Siew Yeen 9905 2515<br />

richard.loiacono@monash.edu<br />

siew.chai@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

ARE THE ADVERSE EFFECTS OF HORMONE REPLACEMENT<br />

THERAPY ON STROKE OUTCOME MEDIATED BY G PROTEIN-<br />

COUPLED ESTROGEN RECEPTOR SIGNALLING?<br />

Supervisors: Dr Brad Broughton & A/Pr<strong>of</strong> Chris Sobey<br />

Location: Vascular Biology & Immunopharmacology Group<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

Stroke is a debilitating disease that can cause permanent neurological damage,<br />

complications, <strong>and</strong> death. At present, there are very few treatment options available<br />

for patients, thus the development <strong>of</strong> new treatments is vital to reduce the damage<br />

caused by stroke. In recent years, growing evidence has revealed that estrogen is<br />

neuroprotective against stroke, but confusingly hormone replacement therapy (HRT)<br />

worsens stroke outcome in post-menopausal women. Interestingly, recent work from our<br />

lab has found that activation <strong>of</strong> a novel estrogen receptor (G Protein-coupled Estrogen<br />

Receptor, GPER), which is widely distributed throughout the brain, increases cerebral<br />

infarct damage in males following stroke. Therefore, we hypothesise that GPER<br />

activity affects outcome <strong>of</strong> HRT therapy in females after stroke.<br />

Project aim:<br />

The aim <strong>of</strong> this project is to investigate the importance <strong>of</strong> GPER in mediating the<br />

adverse effects <strong>of</strong> HRT in stroke. The outcomes <strong>of</strong> the proposed project are<br />

expected to confirm that estrogen therapy has an adverse effect on stroke<br />

outcome in young ovariectomised mice due to activation <strong>of</strong> GPER signalling.<br />

Techniques:<br />

Techniques that will be used during this project include treating mice with estrogen <strong>and</strong><br />

various GPER lig<strong>and</strong>s, histochemical approaches to measure cerebral infarct damage,<br />

immunohistochemistry to examine the distribution <strong>of</strong> GPER in the brain <strong>and</strong> Western<br />

blotting to determine GPER expression levels.<br />

Contacts:<br />

Dr Brad Broughton & A/Pr<strong>of</strong> Chris Sobey<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 0915, Rm EG20<br />

bradley.broughton@monash.edu<br />

chris.sobey@monash.edu<br />

Vehicle Estradiol<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

STEM CELL THERAPY FOR THE TREATMENT OF HIGH BLOOD<br />

PRESSURE<br />

Supervisors: A/Pr<strong>of</strong> Grant Drummond & Dr Rebecca Lim<br />

Location: Vascular Biology & Immunopharmacology Group<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

Hypertension (high blood pressure) is a major cause <strong>of</strong> heart failure, heart attacks <strong>and</strong><br />

strokes. Over 2 million people in Australia suffer from hypertension <strong>and</strong> alarmingly, in up<br />

to 30% <strong>of</strong> these individuals, the condition is not controlled by current blood pressure<br />

medications. A recent development in the field <strong>of</strong> hypertension research is the recognition<br />

that the disease is caused by an influx <strong>of</strong> immune cells (i.e. T cells <strong>and</strong> macrophages) into<br />

the artery wall. Activation <strong>of</strong> these immune cells leads to the release <strong>of</strong> free radicals <strong>and</strong><br />

cytokines that promote an inflammatory response, causing the arteries to become ‘stiff’<br />

<strong>and</strong> constricted <strong>and</strong> exacerbating elevations in blood pressure. Hence, strategies that<br />

reduce inflammation in the vascular wall hold promise as future treatments for<br />

hypertension.<br />

Project aims:<br />

Stem cells have long been touted as possible cures for degenerative diseases due to their<br />

tissue regenerative properties. However, stem cells also display powerful antiinflammatory<br />

properties suggesting that they may also have therapeutic potential in<br />

inflammatory conditions such as hypertension. The human amniotic membrane is a readily<br />

available tissue (discarded after childbirth) <strong>and</strong> a rich source <strong>of</strong> stem cells. The aim <strong>of</strong><br />

this project is to examine whether human amnion stem cells prevent hypertension in<br />

mice by exerting anti-inflammatory effects in the artery wall <strong>and</strong> in other organs<br />

that regulate blood pressure, such as the kidney <strong>and</strong> brain.<br />

Techniques:<br />

This project will involve surgeries on mice to induce hypertension, in vivo blood pressure<br />

monitoring, <strong>and</strong> the use <strong>of</strong> assays to measure immune cell numbers (flow cytometry,<br />

immunohistochemistry), <strong>and</strong> expression <strong>of</strong> inflammatory mediators (real-time PCR, ELISA,<br />

Western blotting) <strong>and</strong> free radicals (luminescence) in blood <strong>and</strong> tissues.<br />

Contact:<br />

A/Pr<strong>of</strong> Grant Drummond<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 4869, Rm E112<br />

Grant.Drummond@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

EXPLORING THE VASOPROTECTIVE ACTIONS OF<br />

NOVEL HNO DONORS<br />

Supervisors: Dr Barbara Kemp-Harper<br />

Location: Vascular Biology & Immunopharmacology Group<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

Nitric oxide (NO ), is an important endogenous vasodilator <strong>and</strong> regulator <strong>of</strong> vascular tone.<br />

In disease states such as hypertension <strong>and</strong> atherosclerosis, the NO signaling pathway is<br />

impaired leading to reduced blood flow to vital organs such as the brain <strong>and</strong> heart. Whilst<br />

nitrovasodilators can be used to overcome dysfunctional NO signaling, the clinical<br />

application <strong>of</strong> these agents is limited due to their susceptibility to scavenging by<br />

superoxide <strong>and</strong> tolerance development.<br />

Importantly, NO can also exist in the reduced state as nitroxyl (HNO) <strong>and</strong> recent<br />

evidence suggests that this nitrogen oxide is also generated endogenously <strong>and</strong> has distinct<br />

pharmacological properties <strong>and</strong> therapeutic advantages over NO. Thus HNO is not<br />

scavenged by superoxide, does not develop tolerance <strong>and</strong> can target distinct signaling<br />

pathways to cause vasorelaxation. As such, the bioavailability <strong>of</strong> HNO is preserved in<br />

disease. With a suite <strong>of</strong> vasoprotective actions, HNO donors have therapeutic potential in<br />

the treatment <strong>of</strong> cardiovascular disease. <strong>Current</strong>ly, however the clinical utility <strong>of</strong> these<br />

donors is limited by their short-half lives byproducts. Excitingly, new pure HNO donors<br />

with longer half-lives are being developed <strong>and</strong> this study will elucidate the therapeutic<br />

potential <strong>of</strong> these drugs in the setting <strong>of</strong> atherosclerosis.<br />

Project aim:<br />

The aim <strong>of</strong> this honours project is to investigate the ability <strong>of</strong> novel HNO donors to serve<br />

as vasoprotective agents in the setting <strong>of</strong> atherosclerosis.<br />

Techniques:<br />

It is anticipated that the project will involve the use <strong>of</strong> techniques to assess the<br />

vasodilator (small vessel myography), anti-aggregatory <strong>and</strong> superoxide suppressing<br />

(chemiluminescence) actions <strong>of</strong> novel HNO donors in control <strong>and</strong> atherosclerotic mice.<br />

Contacts:<br />

Dr Barbara Kemp-Harper<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 4674, Rm E140<br />

Barbara.Kemp@monash.edu<br />

Proliferation<br />

Platelet<br />

Aggregation<br />

HNO VSMC<br />

Relaxation<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.<br />

. -<br />

O<br />

2<br />

.<br />

O 2<br />

-<br />

. -<br />

O<br />

2<br />

Superoxide<br />

Production


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

EXPLORING NOVEL THERAPEUTIC STRATEGIES TO<br />

PREVENT ATHEROSCLEROTIC PLAQUE RUPTURE<br />

Supervisors: Dr Barbara Kemp-Harper & A/Pr<strong>of</strong> Grant Drummond<br />

Location: Vascular Biology & Immunopharmacology Group<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

Atherosclerosis is characterised by the formation <strong>of</strong> lipid filled lesions in the arterial<br />

wall. These plaques are relatively harmless until they become unstable <strong>and</strong> rupture leading<br />

to thrombus formation <strong>and</strong> vessel occlusion resulting in myocardial infarction <strong>and</strong> stroke.<br />

Macrophages, in particular, play a central role in plaque rupture secreting inflammatory<br />

molecules, reactive oxygen species (ROS) <strong>and</strong> metalloproteinases. As such, new therapies<br />

are sought which reduce plaque macrophage content <strong>and</strong> activation. We have evidence to<br />

suggest that nitroxyl (HNO), a novel redox form <strong>of</strong> nitric oxide, targets Nox2 oxidase to<br />

limit vascular <strong>and</strong> inflammatory cells ROS generation <strong>and</strong> may stabilize atherosclerotic<br />

plaques.<br />

Project aim:<br />

The aim <strong>of</strong> this honours project is to investigate the ability <strong>of</strong> HNO <strong>and</strong> NADPH oxidase<br />

inhibitors to serve as protective agents against atherosclerotic plaque rupture. This study<br />

will elucidate the potential vasoprotective effects <strong>of</strong> HNO <strong>and</strong> NAPDH oxidase inhibitors<br />

<strong>and</strong> may lead to the development <strong>of</strong> more effective therapies for the treatment <strong>of</strong><br />

atherosclerosis.<br />

Techniques:<br />

It is anticipated that this will involve the use <strong>of</strong> assays to detect inflammation (RT-PCR,<br />

western blotting, immunohistochemistry), reactive oxgen species generation<br />

(chemiluminescence), vascular dysfunction (small vessel myography) <strong>and</strong> plaque formation<br />

<strong>and</strong> stability in atherosclerotic mice.<br />

Contacts:<br />

Dr Barbara Kemp-Harper <strong>and</strong> A/Pr<strong>of</strong> Grant Drummond<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 4674, Rm E140<br />

Barbara.Kemp@monash.edu<br />

Grant.Drummond@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.<br />

norm<br />

al<br />

arter<br />

mid-stage<br />

atherosclero<br />

sis<br />

late-stage<br />

atherosclero<br />

sis


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

EXPLORING THE ROLE OF ENDOTHELIAL NF-κB ON ISCHEMIC<br />

STROKE OUTCOME<br />

Supervisors: Dr Helena Kim & A/Pr<strong>of</strong> Chris Sobey<br />

Location: Vascular Biology & Immunopharmacology Group<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

Stroke is the second leading cause <strong>of</strong> death worldwide <strong>and</strong> is caused by an interruption <strong>of</strong><br />

cerebral blood flow by a blockage (ischemia) or a rupture (haemorrhage) in the blood<br />

vessel. Cerebral ischemia triggers a cascade <strong>of</strong> complex events including excitotoxicity,<br />

production <strong>of</strong> reactive oxygen species, up-regulation <strong>of</strong> pro-inflammatory molecules <strong>and</strong><br />

transcription factors that lead to apoptosis. Inflammatory mechanisms have come into the<br />

focus <strong>of</strong> stroke research because they contribute substantially to secondary brain damage.<br />

Nuclear factor kappa B (NF-κB) is a transcription factor that modulates gene expression<br />

<strong>of</strong> a range <strong>of</strong> cytokines <strong>and</strong> other pro-inflammatory molecules. Recent findings suggest<br />

that inhibition <strong>of</strong> NF-κB, especially those expressed in neurons, provide neuroprotection<br />

following stroke.<br />

Project aim:<br />

The aim <strong>of</strong> this honours project is to investigate the role <strong>of</strong> endothelial NF-κB on stroke<br />

outcome using mice overexpressing NF-κB inhibitor, IκBα. This study will elucidate the<br />

potential effect <strong>of</strong> NF-κB inhibition <strong>and</strong> blocking <strong>of</strong> further inflammatory response<br />

following stroke <strong>and</strong> may lead to the development <strong>of</strong> more effective therapies for the<br />

treatment <strong>of</strong> stroke.<br />

Techniques:<br />

This project will involve the use <strong>of</strong> assays to measure behavioural deficits (hanging wire<br />

test, neurological score), brain infarct size (thionin stain, Image J s<strong>of</strong>tware) <strong>and</strong> immune<br />

responses (immunohistochemistry) in IκBα-overexpressing mice following stroke.<br />

Contacts:<br />

Dr Helena Kim <strong>and</strong> A/Pr<strong>of</strong> Chris Sobey<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 1146, Rm E139<br />

Helena.Kim@monash.edu<br />

Chris.Sobey@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

INVESTIGATING MECHANISMS LINKING HYPERTENSION AND<br />

COGNITIVE IMPAIRMENT<br />

Supervisors: Dr Alyson Miller<br />

Location: Vascular Biology & Immunopharmacology Group<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

As the population <strong>of</strong> the world ages, the incidence <strong>of</strong> cognitive impairment <strong>and</strong> dementias<br />

is expected to rise exponentially. In fact, it is predicted that the number <strong>of</strong> dementia<br />

patients worldwide will quadruple over the next three decades to reach approximately 81<br />

million by 2040. Although Alzheimer disease (AD) is the most commonly diagnosed cause<br />

<strong>of</strong> cognitive dysfunction among the aged, cognitive impairment caused by cardiovascular<br />

diseases are also important independent causes <strong>and</strong> contributors to cognitive dysfunction.<br />

For example, there is growing evidence hypertension during mid-life increases the risk for<br />

cognitive impairment later in life. Despite this evidence, the mechanistic interactions<br />

between hypertension <strong>and</strong> cognitive dysfunction are still an enigma.<br />

Project aims:<br />

The aims <strong>of</strong> this <strong>Honours</strong> project are to firstly verify that hypertension causes cognitive<br />

dysfunction. Secondly, this project will explore potential pathways <strong>and</strong> mediators that<br />

mechanistically link hypertension <strong>and</strong> cognitive impairment, with a particular focus on<br />

oxidative <strong>and</strong> inflammatory processes.<br />

Techniques:<br />

Techniques that will be used during this project include behavioural tests to assess<br />

cognitive function, small vessel myography to assess vascular function, chemiluminescence<br />

based assays to assess oxidative stress, <strong>and</strong> molecular approaches <strong>and</strong> FACS analysis to<br />

assess inflammation.<br />

Contact:<br />

Dr Alyson Miller<br />

Monash University<br />

Phone: 9905 3817<br />

Alyson.Miller@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

NOVEL PHARMACOLOGICAL TARGETS FOR INFLUENZA A VIRUS<br />

INDUCED LUNG INFLAMMATION<br />

Supervisors: Dr Stavros Selemidis<br />

Dr Ross Vlahos<br />

Location: Department <strong>of</strong> Pharmacology, Monash University, Clayton <strong>and</strong><br />

Department <strong>of</strong> Pharmacology, Melbourne University.<br />

Background:<br />

<strong>Current</strong> drug therapies to treat influenza pathology are focused primarily on halting<br />

mechanisms <strong>of</strong> viral infection <strong>and</strong> replication. Far less attention has been directed to<br />

identifying immunopathological mechanisms <strong>of</strong> host defence that lead to the acute lung<br />

injury. Animal <strong>and</strong> human studies provide compelling evidence that the acute lung injury<br />

following influenza A virus infection is caused by reactive oxygen species (ROS)<br />

production such as superoxide anion. Importantly, the primary sites <strong>of</strong> influenza A virus<br />

infection, namely airway epithelial cells, <strong>and</strong> resident <strong>and</strong> infiltrating macrophages are key<br />

sites <strong>of</strong> ROS production via Nox1- <strong>and</strong> Nox2-containing NADPH oxidases, respectively.<br />

Our preliminary studies have identified the Nox1 <strong>and</strong> Nox2 enzymes as novel targets for<br />

modulating pathology irrespective <strong>of</strong> the infecting influenza strain.<br />

Project aims: The aim <strong>of</strong> this honours project is to investigate the roles <strong>of</strong> NADPH<br />

oxidases in influenza A virus induced lung injury <strong>and</strong> inflammation. To achieve this a<br />

multidisciplinary honours project has been devised. It is anticipated that these studies<br />

will have a major impact on modern drug discovery focusing on oxidative stress caused by<br />

influenza A viruses.<br />

Techniques:<br />

It is anticipated that this will involve the use <strong>of</strong> cell culture <strong>and</strong> in vivo animal models; <strong>and</strong><br />

assays to detect NADPH oxidase expression <strong>and</strong> localization (western blotting,<br />

immunohistochemistry), reactive oxygen species generation (chemiluminescence) <strong>and</strong><br />

inflammatory cell characterization (flow cytometry).<br />

Contacts:<br />

Dr Stavros Selemidis<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 5756, Rm E137<br />

Stavros.selemidis@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

UNDERSTANDING THE BIOLOGY OF REACTIVE OXYGEN<br />

SPECIES IN CANCER<br />

Supervisors: Dr Stavros Selemidis<br />

Location: Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

The socio-economic burden <strong>of</strong> prostate cancer is unquestionable claiming many lives<br />

worldwide. Angiogenesis is necessary for tumours to grow <strong>and</strong> metastasise, <strong>and</strong> much<br />

effort has been directed towards identification <strong>of</strong> the genetic <strong>and</strong> molecular mechanisms<br />

that lead to angiogenesis <strong>and</strong> in the development <strong>of</strong> pharmacological agents to halt<br />

angiogenesis. It has long been known that tumour <strong>and</strong> endothelial cell proliferation <strong>and</strong><br />

survival are crucial for a sustained angiogenic process. Emerging evidence implicates<br />

reactive oxygen species (ROS) such as superoxide <strong>and</strong> hydrogen peroxide (H2O2) to play a<br />

fundamental role in facilitating angiogenesis. The primary sources <strong>of</strong> ROS are the Nox2-<br />

<strong>and</strong> Nox4-containing NADPH oxidases.<br />

Project aims: The aim <strong>of</strong> this honours project is to investigate the roles <strong>of</strong> NADPH<br />

oxidases in prostate tumour-associated angiogenesis. The findings from this study may<br />

provide a new underst<strong>and</strong>ing <strong>of</strong> the molecular mechanisms that underpin angiogenesis, <strong>and</strong><br />

a rationale for the combined suppression <strong>of</strong> NADPH oxidases to inhibit angiogenesis in<br />

prostate cancer.<br />

Techniques:<br />

It is anticipated that this will involve the use <strong>of</strong> cell culture <strong>and</strong> in vivo animal models; <strong>and</strong><br />

assays to detect NADPH oxidase expression <strong>and</strong> localization (western blotting,<br />

immunohistochemistry), reactive oxygen species generation (chemiluminescence) <strong>and</strong><br />

immune cell characterization (flow cytometry).<br />

Contacts:<br />

Dr Stavros Selemidis<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 5756, Rm E137<br />

Stavros.selemidis@monash.edu<br />

Figure: Preliminary data from our laboratory<br />

demonstrating a significant reduction in<br />

prostate tumour development in a NADPH<br />

oxidase-deficient mouse (i.e Nox2 is<strong>of</strong>orm)<br />

compared to a WT control mouse.<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

VENOMS AND TOXINS PROJECTS<br />

Supervisors: Pr<strong>of</strong>essor Wayne Hodgson<br />

Location: Monash Venom Group<br />

Department <strong>of</strong> Pharmacology<br />

Monash University, Clayton<br />

Background:<br />

The Monash Venom Group investigates the toxinology <strong>of</strong> venomous snakes, spiders <strong>and</strong><br />

sea creatures.<br />

<strong>Projects</strong>:<br />

1. Molecular toxinology <strong>of</strong> Australia’s lesser known venomous snakes<br />

2. Pr<strong>of</strong>iling Australian <strong>and</strong> Malaysian snake venoms to guide treatment strategies<br />

Techniques:<br />

Venom fractionation, HPLC, organ bath experimentation, blood coagulation assays,<br />

myotoxicity assays<br />

Contacts:<br />

Pr<strong>of</strong>essor Wayne Hodgson<br />

Department <strong>of</strong> Pharmacology<br />

Monash University<br />

Phone: 9905 4861<br />

Wayne.Hodgson@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

DEFINING THE FUNCTION OF THE HUMAN PLATELET<br />

THROMBIN RECEPTOR, PAR4, IN ARTERIAL THROMBOSIS<br />

Supervisors: Dr Justin Hamilton & A/Pr<strong>of</strong> Grant Drummond<br />

Location: Australian Centre for Blood Diseases<br />

Alfred Medical Research & Education Precinct<br />

Alfred Hospital, Prahran<br />

Background:<br />

Arterial thrombosis causes heart attack <strong>and</strong> stroke is the leading cause <strong>of</strong> death <strong>and</strong><br />

disability in most countries. Platelets are the blood cells responsible for forming arterial<br />

thrombi, <strong>and</strong> new drugs which block this function <strong>of</strong> platelets are sought for effective<br />

antithrombotic therapy. We have shown that thrombin activates platelets via proteaseactivated<br />

receptors (PARs) <strong>and</strong> that PAR-deficient mice are protected against thrombosis<br />

but do not exhibit spontaneous bleeding (see Figure). Our work has led to the<br />

development <strong>of</strong> PAR antagonists – two <strong>of</strong> which are currently in Phase 3 clinical trials for<br />

the prevention <strong>of</strong> arterial thrombosis. However, there are two thrombin receptors on<br />

human platelets, PAR1 <strong>and</strong> PAR4, <strong>and</strong> current antagonists target only PAR1. Indeed, little<br />

is known about PAR4. To address this, we have recently developed the first specific PAR4<br />

antagonist which will allow us for the first time to determine the role <strong>of</strong> PAR4 in human<br />

platelet function.<br />

Project aims:<br />

This project will examine the contribution <strong>of</strong> PAR4 to human platelet activation <strong>and</strong><br />

thrombus formation <strong>and</strong> will help determine whether PAR4 is a suitable c<strong>and</strong>idate for the<br />

development <strong>of</strong> anti-thrombotic therapies.<br />

Techniques:<br />

The project will utilise many useful techniques including functional experiments on isolated<br />

platelets, ex vivo whole blood thrombosis experiments, confocal microscopy, <strong>and</strong> in vivo<br />

mouse models <strong>of</strong> thrombosis <strong>and</strong> haemostasis, <strong>and</strong> will appeal to students interested in<br />

researching <strong>and</strong> developing future therapies for heart attack <strong>and</strong> stroke.<br />

Contact:<br />

Dr Justin Hamilton<br />

Australian Centre for Blood<br />

Diseases<br />

Monash University<br />

Phone: 9903 0125<br />

Justin.Hamilton@monash.edu<br />

In vivo thrombosis: Shown are platelets (red) in<br />

arterioles <strong>of</strong> control or PAR4-deficient mice at various<br />

times after laser-induced vascular injury. Note the<br />

limited growth <strong>of</strong> the thrombus in PAR4-deficient mice.<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

WHAT TURNS YOU ON? USING A NOVEL RNAi-BASED<br />

APPROACH TO DETERMINE PLATELET ACTIVATION SIGNALS<br />

IN THROMBOSIS<br />

Supervisors: Dr Justin Hamilton & A/Pr<strong>of</strong> Grant Drummond<br />

Location: Australian Centre for Blood Diseases<br />

Alfred Medical Research & Education Precinct<br />

Alfred Hospital, Prahran<br />

Background:<br />

Thrombosis is the formation <strong>of</strong> blood clots leading to heart attack <strong>and</strong> stroke. Platelets<br />

are the blood cells which form these clots, but they must be activated before they will do<br />

so. Therefore we are interested in determining what controls the activation <strong>of</strong> platelets<br />

during blood clot formation. We have recently established a novel transgenic strategy for<br />

inducible knockdown <strong>of</strong> genes <strong>of</strong> interest in mouse platelets in vivo. We are now using this<br />

approach to perform the first studies on the intracellular signalling enzyme, PI3K-C2α, in<br />

platelet function. We have shown that PI3K-C2α-deficient mice have dysregulated<br />

platelet function in vitro <strong>and</strong> in vivo (e.g. see figure).<br />

Project aims:<br />

This project will use our novel RNAi-based mouse genetic model to further define the<br />

contribution <strong>of</strong> PI3K-C2α to platelet function to determine whether inhibition <strong>of</strong> PI3K-<br />

C2α is an effective strategy for the prevention <strong>of</strong> arterial thrombosis.<br />

Techniques:<br />

The project will use a range <strong>of</strong> techniques, including a novel mouse genetic approach,<br />

molecular biology (RT-PCR, Western blotting, etc), as well as basic techniques in blood<br />

h<strong>and</strong>ling <strong>and</strong> platelet preparation <strong>and</strong> manipulation <strong>and</strong> will suit students interested in<br />

gaining experience in a variety <strong>of</strong> laboratory techniques <strong>and</strong> those interested in<br />

researching a potential new drug target for the prevention <strong>of</strong> heart attack <strong>and</strong> stroke.<br />

Contact:<br />

Dr Justin Hamilton<br />

Australian Centre for Blood Diseases<br />

Monash University<br />

Phone: 9903 0125<br />

Justin.Hamilton@monash.edu<br />

PI3K-C2α-deficient mice do not clot as<br />

efficiently as control mice. The time taken<br />

for mice to stop bleeding following a defined<br />

wound cut into their tail was recorded.<br />

Wild-type PI3K-C2α<br />

mutant<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

THE ROLE OF AT2R IN MACROPHAGE POLARIZATION<br />

Supervisors: Pr<strong>of</strong> Jaye Chin-Dusting, Dr Jennifer Irvine &<br />

Pr<strong>of</strong> Rob Widdop<br />

Location: Vascular Pharmacology<br />

Baker IDI Heart & Diabetes Institute<br />

Prahran<br />

Background:<br />

Atherosclerosis has been identified as an underlying cause <strong>of</strong> cardiovascular disease<br />

(CVD). Vascular inflammation, a critical early event in the development <strong>of</strong> atherosclerosis,<br />

involves the recruitment <strong>and</strong> adhesion <strong>of</strong> monocytes to the endothelium. The monocytes<br />

then transmigrate through the vessel wall into tissue, where they then differentiate into<br />

macrophages. Under physiological conditions, the monocyte-derived macrophage is <strong>of</strong> the<br />

M2 type playing a normal protective role in the organism. When pathological factors<br />

(including inflammation) are present, the differentiated macrophage possesses features<br />

which damage tissue, <strong>and</strong> this type <strong>of</strong> macrophage is normally termed M1. M1 macrophages<br />

play an important role in CVD.<br />

The Renin-Angiotensin System (RAS) regulates cardiovascular homeostasis largely via<br />

activation <strong>of</strong> the angiotensin type 1 receptor (AT1R), which exhibits, amongst others, proinflammatory<br />

effects. However, despite widespread use <strong>of</strong> AT1R blockers in the<br />

treatment <strong>of</strong> CVD diseases such as hypertension, many patients still exhibit endothelial<br />

dysfunction <strong>and</strong> remain at risk <strong>of</strong> death from complications such as heart attack <strong>and</strong><br />

stroke. Less well established, the angiotensin type 2 receptor (AT2R) <strong>and</strong> the Mas<br />

receptor (MasR) subtypes are thought to counter-regulate the effects <strong>of</strong> the AT1R.<br />

Project aims:<br />

This project aims to explore the potential effects <strong>of</strong> direct AT2R activation on<br />

macrophage polarization. We will isolate monocytes from human blood, differentiate them<br />

into macrophages, <strong>and</strong> induce the macrophages into M1 or M2 states respectively. We will<br />

then study the influence <strong>of</strong> AT2R activation on the induction <strong>of</strong> M1 or M2 macrophages<br />

<strong>and</strong> AT2R-induced alteration <strong>of</strong> macrophage polarization.<br />

Techniques:<br />

This project involves a range <strong>of</strong> technologies <strong>of</strong> cellular <strong>and</strong> molecular biology, such as cell<br />

culture, flow cytometry, real-time PCR, western blot <strong>and</strong> cellular imaging etc.<br />

Contacts:<br />

Pr<strong>of</strong> Jaye Chin-Dusting & Dr Jennifer Irvine<br />

Vascular Pharmacology<br />

Baker IDI Heart & Diabetes Institute<br />

jaye.chin-dusting@bakeridi.edu.au, jennifer.irvine@bakeridi.edu.au<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

EFFECTS OF POSITIVE ALLOSTERIC MODULATOR OF GABAA<br />

RECEPTORS ON HYPERTENSION AND STRESS<br />

Supervisors: Pr<strong>of</strong>essor Ge<strong>of</strong>frey A. Head, Dr Pamela Davern<br />

Location: Neuropharmacology Laboratory<br />

BakerIDI Heart & Diabetes Institute<br />

Prahran<br />

Background:<br />

Allopregnanolone (AlloP), a neurosteroid produced in the brain is a potent endogenous<br />

positive allosteric modulator <strong>of</strong> GABA action at GABAA receptors. Increased NPY Y1<br />

receptor gene expression in the amygdala is a major mechanism <strong>of</strong> AlloP anxiolytic<br />

effects. Systemic administration <strong>of</strong> AlloP or drugs that reverse the reduction <strong>of</strong> AlloP (eg.<br />

Fluoxetine) in the brain induced by social isolation, reduce anxiety <strong>and</strong> aggressive<br />

behavioural abnormalities. However, the cardiovascular consequences <strong>of</strong> the direct actions<br />

on stress have been largely overlooked. We suggest that Schlager hypertensive mice may<br />

be deficient in production <strong>of</strong> AlloP leading to the dysfunction <strong>of</strong> the GABAA receptor <strong>and</strong><br />

that this can be reversed by AlloP administration. This will determine whether the defect<br />

is reversible or not. The latter would suggest the genetic differences are a primary<br />

deficit rather than induced by a lifetime <strong>of</strong> heightened stress<br />

Project aims:<br />

The aim is to determine the effect <strong>of</strong> chronic systemic administration <strong>of</strong> AlloP on blood<br />

pressure <strong>and</strong> response to stress in BPH <strong>and</strong> BPN mice.<br />

Techniques:<br />

The project involves radiotelemetry measurement <strong>of</strong> blood pressure in conscious mice,<br />

behavioural testing using a number <strong>of</strong> mild stressors such as restraint <strong>and</strong><br />

immunohistochemistry to examine thec ahnges in activation <strong>of</strong> neuropeptide Y <strong>and</strong> GABA<br />

neurons in the amygdala <strong>and</strong> hypothalamus.<br />

Contacts:<br />

Pr<strong>of</strong>. Ge<strong>of</strong>frey Head <strong>and</strong> Dr Pamela Davern<br />

Department <strong>of</strong> Pharmacology<br />

Baker IDI Heart & Diabetes Institute<br />

Phone: 03 8532 1332<br />

Ge<strong>of</strong>f.head@bakeridi.edu.au<br />

Pamela.Davern@bakeridi.edu.au<br />

In BPH hypertensive mice many neurons expressing GABAA<br />

receptors (orange) were also immunoreactive for NPY (red) <strong>and</strong><br />

each <strong>of</strong> these were triple labeled with Fos (yellow) in the<br />

paraventricular nucleus (PVN) <strong>and</strong> medial amygdala (MeAm).<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

ROLE OF BRAIN PATHWAYS IN CHRONIC STRESS<br />

Supervisors: Pr<strong>of</strong>essor Ge<strong>of</strong>frey A. Head, Dr Pamela Davern<br />

& S<strong>and</strong>ra Burke<br />

Location: Neuropharmacology Laboratory<br />

BakerIDI Heart & Diabetes Institute<br />

Prahran<br />

Background:<br />

There is now strong evidence to suggest that the sympathetic nervous system (SNS)<br />

makes a significant contribution to certain forms <strong>of</strong> hypertension. One <strong>of</strong> the suggested<br />

mechanisms through which the SNS may be “activated” in hypertension is through the<br />

renin‐angiotensin system. We have recently demonstrated that a very low “subpressor”<br />

dose <strong>of</strong> Angiotensin given for several months to rabbits can produce a very modest<br />

increase in blood pressure but appears to amplify the effects <strong>of</strong> chronic stress. Using<br />

immuno‐histochemistry, we have found that specific areas <strong>of</strong> the brain are activated by<br />

infusing angiotensin, an action which is mediated through areas <strong>of</strong> the brain that does not<br />

have a blood brain barrier. These central pathways appear to be “sensitised”, reflecting a<br />

type <strong>of</strong> positive feed forward CNS plasticity.<br />

Project aims:<br />

We now wish to determine which areas <strong>of</strong> the brain <strong>and</strong> in particular the hypothalamus<br />

may be responsible for this effect on chronic stress<br />

Techniques:<br />

The project will involve some animal surgery, experiments to measure blood pressure<br />

before <strong>and</strong> during a relatively mild air jet stress <strong>and</strong> then perfusion fixation <strong>of</strong> the brain<br />

to process for immunohistochemistry. The use <strong>of</strong> specific antibodies, fluorescence <strong>and</strong><br />

confocal imaging will then be used to identify brain regions <strong>and</strong> specific neurochemical<br />

involved in the response to acute <strong>and</strong> chronic stress.<br />

Contacts:<br />

Pr<strong>of</strong>. Ge<strong>of</strong>frey Head <strong>and</strong> Dr Pamela Davern<br />

Department <strong>of</strong> Pharmacology<br />

Baker IDI Heart & Diabetes Institute<br />

Phone: 03 8532 1332<br />

Ge<strong>of</strong>f.head@bakeridi.edu.au<br />

Pamela.Davern@bakeridi.edu.au<br />

Fos Immunohistochemistry: Effect <strong>of</strong><br />

Chronic Stress <strong>and</strong> 6wks low dose Ang II<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

WHICH PARTS OF THE BRAIN ARE AFFECTED IN OBESITY<br />

RELATED HYPERTENSION?<br />

Supervisors: Pr<strong>of</strong>essor Ge<strong>of</strong>frey A. Head, Dr Pamela Davern <strong>and</strong><br />

Dr Joon Lim<br />

Location: Neuropharmacology Laboratory<br />

BakerIDI Heart & Diabetes Institute<br />

Prahran<br />

Background:<br />

Cardiovascular disease <strong>and</strong> its ultimate consequences are a significant burden on health<br />

systems in Australia <strong>and</strong> around the developed world. It is increasingly apparent that the<br />

high rate <strong>of</strong> hypertension in many societies can be attributed to an equally alarming rate<br />

<strong>of</strong> obesity. Despite this link, there is a less than complete underst<strong>and</strong>ing <strong>of</strong> the manner by<br />

which obesity alters central nervous system function to result in hypertension. It is known<br />

that the sympathetic nervous system (SNS), responsible in part for the control <strong>of</strong> blood<br />

pressure <strong>and</strong> is overactive in obese humans. It is also known that several peripheral<br />

signalling chemicals act at specific brain sites to modulate both appetite <strong>and</strong> also the<br />

activity <strong>of</strong> the SNS . For example, leptin, produced by fat <strong>and</strong> responsible for reducing<br />

appetite by acting at specific sites in the brain may act also to either stimulate or inhibit<br />

SNS activity depending on which neurons it acts at in the hypothalamus. Nonetheless,<br />

despite a comprehensive underst<strong>and</strong>ing <strong>of</strong> changes in appetite systems in the brain in<br />

obesity, <strong>and</strong> a known link between some <strong>of</strong> these appetite centres <strong>and</strong> other nuclei that<br />

control SNS outflow there is little underst<strong>and</strong>ing <strong>of</strong> how the sympathetic nervous system<br />

is activated during the development <strong>of</strong> obesity.<br />

Project aims:<br />

We seek to determine the manner by which various brain centres respond in obesity by<br />

studying patterns <strong>of</strong> neural activation <strong>and</strong> measuring SNS activity <strong>and</strong> blood pressure<br />

during the development <strong>of</strong> obesity using rabbits.<br />

Techniques:<br />

This project will involve surgical implantation <strong>of</strong> telemetric probes (for measurement <strong>of</strong><br />

renal sympathetic nerve activity <strong>and</strong> haemodynamic variables) <strong>and</strong> guide cannulae into<br />

distinct brain regions to allow injection <strong>of</strong> drugs. Students will also learn<br />

immunohistochemical techniques in order to identify by immun<strong>of</strong>luorescent labelling those<br />

neurons that are responsible for the genesis <strong>of</strong> obesity related hypertension.<br />

Contacts:<br />

Pr<strong>of</strong>. Ge<strong>of</strong>frey Head, Dr Pamela<br />

Davern & Dr Joon Lim<br />

Baker IDI Heart & Diabetes Institute<br />

Phone: 03 8532 1332<br />

Ge<strong>of</strong>f.head@bakeridi.edu.au<br />

Joon.Lim@bakeridi.edu.au<br />

Pamela.Davern@bakeridi.edu.au<br />

NITROXYL, A RELATIVE OF<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

HNO, IS A NATURALLY-OCCURRING CARDIOPROTECTIVE<br />

MOLECULE<br />

Supervisors: A/Pr<strong>of</strong> Rebecca Ritchie & Dr Barbara Kemp-Harper<br />

Location: Heart Failure Pharmacology Laboratory<br />

Baker IDI Heart & Diabetes Institute<br />

75 Commercial Rd, Melbourne<br />

Background:<br />

The nitric oxide (NO•)/cGMP signalling system is as a powerful cardiac antihypertrophic<br />

mechanism. Nitroxyl (HNO), a novel redox sibling <strong>of</strong> NO•, has several therapeutic<br />

advantages for the treatment <strong>of</strong> cardiovascular diseases. The nitric oxide (NO•)/cGMP<br />

signalling system is as a powerful cardiac antihypertrophic mechanism. Nitroxyl (HNO), a<br />

novel redox sibling <strong>of</strong> NO•, has several therapeutic advantages for the treatment <strong>of</strong><br />

cardiovascular diseases. We have shown that HNO prevents cardiomyocyte hypertrophy<br />

(abnormal pathological growth) <strong>and</strong> generation <strong>of</strong> superoxide, while concomitantly<br />

enhancing cardiac function (the latter in direct contrast to NO•).<br />

Project aims:<br />

This project explores whether HNO pharmacotherapy limits myocardial dysfunction,<br />

induced by hypertension, heart failure or diabetes, <strong>and</strong> the mechanisms for these actions.<br />

Putative independent mediators <strong>of</strong> chronic HNO cardioprotection include cGMP-mediated<br />

suppression <strong>of</strong> reactive oxygen species (ROS), <strong>and</strong> thiol-mediated preservation <strong>of</strong> cardiac<br />

calcium h<strong>and</strong>ling protein activity (e.g. SERCA2a, RyR2), whose activity is abnormally<br />

affected in cardiac pathologies. This project will be tailored depending on the student’s<br />

abilities <strong>and</strong> interests. The outcome <strong>of</strong> this project will be definitive information<br />

regarding the mechanism(s) <strong>and</strong> effectiveness <strong>of</strong> HNO-mediated rescue <strong>of</strong> myocardial<br />

dysfunction. Ultimately, HNO-based strategies may <strong>of</strong>fer new treatment options for<br />

cardiac disease, either alone or on top <strong>of</strong> st<strong>and</strong>ard care.<br />

Techniques:<br />

This project will provide the opportunity for learning a range <strong>of</strong> techniques, including cell<br />

culture (cardiomyocytes <strong>and</strong>/or cardiac fibroblasts), pharmacological (e.g. isolated rodent<br />

hearts ex vivo or in vivo) models <strong>of</strong> cardiac disease for assessing cardiac function <strong>and</strong><br />

blood pressure, biochemical (Westerns, ROS detection, ELISA, real-time PCR) <strong>and</strong>/or<br />

histological techniques.<br />

Contact:<br />

A/Pr<strong>of</strong> Rebecca Ritchie<br />

Heart Failure Pharmacology Laboratory<br />

Baker IDI Heart & Diabetes Institute<br />

Phone: 8532 1392<br />

Email: rebecca.ritchie@bakeridi.edu.au<br />

Heart disease (e.g.<br />

Hypertension, Diabetes)<br />

↑LV hypertrophy<br />

↑LV fibrosis<br />

X X<br />

↓ LV<br />

function<br />

↓SERCA/RyR<br />

function<br />

X<br />

↑SERCA/RyR<br />

function<br />

↑ cGMP thiol reactivity<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.<br />

↑ROS<br />

Nitroxyl<br />

heart failure<br />

death<br />

X ?


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

NEW STRATEGIES TO RESCUE<br />

DIABETES-INDUCED CARDIAC DYSFUNCTION<br />

Supervisors: A/Pr<strong>of</strong> Rebecca Ritchie<br />

Location: Heart Failure Pharmacology Laboratory<br />

Baker IDI Heart & Diabetes Institute<br />

75 Commercial Rd, Melbourne<br />

Background:<br />

Diabetes is Australia’s fastest growing chronic disease; one million Australians have been<br />

diagnosed, with close to one million more yet to be identified. Diabetes impairs left<br />

ventricular (LV) function, increasing the risk <strong>of</strong> death from heart failure > 2-fold. New<br />

therapies for restoring cardiac function in the diabetic heart are thus highly desirable.<br />

The aetiology <strong>of</strong> diabetic heart disease is distinct from other causes <strong>of</strong> LV dysfunction, as<br />

it is characterised initially by diastolic dysfunction, where relaxation <strong>of</strong> the cardiac<br />

muscle following contraction is prolonged. Our laboratory has demonstrated that<br />

antioxidant <strong>and</strong>/or ROS-suppressing approaches, as well as activation <strong>of</strong> cardioprotective<br />

signalling <strong>and</strong> negative regulators <strong>of</strong> LV hypertrophy, are beneficial for treating the<br />

cardiac complications <strong>of</strong> type 1 <strong>and</strong> type 2 diabetes in the intact heart.<br />

Project aims:<br />

This project explores a novel potential therapeutic strategy for rescuing cardiac function<br />

<strong>and</strong> structure in the diabetic heart, determining whether post-translational protein<br />

modifications induced by high glucose play a causal role in development <strong>of</strong> diabetic<br />

cardiomyopathy, <strong>and</strong> investigate whether pharmacological <strong>and</strong>/or gene-based strategies<br />

targeted at limiting these modifications can prevent diabetes-induced LV dysfunction, LV<br />

fibrosis, hypertrophy <strong>and</strong> excess generation <strong>of</strong> reactive oxygen species (ROS) such<br />

superoxide. Ultimately, treatment strategies that may emerge from these studies may<br />

provide significant benefits alone or in combination with current st<strong>and</strong>ard care, to<br />

ultimately reduce progression to heart failure <strong>and</strong> death in diabetic patients.<br />

Techniques:<br />

This project will provide the opportunity for learning pharmacological (e.g. isolated rodent<br />

hearts ex vivo or in vivo) models <strong>of</strong> cardiac disease for assessing cardiac function <strong>and</strong><br />

blood pressure, biochemical (Westerns, ROS detection, ELISA, real-time PCR) <strong>and</strong>/or<br />

histological techniques.<br />

Contact:<br />

A/Pr<strong>of</strong> Rebecca Ritchie<br />

Heart Failure Pharmacology Laboratory<br />

Baker IDI Heart & Diabetes Institute<br />

Phone: 8532 1392<br />

Email: rebecca.ritchie@bakeridi.edu.au<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

TARGETING ANTI-INFLAMMATORY PROTEIN ANNEXIN-A1<br />

FOR PROTECTION FROM MYOCARDIAL INFARCTION<br />

(HEART ATTACK)<br />

Supervisors: A/Pr<strong>of</strong> Rebecca Ritchie<br />

Location: Heart Failure Pharmacology Laboratory<br />

Baker IDI Heart & Diabetes Institute<br />

75 Commercial Rd, Melbourne<br />

Background:<br />

Myocardial infarction (a sustained impairment in coronary bloodflow) <strong>and</strong> the resultant<br />

heart failure is a major cause <strong>of</strong> death in Western societies. The therapeutic potential <strong>of</strong><br />

the endogenous anti-inflammatory mediator annexin-A1 (ANX-A1) has been recognized in a<br />

range <strong>of</strong> inflammatory disorders. We have shown that ANX-A1 has powerful protective<br />

actions against cardiac injury <strong>and</strong> loss <strong>of</strong> LV contractile function. This project explores<br />

the potential for ANX-A mimetics to reduce cardiac ischaemia-reperfusion injury.<br />

Project aims:<br />

The project will test the hypothesis that ANX-A1 represents a novel modulator <strong>of</strong><br />

myocardial viability <strong>and</strong> LV contractile function following ischaemia-reperfusion, <strong>and</strong> will<br />

seek to investigate the cardioprotective function <strong>of</strong> endogenous ANX-A1 in I-R injury, the<br />

receptors responsible for cardioprotection elicited by ANX-A1 <strong>and</strong> its mimetics (likely the<br />

formyl peptide receptor family, FPR1 <strong>and</strong>/or FPR2), <strong>and</strong> examine the potential therapeutic<br />

opportunities <strong>of</strong>fered by exogenous ANX-A1 mimetics after I-R injury in the intact heart.<br />

Development <strong>of</strong> therapeutic strategies for treating myocardial infarction, alone or<br />

concurrent with st<strong>and</strong>ard care, will ultimately reduce progression to heart failure <strong>and</strong><br />

death in affected patients.<br />

Techniques:<br />

The project provides the opportunity for learning a range <strong>of</strong> techniques, including cell<br />

culture, as well as physiological ex vivo <strong>and</strong>/or in vivo models <strong>of</strong> cardiac ischaemia for<br />

studying cardiac function <strong>and</strong> structure, biochemical (<strong>and</strong>/or histological techniques.<br />

Contact:<br />

A/Pr<strong>of</strong> Rebecca Ritchie<br />

Heart Failure Pharmacology Laboratory<br />

Baker IDI Heart & Diabetes Institute<br />

Phone: 8532 1392<br />

Email: rebecca.ritchie@bakeridi.edu.au<br />

↓ inflammation<br />

in vivo<br />

preservation <strong>of</strong><br />

cardiac morphology<br />

ANX-A1 mimetics<br />

FPR2 FPR1<br />

G q/11 G<br />

↑ myocardial<br />

viability<br />

heart failure<br />

death<br />

early rescue <strong>of</strong><br />

contractile function<br />

preservation <strong>of</strong><br />

contractile function<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.<br />

X<br />

P-Akt<br />

G i/o<br />

X


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

SIGNALLING BIAS OF CALCITONIN RECEPTOR POLYMORPHS<br />

Supervisors: Dr Sebastian Furness <strong>and</strong> Pr<strong>of</strong> Patrick Sexton<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background:<br />

The Calcitonin receptor (CTR) is involved in calcium homeostasis by regulating its secretion<br />

in kidneys <strong>and</strong> the resorbtion <strong>of</strong> bone by osteoclasts in response to Calcitonin. The CTR is<br />

polymorphic in the C-terminal tail with a number <strong>of</strong> epidemiological studies correlating this<br />

polymorphism with osteoporotic risk. One previous study failed to uncover differences<br />

between CTR polymorhs. We have used several model cell backgrounds to analyse<br />

differences in signaling arising from these polymorphs. We have shown that there are a<br />

number <strong>of</strong> polymorphism-dependant changes in signaling efficacy toward adenylate<br />

cyclase, extracellular signal-regulated kinase (ERK) phosphorylation <strong>and</strong> intracellular<br />

calcium release. Further, we have demonstrated that these signaling biases are dependent<br />

on cellular background.<br />

Project aim:<br />

This project will extend the existing signalling bias studies using bioluminescence<br />

resonance energy transfer (BRET) to examine the interaction between CTR <strong>and</strong> members<br />

<strong>of</strong> the arrestin <strong>and</strong> GRK families.<br />

Techniques:<br />

Molecular Biology: basic molecular biology procedures including isolation <strong>and</strong> preparation <strong>of</strong><br />

DNA, cell culture <strong>and</strong> cell transfection. Protein-protein interaction will be measured by<br />

resonance energy transfer <strong>and</strong> via fluorescence imaging. Pharmacological characterization<br />

<strong>of</strong> receptors will be performed using high-throughput assays for measurement <strong>of</strong><br />

intracellular signaling including cAMP formation, ERK phosphylation <strong>and</strong> intracellular<br />

calcium mobilization.<br />

Contacts:<br />

Dr Sebastian Furness<br />

Phone: 9903 9055, Rm 5-09, Building 404;<br />

Sebastian.furness@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

COMPARTMENTALISATION OF RELAXIN-STIMULATED<br />

SIGNALLING IN SINGLE CELLS<br />

Supervisors: Dr Michelle Halls & Pr<strong>of</strong> Roger Summers<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background:<br />

Relaxin is a pleiotropic hormone that is implicated in multiple physiological systems, <strong>and</strong><br />

has therapeutic potential in heart failure (currently in phase III clinical trials), fibrosis<br />

<strong>and</strong> cancer metastases. Relaxin binding to its G protein - coupled receptor, RXFP1, induces<br />

the activation <strong>of</strong> multiple signalling pathways. The major consequence <strong>of</strong> receptor<br />

stimulation is increased cAMP production, which occurs via a complex, simultaneous<br />

activation <strong>of</strong> three distinct G protein pathways: Gαs stimulation <strong>of</strong> adenylyl cyclase (AC)<br />

activity to increase cAMP, GαoB inhibition <strong>of</strong> AC, <strong>and</strong> Gαi3 -activation <strong>of</strong> AC.<br />

The activation <strong>of</strong> three separate pathways to independently affect the same second<br />

messenger, suggests that relaxin-stimulated cAMP production is compartmentalised within<br />

the cell. Compartmentalisation, or the specific localisation <strong>of</strong> signalling mediators into<br />

discrete regions <strong>of</strong> the cell, allows cells to specifically tailor a physiological response to a<br />

distinct stimulus.<br />

Project aim:<br />

This project will explore the extent <strong>of</strong> signalling compartmentalisation following relaxin<br />

activation <strong>of</strong> RXFP1. Specifically, we will use state-<strong>of</strong>-the-art biosensors, which can be<br />

targeted to different areas <strong>of</strong> the cell, to examine the compartmentalisation <strong>of</strong> cAMP,<br />

protein kinase C (PKC) <strong>and</strong> extracellular signal regulated kinase ( -RK) phosphorylation in<br />

single live cells. Information gained from this project will demonstrate the importance <strong>of</strong><br />

signalling compartmentalisation in the generation <strong>of</strong> a defined physiological response.<br />

Further, underst<strong>and</strong>ing the compartmentalisation <strong>of</strong> relaxin -mediated signalling may make<br />

clear some <strong>of</strong> the clinically relevant effects <strong>of</strong> relaxin in conditions such as heart failure.<br />

Techniques:<br />

Molecular'Biology: basic molecular biology procedures including isolation <strong>and</strong> preparation<br />

<strong>of</strong> DNA, cell culture <strong>and</strong> cell transfection. Single-Cell Imaging: cutting edge, high<br />

throughput imaging will be used to measure activation <strong>of</strong> cAMP <strong>and</strong> phosphorylation <strong>of</strong> PKC<br />

<strong>and</strong> ERK by the use <strong>of</strong> fluorescence -resonance energy transfer (FRET) -based biosensors<br />

targeted to the plasma membrane, bulk cytosol or nucleus <strong>of</strong> single cells. Pharmacological:<br />

the intracellular signalling pathways leading to each <strong>of</strong> the compartmentalised responses<br />

will be elucidated by the use <strong>of</strong> pharmacological inhibitors <strong>of</strong> signalling.<br />

Contacts:<br />

Dr Michelle Halls<br />

Phone: 9903 9094, Rm 324, Building 404<br />

michelle.halls@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

MOLECULAR MECHANISMS OF A NOVEL ALLOSTERIC<br />

MODULATOR OF THE D2-LIKE DOPAMINE RECEPTORS<br />

Supervisors: Dr Rob Lane, Pr<strong>of</strong> Arthur Christopoulos<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background & Project Details:<br />

The neurotransmitter dopamine mediates its action via five G protein coupled receptors<br />

(the dopamine receptors D1-5R) <strong>and</strong> been shown to play a vital role in a range <strong>of</strong> central<br />

nervous system functions including voluntary movement, reward, memory <strong>and</strong> learning.<br />

Dysregulation <strong>of</strong> dopamine signalling is associated with many human disorders, including<br />

schizophrenia <strong>and</strong> Parkinson’s disease.<br />

Although, antipsychotic drug discovery at the dopamine receptors has focused on<br />

targeting the binding site for endogenous dopamine (the ‘orthosteric’ site) this approach is<br />

associated with severe extrapyramidal side-effects <strong>and</strong> poor receptor subtype selectivity.<br />

Targeting a spatially distinct ‘allosteric’ site on the receptor may <strong>of</strong>fer greater subtype<br />

selectivity with fewer side-effects. Recently, the first drug-like allosteric lig<strong>and</strong> <strong>of</strong> the<br />

dopamine receptor was described. We have demonstrated that this lig<strong>and</strong> adopts a<br />

simultaneous orthosteric/allosteric (bitopic) mode <strong>of</strong> binding at the dopamine D2<br />

receptor. However, to enable us to develop novel improved modulators <strong>of</strong> this receptor we<br />

need to underst<strong>and</strong> how this drug exerts its modulatory effect.<br />

To determine the mechanism <strong>and</strong> site <strong>of</strong> action <strong>of</strong> this novel drug, we will use a combined<br />

approach. First, we have generated a library <strong>of</strong> receptors in which we have mutated amino<br />

acids within the receptor that have been implicated in the binding <strong>of</strong> both orthosteric<br />

lig<strong>and</strong>s or potential allosteric lig<strong>and</strong>s. Second, we have synthesised a number <strong>of</strong><br />

derivatives <strong>of</strong> this drug in which key parts <strong>of</strong> the drug have been modified. These mutant<br />

receptors <strong>and</strong> novel drugs will be characterized using a number <strong>of</strong> state <strong>of</strong> the art<br />

functional assays. Information gained from this project <strong>and</strong> the recently published crystal<br />

structure <strong>of</strong> the dopamine D3 receptor can be used to predict the mode <strong>of</strong> binding <strong>of</strong> this<br />

receptor <strong>and</strong> will be the starting point towards the development <strong>of</strong> novel allosteric drugs<br />

for this important therapeutic target.<br />

Techniques:<br />

Structural: Site-directed mutagenesis <strong>and</strong> molecular modeling, to determine the binding<br />

mode <strong>of</strong> SB269652. Pharmacological: Receptor function will be quantified by downstream<br />

signalling using state-<strong>of</strong>-the-art, high throughput assays <strong>of</strong> extracellular signal regulated<br />

kinases (ERK1/2), intracellular cAMP, <strong>and</strong> resonance energy transfer techniques to detect<br />

interaction with β arrestin. Molecular biological: As part <strong>of</strong> the project you will also learn<br />

basic molecular biological procedures including isolation <strong>and</strong> preparation <strong>of</strong> DNA, cell<br />

culture <strong>and</strong> cell transfection.<br />

Contacts:<br />

Dr Rob Lane<br />

Phone: 9903 9095, Rm 325, Building 404<br />

rob.lane@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

PROBING AN ALLOSTERIC BINDING SITE IN THE HUMAN<br />

CASR<br />

Supervisors: Dr Katie Leach, Dr Laura Lopez, Pr<strong>of</strong> Arthur Christopoulos<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background & Project Details:<br />

The human calcium sensing receptor (CaSR) is a family C G protein coupled receptor<br />

(GPCR) that plays a pivotal role in extracellular calcium (Ca2+o) homeostasis <strong>and</strong><br />

parathyroid hormone (PTH) secretion. It is expressed on the surface <strong>of</strong> a variety <strong>of</strong> cell<br />

types throughout the body, where it responds to small increases in free Ca2+o<br />

concentrations. Over 200 clinically relevant polymorphisms have been identified in the<br />

CaSR <strong>and</strong> unfortunately they can result in a number <strong>of</strong> disorders, such as autosomal<br />

dominant hypocalcaemia (ADH), Bartter syndrome type V, familial hypocalciuric<br />

hypercalcaemia (FHH), familial benign hypercalcaemia (FBH) <strong>and</strong> neonatal severe<br />

hyperparathyroidism (NSHPT). These disorders are generally characterised by an<br />

imbalance <strong>of</strong> PTH <strong>and</strong>/or Ca2+o.<br />

Cinacalcet, a positive allosteric CaSR modulator prescribed for the treatment <strong>of</strong> primary<br />

<strong>and</strong> secondary hyperparathyroidism was recently shown to successfully normalise serum<br />

Ca2+o in individuals with loss-<strong>of</strong>-function CaSR mutations, suggesting that cinacalcet <strong>and</strong><br />

other allosteric CaSR modulators could be invaluable in the treatment <strong>of</strong> disorders linked<br />

to CaSR mutations. Recent molecular modelling <strong>and</strong> mutagenesis studies have predicted<br />

that the binding site(s) for allosteric CaSR modulators comprises residues located in the<br />

transmembrane (TM) domains <strong>and</strong> extracellular loops <strong>of</strong> the receptor, where a growing<br />

number <strong>of</strong> naturally occurring mutations are being identified. However, although<br />

mutagenesis studies, which have involved alanine substitution <strong>of</strong> amino acids predicted to<br />

interact with allosteric drugs, have shown effects on the actions <strong>of</strong> allosteric modulators<br />

at the CaSR, no study has differentiated between effects <strong>of</strong> these mutations on the<br />

binding affinity <strong>of</strong> the allosteric modulator, versus the cooperativity <strong>of</strong> the modulator;<br />

that is to say the ability <strong>of</strong> the drug to modulate the actions <strong>of</strong> the endogenous agonist,<br />

Ca2+o. For instance, mutation <strong>of</strong> Iso841 to Ala (I841A) in the 7th TM domain <strong>of</strong> the<br />

receptor was previously reported to attenuate the binding affinity <strong>of</strong> the negative<br />

allosteric modulator, Calhex 231, due to effects <strong>of</strong> the mutation on the ability <strong>of</strong> Calhex<br />

231 to reduce Ca2+o signalling. However, we have shown that these mutational effects are<br />

driven by changes in the cooperativity between Ca2+o <strong>and</strong> the negative allosteric<br />

modulator, whilst the binding affinity <strong>of</strong> the drug is unaltered at the I841A mutant. Thus,<br />

it is imperative that mutagenesis data be interpreted correctly if we want to define the<br />

binding site <strong>of</strong> allosteric CaSR drugs.<br />

Techniques: Cell culture: This project requires routine growth <strong>and</strong> culturing <strong>of</strong> human<br />

kidney embryonic (HEK293) cell lines expressing the wild type <strong>and</strong> headless human CaSRs.<br />

Pharmacological: Receptor function will be quantified by downstream signalling using high<br />

throughput assays that measure extracellular signal regulated kinases (ERK1/2),<br />

intracellular Ca2+o mobilisation <strong>and</strong> inositol phosphate accumulation. Structural: Alanine<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

substitution <strong>of</strong> amino acids that have either previously been implicated in the binding <strong>of</strong><br />

allosteric CaSR modulators, or that have been identified from a recent investigation into<br />

the effects <strong>of</strong> naturally occurring amino acids performed in our laboratory, will be used to<br />

determine the involvement <strong>of</strong> amino acids in the function <strong>of</strong> allosteric modulators. This<br />

information will be used to aid the design <strong>of</strong> more accurate molecular models <strong>of</strong> the CaSR,<br />

which are currently lacking.<br />

Contacts:<br />

Dr Katie Leach<br />

Phone: 9903 9089, Rm 344, Building 404<br />

Katie.leach@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

ALLOSTERIC PHARMACOREGULATION OF A HEADLESS CASR<br />

Supervisors: Dr Katie Leach, Pr<strong>of</strong> Arthur Christopoulos<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background & Project Details:<br />

The human calcium sensing receptor (CaSR) is a family C G protein coupled receptor<br />

(GPCR) that plays a pivotal role in extracellular calcium (Ca2+o) homeostasis <strong>and</strong><br />

parathyroid hormone (PTH) secretion. It is expressed on the surface <strong>of</strong> a variety <strong>of</strong> cell<br />

types throughout the body, where it responds to small increases in free Ca2+o<br />

concentrations. Although the primary Ca2+- (orthosteric) binding site has been localised<br />

to the large extracellular N-terminal region <strong>of</strong> the receptor [1-3], otherwise known as the<br />

venus flytrap (VFT) domain, evidence has pointed towards an additional binding site within<br />

the transmembrane spanning regions <strong>of</strong> the receptor [4-5]. This second site is<br />

topographically distinct from the region that binds allosteric calcilytics <strong>and</strong> calcimimetics<br />

[5], which modulate the activity <strong>of</strong> Ca2+ o at the CaSR, although it is unclear which amino<br />

acids form this second binding site for Ca2+ o. Thus, the aim <strong>of</strong> this study is to<br />

investigate the activity <strong>of</strong> orthosteric <strong>and</strong> allosteric lig<strong>and</strong>s at a “headless” CaSR that<br />

lacks the first 599 amino acids that comprise the VFT domain <strong>and</strong> is thus composed <strong>of</strong> the<br />

TM domains <strong>and</strong> a functional but truncated C-terminal tail, which lacks the last 176 amino<br />

acids <strong>of</strong> the receptor. The extent to which allosteric modulators retain their ability to<br />

regulate CaSR activity in the absence <strong>of</strong> the primary Ca2+o-binding site will be<br />

investigated to gain an underst<strong>and</strong>ing <strong>of</strong> the mechanism <strong>of</strong> allosteric modulation at the<br />

human CaSR.<br />

1. Huang, Y., Zhou, Y., Castiblanco, A., Yang, W., Brown, E. M., <strong>and</strong> Yang, J. J. (2009)<br />

Biochemistry 48, 388-398<br />

2. Huang, Y., Zhou, Y., Yang, W., Butters, R., Lee, H. W., Li, S., Castiblanco, A., Brown,<br />

E. M., <strong>and</strong> Yang, J. J. (2007) J Biol Chem 282, 19000-19010<br />

3. Silve, C., Petrel, C., Leroy, C., Bruel, H., Mallet, E., Rognan, D., <strong>and</strong> Ruat, M. (2005)<br />

J Biol Chem 280, 37917-37923<br />

4. Ray, K., Tisdale, J., Dodd, R. H., Dauban, P., Ruat, M., <strong>and</strong> Northup, J. K. (2005) J<br />

Biol Chem 280, 37013-37020<br />

5. Ray, K., <strong>and</strong> Northup, J. (2002) J Biol Chem 277, 18908-18913<br />

Techniques: Cell culture: This project requires routine growth <strong>and</strong> culturing <strong>of</strong> human<br />

kidney embryonic (HEK293) cell lines expressing the wild type <strong>and</strong> headless human CaSRs.<br />

Pharmacological: Receptor function will be quantified by downstream signalling using high<br />

throughput assays that measure extracellular signal regulated kinases (ERK1/2),<br />

intracellular Ca2+o mobilisation <strong>and</strong> inositol phosphate accumulation.<br />

Contacts:<br />

Dr Katie Leach<br />

Phone: 9903 9089, Rm 344, Building 404<br />

Katie.leach@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

INVESTIGATION OF MUSCARINIC M4 RECEPTOR FUNCTION<br />

USING LABEL FREE IMPEDANCE TECHNOLOGY<br />

Supervisors: Dr Patricia Rueda, Pr<strong>of</strong> Arthur Christopoulos, Dr Chris Langmead<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background & Project Details:<br />

The muscarinic M4 receptor plays a role in a range <strong>of</strong> central nervous system functions<br />

including voluntary movement, reward, memory <strong>and</strong> learning. The metabotropic actions <strong>of</strong><br />

acetylcholine are mediated by its interaction with five G protein-coupled receptors<br />

(GPCRs), the muscarinic M1-M5 receptors. Because acetylcholine is involved in such critical<br />

physiological processes, it is not surprising that many human disorders, including<br />

schizophrenia, Parkinson’s disease <strong>and</strong> Alzheimer’s disease, have been related to<br />

cholinergic dysfunction.<br />

The muscarinic M4 receptor has been studied as a target <strong>of</strong> interest for the treatment <strong>of</strong><br />

schizophrenia; agonists or positive allosteric modulators (PAMs) are under consideration<br />

as potential antipsychotic agents.<br />

Recently, GPCR research has been enriched by the development <strong>of</strong> label-free technologies<br />

(using cellular impedance) which allow phenotypic measurement <strong>of</strong> cell function <strong>and</strong> are not<br />

restricted to single signalling pathways. Using this technology it is possible to evaluate<br />

whole cell responses to drug challenges <strong>and</strong> dissect kinetics <strong>of</strong> responses as well as biased<br />

agonism.<br />

Using this label-free technology, the pharmacology novel agonists <strong>and</strong> PAMs <strong>of</strong> the<br />

muscarinic M4 receptor will be studied; inhibitors <strong>of</strong> G protein coupling <strong>and</strong> signal<br />

transduction pathways will be used to characterise the functional responses; these will<br />

also be verified in orthogonal assays <strong>of</strong> muscarinic M4 receptor function. This will lead to<br />

a greater underst<strong>and</strong>ing <strong>of</strong> activation <strong>and</strong> allosteric modulation <strong>of</strong> the muscarinic M4<br />

receptor which will be <strong>of</strong> benefit in the development <strong>of</strong> novel antipsychotic drugs.<br />

Techniques:<br />

Pharmacological: Receptor function will be quantified by downstream signalling using state<strong>of</strong>-the-art,<br />

high throughput assays <strong>of</strong> cellular impedance <strong>and</strong> possibly extracellular signal<br />

regulated kinases (ERK1/2), intracellular cAMP or resonance energy transfer techniques to<br />

detect interaction with β arrestin.<br />

Molecular biological: As part <strong>of</strong> the project you will also learn basic molecular biological<br />

procedures including isolation <strong>and</strong> preparation <strong>of</strong> DNA, cell culture <strong>and</strong> cell transfection.<br />

Contacts:<br />

Dr Chris Langmead<br />

Phone: 9903 9096, Rm 515, Building 404<br />

chris.langmead@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

INVESTIGATING THE SIGNALLING CASCADE OF NOVEL<br />

ADENOSINE RECEPTOR LIGANDS AT THE ADENOSINE A2B<br />

RECEPTOR (A2B-AR)<br />

Supervisors: Dr Celine Valant, Dr Lauren May<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background & Project Details:<br />

Since its discovery as a low-affinity adenosine receptor (AR), the A2B receptor (A2B-AR),<br />

has proven enigmatic in its function. The lack <strong>of</strong> specific pharmacological agents targeting<br />

the A2B-AR made its initial characterization challenging.<br />

However, the importance <strong>of</strong> this receptor was reconsidered when it was observed that the<br />

A2B-AR is highly transcriptionally regulated by factors implicated in inflammatory<br />

hypoxia. The notion that during ischemia or inflammation extracellular adenosine is<br />

dramatically elevated to levels sufficient for A2B-AR activation, indicated that A2B-AR<br />

signaling may be important to dampen inflammation particularly during tissue hypoxia.<br />

Additionally, the involvement <strong>of</strong> this receptor in processes such as interleukins secretion,<br />

Ca2+ mobilization, hepatic glucose regulation, tumor vascularisation, <strong>and</strong> cardioprotection<br />

have stimulated many researchers to develop specific agonists <strong>and</strong> antagonists. For many<br />

years, the lack <strong>of</strong> potent <strong>and</strong> selective A2B-AR lig<strong>and</strong>s precluded a deep exploration <strong>of</strong><br />

their therapeutic prospective; at present, much progress in the field <strong>of</strong> antagonists led to<br />

preclinical studies for different compounds. Less populated is the universe <strong>of</strong> A2B-AR<br />

agonists, but really promising for the involvement in ischemic preconditioning.<br />

Here we propose to study the pharmacological properties <strong>of</strong> various adenosine lig<strong>and</strong>s,<br />

including the recently in-house identified highly potent A2B-AR agonist, VCP746. Following<br />

receptor activation, the A2B-AR couple predominantly to Gs protein to induce cAMP<br />

production via stimulation <strong>of</strong> adenylate cyclases. As such cAMP production will be initially<br />

used as a st<strong>and</strong>ard assay to quantify agonist potency in the absence as well as in the<br />

presence <strong>of</strong> selective pathway inhibitors <strong>of</strong> various stages <strong>of</strong> the signalling cascade <strong>of</strong> the<br />

adenosine A2B-AR. Ultimately, if time permits, other signalling assays will be investigated,<br />

such as Ca2+ mobilization <strong>and</strong> ERK1/2 phosphorylation.<br />

Techniques:<br />

Cell culture: In this project you will learn the basics <strong>of</strong> cell culture, such as splitting <strong>and</strong><br />

seeding cells.<br />

Pharmacology: Various singalling pathways will be learnt. Initially, cAMP production will be<br />

the main signaling assay investigated in this study but if the time permits, other<br />

downstream signalling can be investigated, such as ERK1/2 phosphorylation, calcium<br />

mobilization or cell survival.<br />

Contacts:<br />

Dr Celine Valant<br />

Phone: 9903 9091, Rm 344, Building 404; celine.valant@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

INVESTIGATION OF AN ABSOLUTELY CONSERVED POCKET AT<br />

THE MUSCARINIC ACETYLCHOLINE M1 RECEPTOR (M1 MACHR)<br />

Supervisors: Dr Celine Valant, Dr Meritxel Canals<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background & Project Details:<br />

Five muscarinic acetylcholine receptor (mAChR) subtypes have been cloned (M1, M2, M3,<br />

M4 <strong>and</strong> M5). For years now, there has been great interest in the structure-function<br />

relationships <strong>of</strong> mAChRs because these prototypical Family A G protein-coupled receptors<br />

(GPCRs) are attractive therapeutic targets for both peripheral <strong>and</strong> central nervous<br />

system disorders. A plethora <strong>of</strong> drugs that act at the mAChRs have been identified over<br />

the last few decades, but many <strong>of</strong> these show minimal selectivity for any one <strong>of</strong> the five<br />

mAChR subtypes over the others, which has hampered their development into<br />

therapeutics due to adverse side effects. The lack <strong>of</strong> drug specificity is primarily due to<br />

high sequence similarity in this family <strong>of</strong> receptor, especially in the orthosteric<br />

(endogenous lig<strong>and</strong>) binding pocket.<br />

Recently, with the success <strong>of</strong> crystallization <strong>of</strong> Family A GPCRs, especially in the mAChR<br />

family, a number <strong>of</strong> residues outside the endogenous lig<strong>and</strong> binding pocket, absolutely<br />

conserved cross all five mAChR subtypes, <strong>and</strong> highly conserved across the family A GPCRs,<br />

have been identified. These thirteen residues appear to form an extended binding pocket<br />

located right below the orthosteric binding site. In an effort to underst<strong>and</strong> the purpose<br />

<strong>of</strong> such highly conserved deep pocket, we propose to investigate one by one the effect <strong>of</strong><br />

each residue, using the M1 mAChR as a template. Starting with alanine mutation <strong>of</strong> each<br />

residue, the binding <strong>and</strong> functional properties <strong>of</strong> a selection <strong>of</strong> muscarinic lig<strong>and</strong>s,<br />

antagonists, non-selective agonists, <strong>and</strong> especially ‘selective allosteric agonists’, such as<br />

77-LH-28, AC-42, TBPB <strong>and</strong> BQCA, will be investigated.<br />

Techniques:<br />

Cell culture: In this project you will learn the basics <strong>of</strong> cell culture, such as splitting <strong>and</strong><br />

seeding cells, as well as cell transfection.<br />

Pharmacology: Radiolig<strong>and</strong> binding assays as well as various signaling assays will be learnt<br />

during this study.<br />

Molecular biology: As part <strong>of</strong> the project you will also learn basic molecular biological<br />

procedures including isolation <strong>and</strong> preparation <strong>of</strong> DNA.<br />

Contacts:<br />

Dr Celine Valant<br />

Phone: 9903 9091, Rm 344, Building 404<br />

celine.valant@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

INVESTIGATION OF AN ABSOLUTELY CONSERVED POCKET AT<br />

THE MUSCARINIC ACETYLCHOLINE M2 RECEPTOR (M2 MACHR)<br />

Supervisors: Dr Celine Valant, Pr<strong>of</strong> Arthur Christopoulos<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background & Project Details:<br />

Five muscarinic acetylcholine receptor (mAChR) subtypes have been cloned (M1, M2, M3,<br />

M4 <strong>and</strong> M5). For years now, there has been great interest in the structure-function<br />

relationships <strong>of</strong> mAChRs because these prototypical Family A G protein-coupled receptors<br />

(GPCRs) are attractive therapeutic targets for both peripheral <strong>and</strong> central nervous<br />

system disorders. A plethora <strong>of</strong> drugs that act at the mAChRs have been identified over<br />

the last few decades, but many <strong>of</strong> these show minimal selectivity for any one <strong>of</strong> the five<br />

mAChR subtypes over the others, which has hampered their development into<br />

therapeutics due to adverse side effects. The lack <strong>of</strong> drug specificity is primarily due to<br />

high sequence similarity in this family <strong>of</strong> receptor, especially in the orthosteric<br />

(endogenous lig<strong>and</strong>) binding pocket.<br />

Recently, with the success <strong>of</strong> crystallization <strong>of</strong> Family A GPCRs, especially in the mAChR<br />

family, a number <strong>of</strong> residues outside the endogenous lig<strong>and</strong> binding pocket, absolutely<br />

conserved cross all five mAChR subtypes, <strong>and</strong> highly conserved across the family A GPCRs,<br />

have been identified. These thirteen residues appear to form an extended binding pocket<br />

located right below the orthosteric binding site at the M2 mAChR. In an effort to<br />

underst<strong>and</strong> the purpose <strong>of</strong> such highly conserved deep pocket, we propose to investigate<br />

one by one the effect <strong>of</strong> each residue. Starting with alanine mutation <strong>of</strong> each residue, the<br />

binding <strong>and</strong> functional properties <strong>of</strong> a selection <strong>of</strong> muscarinic lig<strong>and</strong>s, such as antagonists,<br />

agonists <strong>and</strong> allosteric modulators will be thoroughly investigated.<br />

Techniques:<br />

Cell culture: In this project you will learn the basics <strong>of</strong> cell culture, such as splitting <strong>and</strong><br />

seeding cells, as well as cell transfection.<br />

Pharmacology: Radiolig<strong>and</strong> binding assays as well as various signaling assays will be learnt<br />

during this study.<br />

Molecular biology: As part <strong>of</strong> the project you will also learn basic molecular biological<br />

procedures including isolation <strong>and</strong> preparation <strong>of</strong> DNA.<br />

Contacts:<br />

Dr Celine Valant<br />

Phone: 9903 9091, Rm 344, Building 404<br />

celine.valant@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.


Department <strong>of</strong> Pharmacology, <strong>Honours</strong> 2013<br />

PROBING STRUCTURE ACTIVITY RELATIONSHIPS FOR<br />

GLUCAGON-LIKE PEPTIDE-1 RECEPTOR ALLOSTERIC LIGANDS<br />

Supervisors: Dr Denise Wootten <strong>and</strong> Pr<strong>of</strong> Patrick Sexton<br />

Location: Drug Discovery Biology theme<br />

Monash Institute <strong>of</strong> Pharmaceutical Sciences<br />

Monash University, Parkville<br />

Background:<br />

The glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) is a family B GPCR that plays an<br />

essential role in nutrient mediated insulin release <strong>and</strong> as such is a therapeutic target for<br />

the treatment <strong>of</strong> type II diabetes. Unlike most GPCRs, the GLP-1R is activated by four<br />

distinct endogenous GLP-1 variants, as well as by further endogenous <strong>and</strong> exogenous<br />

lig<strong>and</strong>s. <strong>Current</strong> GLP-1R based therapeutics for treatment <strong>of</strong> type II diabetes is<br />

restricted to daily intravenous or subcutaneous administrations <strong>of</strong> peptide analogues <strong>of</strong><br />

GLP-1. Therefore, orally active, small molecule agonists <strong>of</strong> the GLP-1 receptor are highly<br />

desired. Recent developments in the GLP-1R field include the discovery <strong>of</strong> nonpeptide<br />

lig<strong>and</strong>s, which bind at sites topographically distinct (allosteric) from the endogenous<br />

peptides. Allosteric lig<strong>and</strong>s <strong>of</strong>fer alternative routes to receptor activation that may be<br />

able to either modulate the existing peptide response <strong>and</strong>/or activate the receptor alone.<br />

We have recently characterised two <strong>of</strong> these nonpeptide GLP-1R allosteric lig<strong>and</strong>s,<br />

Compound 2 <strong>and</strong> BETP. In recombinant cell systems, Compound 2 was a strong partial<br />

agonist in cAMP signalling <strong>and</strong> a positive allosteric modulator <strong>of</strong> oxyntomdulin activity (but<br />

not <strong>of</strong> other orthosteric agonists), whereas BETP displayed only weak partial intrinsic<br />

agonsim in cAMP, but similarly to Compound 2, selectively modulated oxyntomodulin<br />

affinity <strong>and</strong> efficacy in cAMP signalling.<br />

Project aim:<br />

The aim <strong>of</strong> this project is to screen in a library <strong>of</strong> compounds based on the scaffolds <strong>of</strong><br />

Compound 2 <strong>and</strong> BETP in the classical signalling pathway (cAMP) to identify a structureactivity<br />

pr<strong>of</strong>ile for intrinsic efficacy <strong>and</strong> modulator activity. In addition, other signalling<br />

pathways (such as pERK1/2, iCa2+ <strong>and</strong> b-arrestin recruitment) will be assessed in an<br />

attempt to discover biased <strong>and</strong>/or probe-dependent lig<strong>and</strong>s which may be used in future<br />

studies to interrogate the in vivo biology <strong>of</strong> the GLP-1R.<br />

Techniques:<br />

Receptor function will be quantified by downstream signalling using state-<strong>of</strong>-theart,<br />

high throughput assays <strong>of</strong> intracellular cAMP, extracellular signal regulated<br />

kinases (ERK1/2), iCa2+ mobilization assays <strong>and</strong> resonance energy transfer<br />

techniques to detect interaction with β arrestins. Radiolig<strong>and</strong> binding assays will be<br />

used to assess lig<strong>and</strong> affinities. As part <strong>of</strong> the project you will also learn various<br />

cell culture techniques.<br />

Contacts: Dr Denise Wootten<br />

Phone: 9903 9088, Rm 5-09, Building 404; Denise.wootten@monash.edu<br />

Copyright © Monash University 2012. All rights reserved. Except as provided in the Copyright Act 1968, this<br />

work may not be reproduced in any form without the written permission <strong>of</strong> the host <strong>Faculty</strong> <strong>and</strong><br />

School/Department.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!