02.05.2015 Views

Institute of Membrane & Systems Biology - Faculty of Biological ...

Institute of Membrane & Systems Biology - Faculty of Biological ...

Institute of Membrane & Systems Biology - Faculty of Biological ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>Institute</strong> <strong>of</strong> <strong>Membrane</strong><br />

& <strong>Systems</strong> <strong>Biology</strong><br />

<strong>Faculty</strong> <strong>of</strong> <strong>Biological</strong> Sciences


<strong>Institute</strong> <strong>of</strong> <strong>Membrane</strong><br />

& <strong>Systems</strong> <strong>Biology</strong><br />

Welcome to the <strong>Institute</strong>’s brochure. Its purpose is to provide an introduction to our<br />

research activity – what we do and why we do it. The brochure is intended for a broad<br />

spectrum <strong>of</strong> readers – approachable for those considering study at the University,<br />

informative for experts wishing to benefit from our research or invest in it. The <strong>Institute</strong><br />

has two main reasons for existence – to educate and to discover. This brochure focuses<br />

mostly on the discovery side. Nevertheless, you should know that we also have extensive<br />

undergraduate and postgraduate education programmes running in parallel.<br />

These programmes are not separate from the research but inter-woven with it. Research informs the direction <strong>of</strong> teaching<br />

and, in many cases, causes it to be revised annually. It also provides a rich and modern experience in research discovery for<br />

undergraduates and postgraduates through projects in our research laboratories. We should also not forget that research is a<br />

central platform for the reputation <strong>of</strong> our University, which will influence the lives <strong>of</strong> most who study at it.<br />

The <strong>Institute</strong> is one <strong>of</strong> three research institutes in the <strong>Faculty</strong>. Its academic staff and research fellows have interests ranging from<br />

fundamental studies <strong>of</strong> the structures and functions <strong>of</strong> membrane proteins, to physiological questions in excitable systems<br />

including the mammalian cardiovascular, muscular, nervous and respiratory systems – all the way up to studies <strong>of</strong> health, exercise<br />

and disease in humans. The <strong>Institute</strong> provides an exciting and interactive environment for modern research relating to animal<br />

and human health, as well as access to “cutting-edge” research equipment, including moderate-to-high through-put protein,<br />

fluorescence and patch-clamp systems. Publication <strong>of</strong> research findings in leading international and high-pr<strong>of</strong>ile scientific<br />

journals is a strong priority for all members <strong>of</strong> the <strong>Institute</strong>. Recent successes include papers in Nature Biotechnology, Molecular<br />

Cell, EMBO Journal and Proceedings <strong>of</strong> the National Academy <strong>of</strong> Science USA. Commensurate with this activity we have about<br />

four thousand square metres <strong>of</strong> newly refurbished laboratory space in the LIGHT Building and Centres for Integrative <strong>Membrane</strong><br />

<strong>Biology</strong> and Sports & Exercise Science. We have constant interest in the next generation <strong>of</strong> researchers and invest major resources<br />

and effort in undergraduate and postgraduate education. Furthermore, space is available for recruitment <strong>of</strong> promising and<br />

established investigators and we would be pleased at any time to hear from talented and committed individuals wishing to join<br />

us. The <strong>Institute</strong>, <strong>Faculty</strong> and University as a whole operate a low-wall principle for research groupings, centres and institutes with<br />

the aim <strong>of</strong> stimulating cross-disciplinary research and integrative approaches to big scientific questions. This places the <strong>Institute</strong><br />

within a powerful scientific environment that encourages and supports (along with its many sponsors) research and researchtraining<br />

at the highest level.<br />

Where ever you are in the world – for example at a school in Leeds, in a biotech firm or from a city on the other side <strong>of</strong> the<br />

globe, if you are interested in our research areas, please consider visiting us, joining us or investing in us. You would be most<br />

welcome. We are an ambitious, productive and open research centre in a big cosmopolitan city adjacent to some <strong>of</strong> the most<br />

beautiful countryside. You can contact any <strong>of</strong> our academics directly through e-mail. For enquiries about our undergraduate<br />

and postgraduate programmes, please see information on our web pages and make contact accordingly.<br />

Pr<strong>of</strong>essor Jim Deuchars<br />

Director, IMSB<br />

Email: d.jones@leeds.ac.uk<br />

Tel: +44 (0)113 343 4272


Group<br />

Research<br />

Figure 2: Fibre orientation maps for apical slab <strong>of</strong> left<br />

ventricular free wall (left). The angle <strong>of</strong> inclination is<br />

colour-coded between -90 and +90 degrees (right).<br />

(Image Pr<strong>of</strong> A. Holden)<br />

Cardiovascular<br />

& Sports Sciences<br />

The Group is part <strong>of</strong> the University’s<br />

Multidisciplinary Cardiovascular<br />

Research Centre (MCRC), which is a<br />

large cross-faculty research organisation<br />

providing a single academic framework<br />

to foster and promote cardiovascular<br />

research across the University. Our<br />

cardiovascular researchers have already<br />

benefited from substantial investment<br />

in the form <strong>of</strong> research fellowships and<br />

PhD studentships. This process <strong>of</strong><br />

investment is continuing with the<br />

relocation, in May 2008, <strong>of</strong> many <strong>of</strong> the<br />

Group’s researchers to purpose built<br />

laboratory space.<br />

The Group serves to extend previous<br />

individual successes <strong>of</strong> its members,<br />

fostering new collaborative research<br />

directions, allowing Leeds to embrace the<br />

area <strong>of</strong> research in its broadest sense:<br />

Current themes range from molecular,<br />

cellular and computational approaches<br />

to sports-related subjects, encompassing<br />

aspects <strong>of</strong> health, fitness, exercise<br />

physiology, anatomy, biomechanics<br />

and motor control. Ultimately, these<br />

diverse approaches feed through to<br />

inform clinical practice and therapeutics.<br />

Research activities complement<br />

the Leeds <strong>Institute</strong> for Genetics and<br />

Health (LIGHT) and the Integrative<br />

<strong>Membrane</strong> <strong>Biology</strong> Research Group by<br />

fostering new and exciting cross-faculty<br />

research opportunities in areas such<br />

as cardiovascular epidemiology and<br />

protein structure studies. This strategy<br />

satisfies the University’s desire to<br />

establish coherent research units, whilst<br />

at the same time developing innovative<br />

graduate training programmes, thereby<br />

placing Leeds at the forefront <strong>of</strong> national<br />

and international cardiovascular and<br />

sports science research.<br />

During the past 5 years MCRC<br />

members have raised over £25M <strong>of</strong><br />

external grant income. This includes<br />

funding from the British Heart<br />

Foundation (BHF), British Cardiac<br />

Association, MRC, Wellcome Trust,<br />

BBSRC, EPSRC, Department <strong>of</strong> Health<br />

and Industry. In addition to >100<br />

project grants, there are currently 5<br />

major programme grants.<br />

The quality and quantity <strong>of</strong> research<br />

output from MCRC continues to<br />

increase, with publications in the top<br />

cardiovascular journals (e.g. Circulation<br />

Research, Circulation, impact factors<br />

≥ 10) and quality multidisciplinary<br />

journals (e.g. Journal <strong>of</strong> <strong>Biological</strong><br />

Chemistry, FASEB J). There is also<br />

an increasing number <strong>of</strong> high pr<strong>of</strong>ile<br />

publications in Science, Nature, Nature<br />

Biotechnology and Molecular Cell.<br />

For more information: http://www.fbs.<br />

leeds.ac.uk/institutes/imsb/<br />

Figure 1: Surface plot <strong>of</strong> a prolonged nuclear Ca2+<br />

release event detected using line-scan confocal<br />

microscopy (Image Dr D. Steele)<br />

Figure 3: Tubulin (green) and microtubule-associated<br />

protein 4 (red) immunostaining in a control myocyte<br />

(lower) and a myocyte from a rat with streptozotocininduced<br />

type 1 diabetes (upper). Scale bar is 20 μm.<br />

(Image Dr S. Calaghan)<br />

Dr Derek Steele (Group leader)<br />

Pr<strong>of</strong> Arun Holden<br />

Pr<strong>of</strong> John Colyer<br />

Pr<strong>of</strong> Ed White<br />

Dr Simon Harrison<br />

Dr Sarah Calaghan 1<br />

Dr Nicola Mutch 1<br />

Dr Keith Dilly 1<br />

Dr Olivier Bernus 1<br />

Dr Matthew Lancaster 2<br />

Dr Karen Birch 2<br />

Dr Harry Rossiter 2<br />

Dr Andrea Utley 2<br />

Dr Ronaldo Ichiyama 2<br />

Dr Jean Aaron 2<br />

Dr Neil Messenger 2<br />

1<br />

Recently appointed tenure track research fellows<br />

2<br />

Sports-related subjects<br />

Figure 4: Map <strong>of</strong> action potential conduction in sinoatrial<br />

node from 3 month old rat. Image from Dr S Jones


Group<br />

Research<br />

Integrative<br />

<strong>Membrane</strong> <strong>Biology</strong><br />

The membranes that surround cells<br />

and the compartments within them<br />

play critical roles in almost all aspects<br />

<strong>of</strong> biology, ranging from the uptake<br />

<strong>of</strong> nutrients and perception <strong>of</strong> the<br />

environment to the transmission <strong>of</strong><br />

information from one part <strong>of</strong> the body<br />

to another.<br />

The importance <strong>of</strong> membranes<br />

is highlighted by the finding that<br />

membrane proteins account for more<br />

than 20% <strong>of</strong> the human genome.<br />

<strong>Membrane</strong> dysfunction is involved in<br />

a panoply <strong>of</strong> common diseases and<br />

membrane proteins represent the<br />

targets <strong>of</strong> more than 50% <strong>of</strong> currently<br />

used therapeutic drugs. Our research<br />

in Integrative <strong>Membrane</strong> <strong>Biology</strong> aims<br />

at a fundamental understanding <strong>of</strong><br />

the molecular mechanisms underlying<br />

these roles <strong>of</strong> membranes at the levels<br />

<strong>of</strong> cells, tissues and whole organisms.<br />

Importantly, we are trying to integrate<br />

the study <strong>of</strong> individual genes and<br />

membrane proteins with investigations<br />

<strong>of</strong> information flow within and<br />

between cells, in order to understand,<br />

for example, how the neural<br />

networks involved in brain function<br />

operate. To enable such a systems<br />

biology approach, we have strong,<br />

multidisciplinary research programmes<br />

in multiple areas, many involving<br />

collaborations with researchers not only<br />

within other <strong>Institute</strong>s and Faculties at<br />

Leeds, but worldwide. For example, at<br />

the molecular level, our researchers are<br />

collaborating with other groups in the<br />

UK and Europe to solve the structures,<br />

and thus understand the mechanisms,<br />

<strong>of</strong> membrane transporters, ion channels<br />

and hormone receptors (Figure 1).<br />

Figure 1: Model <strong>of</strong> the human membrane protein GLUT1,<br />

which transports glucose across the blood-brain barrier<br />

Work on these experimentallychallenging<br />

molecules has recently<br />

been enhanced by establishment<br />

<strong>of</strong> cutting-edge facilities for highthroughput<br />

protein production and<br />

crystallisation using robotic techniques.<br />

The detailed examination <strong>of</strong> the<br />

ligand binding sites <strong>of</strong> membrane<br />

proteins necessary to inform drug<br />

design has been made possible by the<br />

development <strong>of</strong> novel solid-state NMR<br />

techniques. In parallel, automated<br />

high-throughput electrophysiological<br />

and fluorescence approaches are being<br />

used to assay membrane function. At<br />

the cellular level, researchers studying<br />

the trafficking <strong>of</strong> membranes between<br />

cellular compartments, a process which<br />

plays critical roles in neurotransmission<br />

and in the response to hormones such<br />

as insulin, are using our state-<strong>of</strong>-the-art<br />

bioimaging facilities (Figure. 2).<br />

These include confocal, deconvolution<br />

and TIRF microscopes which can<br />

be used for real-time investigations<br />

on living cells. Current research on<br />

the role <strong>of</strong> membranes in tissue and<br />

whole organism function includes<br />

electrophysiology on complex neuronal<br />

networks and use <strong>of</strong> transgenic<br />

organisms. These investigations <strong>of</strong><br />

normal physiology are complemented<br />

by studies on the role <strong>of</strong> membranes<br />

in disease, including Alzheimer’s<br />

disease, hypertension, cardiovascular<br />

disease, neuropathic pain, epilepsy<br />

and diabetes. Via such an integrative<br />

approach to membrane biology, we<br />

hope not only to gain an understanding<br />

<strong>of</strong> these key components <strong>of</strong> living<br />

organisms, but also to address some<br />

<strong>of</strong> the major healthcare problems in<br />

the UK.<br />

Figure 2: Surface location in a cultured human cell<br />

<strong>of</strong> a green fluorescent protein-labelled component <strong>of</strong><br />

the exocyst complex, which plays a critical role in the<br />

secretory pathway <strong>of</strong> eukaryotes


Stephen Baldwin<br />

BA, MA (Cambridge)<br />

PhD (Cambridge)<br />

Postdoctoral research at Dartmouth Medical School, USA<br />

Pr<strong>of</strong>essor <strong>of</strong> Biochemistry (1998-)<br />

Leader, <strong>Membrane</strong> <strong>Biology</strong> Research Group (200-)<br />

Associate Editor for UK and Europe, Molecular <strong>Membrane</strong> <strong>Biology</strong> (1993-2005)<br />

Contact: s.a.baldwin@leeds.ac.uk<br />

Molecular<br />

membrane biology<br />

We aim to understand how membrane<br />

proteins mediate the flow <strong>of</strong> molecules<br />

across cell membranes and how they<br />

are regulated by hormones. A better<br />

understanding should lead to the<br />

development <strong>of</strong> improved therapeutic<br />

drugs. Our current research falls into<br />

four main areas.<br />

Structural proteomics <strong>of</strong> membrane<br />

proteins. As part <strong>of</strong> the UK <strong>Membrane</strong><br />

Protein Structure Initiative we are<br />

developing methods for the robotic<br />

high-throughput cloning, expression and<br />

functional characterization <strong>of</strong> membrane<br />

transporters and ion channels. In<br />

particular we are working on bacterial<br />

proteins related to important human<br />

proteins (see Figure 1).<br />

Folding <strong>of</strong> membrane proteins. We<br />

are also interested in how membrane<br />

proteins achieve their final, folded state.<br />

Current work is focused on an enzyme,<br />

PagP, from the outer membrane <strong>of</strong><br />

Escherichia coli. We are using this<br />

protein as a model system to explore<br />

protein folding.<br />

Figure 1: Binding <strong>of</strong> the neurotransmitter glutamate to a<br />

human neuronal transporter, modelled on the structure<br />

<strong>of</strong> a bacterial homologue<br />

Figure 2: Immun<strong>of</strong>luorescent imaging <strong>of</strong> adenosine<br />

transporters (green) in a muscle cell from rat heart<br />

Adenosine transporters as therapeutic<br />

targets. Adenosine regulates many<br />

processes, including cardiac output,<br />

neuronal signalling and sleep.<br />

Adenosine transporters are also the<br />

route <strong>of</strong> uptake <strong>of</strong> anti-cancer and<br />

chemotherapeutic drugs. Members <strong>of</strong><br />

the transporter family responsible for<br />

adenosine transport in most human<br />

tissues were first identified and cloned<br />

in our laboratory (see Figure 2). We are<br />

studying these proteins to understand<br />

their biological roles and develop new<br />

drugs for the treatment <strong>of</strong>, for example,<br />

chronic pain and malaria.<br />

Role <strong>of</strong> the exocyst in vesicular<br />

trafficking. The exocyst is a multiprotein<br />

complex essential for<br />

membrane-trafficking processes.<br />

We are using a combination <strong>of</strong> cell and<br />

structural biological approaches (see<br />

Figure 3) to understand the molecular<br />

mechanism <strong>of</strong> this complex and how it<br />

contributes to defects in trafficking seen<br />

in disease states.<br />

Figure 3: Model <strong>of</strong> the exo70 component <strong>of</strong> the<br />

mouse exocyst complex, showing its extended,<br />

four-domain structure<br />

Funding: Wellcome Trust; British Heart<br />

Foundation; Government agencies;<br />

MRC; BBSRC; Invitrogen; Astra-Zeneca<br />

Overseas collaborators: James Young,<br />

Carol Cass (Canada)<br />

More information:<br />

http://www.bmb.leeds.ac.uk/staff/sab/<br />

Representative Publications<br />

Baldwin, SA, Yao SYM, Hyde RJ et al.<br />

(2005) Functional characterisation <strong>of</strong> novel<br />

human and mouse equilibrative nucleoside<br />

transporters (hENT3 and mENT3) located in<br />

intracellular membranes. Journal <strong>of</strong> <strong>Biological</strong><br />

Chemistry 280: 15880–15887.<br />

Barnes, K, McIntosh, E, Whetton, AD, Daley,<br />

GQ, Bentley, J & Baldwin, SA (2005) Chronic<br />

myeloid leukaemia: an investigation into<br />

the role <strong>of</strong> Bcr-Abl-induced abnormalities in<br />

glucose transport regulation. Oncogene 24:<br />

3257–3267.<br />

Fielding, AB, Schonteich, E, Matheson, J et<br />

al. (2005) Rab11-FIP3 and FIP4 interact with<br />

Arf6 and the Exocyst to control membrane<br />

traffic in cytokinesis. EMBO Journal 24:<br />

3389–3399.


Alan Bateson<br />

BSc (Brunel)<br />

PhD (Kings College, London)<br />

Postdoctoral research at MRC Molecular Neurobiology Unit, Cambridge<br />

Assistant/Associate Pr<strong>of</strong>essor in Pharmacology, Neuroscience and Psychiatry, University <strong>of</strong> Alberta, Canada<br />

Senior Lecturer (2001-); Chair, Canadian <strong>Institute</strong>s <strong>of</strong> Health Research Pharmacology and Toxicology Grants<br />

Committee (2003-2006); Council Member, British Association for Psychopharmacology, (2004-2008); Meetings<br />

Secretary, British Association for Psychopharmacology, (2007-2010)<br />

Contact: a.n.bateson@leeds.ac.uk<br />

Regulation and expression<br />

<strong>of</strong> ion channels<br />

My research centres on the mechanisms<br />

that regulate the expression and<br />

function <strong>of</strong> ion channels, particularly<br />

in the nervous system.<br />

GABA A<br />

receptors are ligand-gated ion<br />

channels responsible for the majority<br />

<strong>of</strong> fast-synaptic inhibition in the central<br />

nervous system (CNS). They are<br />

important drug targets for a variety <strong>of</strong><br />

conditions including anxiety and sleep<br />

disorders, and some forms <strong>of</strong> epilepsy.<br />

Benzodiazepines, such as diazepam<br />

(Valium), act at GABA A<br />

receptors by<br />

potentiating the actions <strong>of</strong> GABA.<br />

Tolerance and dependence develops<br />

following long-term benzodiazepine<br />

exposure. We have examined the<br />

mechanism by which such exposure<br />

produces a change in GABA A<br />

receptor<br />

expression (Figure 1). Understanding<br />

these mechanisms may allow the<br />

identification <strong>of</strong> better drugs or different<br />

drug targets.<br />

Figure 1: Schematic illustrating the mechanism by<br />

which GABA A<br />

receptor activation alters its own expression<br />

following chronic exposure to receptor activation or<br />

modulation.<br />

We recently found that GABA A<br />

receptors<br />

functionally associate with L-type Ca 2+<br />

channels. Thus, activation <strong>of</strong> excitatory<br />

GABA A<br />

receptors early in development<br />

causes a depolarization-linked Ca 2+<br />

entry via L-type Ca 2+ channels. Further,<br />

a novel inhibition <strong>of</strong> L-type Ca 2+ channel<br />

function was revealed by repeated<br />

GABA A<br />

receptor activation. We also<br />

demonstrated a role for Ca 2+ entry via<br />

L-type Ca 2+ channels in the regulation <strong>of</strong><br />

GABA A<br />

receptor gene expression.<br />

Figure 2: Cultured cerebellar granule neurons (CGNs).<br />

(A) Bright-field image. (B) CGNs filled with the Ca 2+sensitive<br />

dye Fluo4 (C) CGNs stimulated by GABA. (D)<br />

CGNs stimulated by high [K + ].<br />

In collaboration with Dr Anne King<br />

(Leeds) we have used biolistic (gene<br />

gun) gene delivery to examine the<br />

regulation <strong>of</strong> expression <strong>of</strong> the<br />

preprotachykinin-A gene promoter in<br />

cultured spinal cords. This methodology<br />

allows transcriptional analysis in intact<br />

tissue without the time and expense <strong>of</strong><br />

constructing transgenic mouse lines.<br />

TRP channels are widely expressed,<br />

including the CNS where they regulate<br />

neuronal growth cones. In collaboration<br />

with Pr<strong>of</strong>essor Beech (Leeds) we<br />

discovered that TRPC5 is activated<br />

by lysophospholipids and other lipids<br />

which may have significance for<br />

neuronal development and function.<br />

Representative Publications<br />

Bahnasi, YM, Wright, HM, Milligan, CJ, Dedman,<br />

AM, Zeng, F, Hopkins, PM, Bateson, AN, Beech,<br />

DJ (2008) Modulation <strong>of</strong> TRPC5 cation channels<br />

by halothane, chlor<strong>of</strong>orm and prop<strong>of</strong>ol. British<br />

Journal Pharmacology 153: 1505-1512.<br />

Flemming, PK, Dedman, AM, Xu, SZ et al. (2006)<br />

Sensing <strong>of</strong> lysophospholipids by TRPC5 calcium<br />

channel. Journal <strong>of</strong> <strong>Biological</strong> Chemistry 281:<br />

4977–4982.<br />

Brazier, SP, Mason, HA, Bateson, AN & Kemp,<br />

PJ (2005) Cloning <strong>of</strong> the human TASK-2<br />

(KCNK5) promoter and its regulation by<br />

chronic hypoxia. Biochemical and Biophysical<br />

Research Communications 336: 1251–1258.<br />

Hilton, KJ, Bateson, AN & King, AE (2004) A<br />

model <strong>of</strong> organotypic rat spinal slice culture<br />

and biolistic transfection to elucidate factors<br />

that drive the preprotachykinin-A promoter.<br />

Brain Research. Brain Research Reviews 46:<br />

191–203.<br />

Bateson, AN (2002) Basic pharmacologic<br />

mechanisms involved in benzodiazepine<br />

tolerance and withdrawal. Current<br />

Pharmaceutical Design 8: 5–21.


David Beech<br />

BSc 1st Class Honours Pharmacology (Manchester)<br />

PhD Pharmacology (University <strong>of</strong> London)<br />

Postdoctoral research, University <strong>of</strong> Washington, USA<br />

Pr<strong>of</strong>essor (2000–)<br />

Head <strong>of</strong> the Multidisciplinary Cardiovascular Research Centre (MCRC)<br />

Member <strong>of</strong> the Medical Research Council Population and <strong>Systems</strong> Medicine Board<br />

Member <strong>of</strong> the Wellcome Trust-DBT India Alliance’s Early Fellowships Selection Committee<br />

Contact: d.j.beech@leeds.ac.uk<br />

Ion channels in vascular disease<br />

General purpose<br />

The overall aim <strong>of</strong> the lab is to make<br />

fundamental discoveries relating to<br />

trans-membrane calcium and sodium ion<br />

movements in mammalian cells, especially<br />

regarding ion channels in cells <strong>of</strong> human<br />

vascular diseases and associated conditions.<br />

The lab aims to reveal molecular mechanisms<br />

and to determine how they integrate into<br />

cells and are regulated by chemical factors. It<br />

aims to use the information to develop novel<br />

therapeutic strategies. The lab’s current<br />

specific interests lie in chemical regulation<br />

<strong>of</strong> newly-discovered calcium-permeable<br />

channels that play roles in tissue remodeling<br />

<strong>of</strong> vascular disease and cancer. Channels <strong>of</strong><br />

interest include the TRP channels and Orai<br />

channels.<br />

Discoveries and innovations<br />

TRPC1 is a key driver <strong>of</strong> remodelling;<br />

chemical reduction <strong>of</strong> the turret by<br />

extracellular redox protein is a mechanism<br />

for channel opening; TRPC channels are<br />

transducers for responses to lipid factors,<br />

including sphingosine-1-phosphjate<br />

and oxidized phospholipids; TRPM3 is a<br />

functional vascular smooth muscle ion<br />

channel conferring reciprocal regulation<br />

by neurosterids and cholesterol; STIM1<br />

is a functional plasma membrane ion<br />

channel subunit <strong>of</strong> vascular cells; TRPM2 is<br />

a molecular basis <strong>of</strong> calcium-activated nonselective<br />

cation channels; NCS1 is a calciumsensor<br />

<strong>of</strong> TRPC5; REST transcription factor is<br />

a switch for ion channel gene regulation in<br />

remodelling; Kv1 channels control arterial<br />

tone; there are non-contractile subcellular<br />

calcium domains <strong>of</strong> arterial smooth muscle<br />

cells; regulatory and molecular features<br />

<strong>of</strong> vascular K-ATP channels are nucleotide<br />

diphosphates and Kir6.1; robotic multiwell<br />

patch-clamp recording for academic<br />

research; patch-clamp methods for intact<br />

microvessels; extracellular blocking<br />

antibodies and design strategies for use<br />

in academic research and pharmaceutical<br />

drug development; routine use <strong>of</strong> fresh<br />

human diseased tissue samples for ion<br />

channel studies.<br />

Example current projects<br />

• Integrated functions <strong>of</strong> TRPC channels in<br />

vascular smooth muscle cells<br />

• TRPM3 and sulphated steroid responses <strong>of</strong><br />

vascular smooth muscle cells<br />

• Oxidized phospholipid ionic transduction<br />

mechanism <strong>of</strong> human vascular cells<br />

Above are images from our lab: Ion channel blocking<br />

antibodies; Robotic patch-clamp for primary cells<br />

(Nanion); Novel calcium channels <strong>of</strong> vascular smooth<br />

muscle cells; Chemical screening and vascular TRPM3.<br />

Funding: Wellcome Trust, British Heart<br />

Foundation, Medical Research Council,<br />

BBSRC, AstraZeneca, Nanion<br />

More information:<br />

http://www.cardiovascular.leeds.ac.uk/<br />

staff/Beech/<br />

Lab members: Ion Channel Retreat 2010 (DJ Beech is 6th<br />

from the right)<br />

Representative Publications<br />

AL-Shawaf E et al (2010) Acute stimulation <strong>of</strong><br />

calcium-permeable TRPC5-containing channels by<br />

oxidized phospholipids. ATVB In Press<br />

Naylor J et al (2010) Pregnenolone sulphate- and<br />

cholesterol-regulated TRPM3 channels coupled to<br />

vascular smooth muscle secretion and contraction.<br />

Circulation Research In Press<br />

Milligan CJ et al (2009) Robotic multi-well planar<br />

patch-clamp for native and primary mammalian<br />

cells. Nature Protocols 4: 244-255.<br />

Li J et al (2008) Interactions, functions and<br />

independence <strong>of</strong> plasma membrane STIM1 and<br />

TRPC1 in vascular smooth muscle cells. Circulation<br />

Research 103: e97-104.<br />

Xu S et al (2008) TRPC channel activation by<br />

extracellular thioredoxin. Nature 451: 69-72.<br />

Kumar B et al (2006) Up-regulated TRPC1 channel<br />

in vascular injury in vivo and its role in human<br />

neointimal hyperplasia. Circulation Research 98:<br />

557–563<br />

Xu S et al (2006) A sphingosine-1-phosphate<br />

activated calcium channel controlling vascular<br />

smooth muscle cell motility. Circulation Research<br />

98: 1381-1389.<br />

Xu S et al (2005) Generation <strong>of</strong> functional<br />

ion channel tools by E3-targeting. Nature<br />

Biotechnology 23: 1289-1293.<br />

Cheong A et al (2005) Down-regulated REST<br />

transcription factor is a switch enabling<br />

critical potassium channel expression and cell<br />

proliferation. Molecular Cell 20: 45–52.<br />

McHugh D et al (2003) Critical intracellular Ca 2+ -<br />

dependence <strong>of</strong> Transient Receptor Potential<br />

Melastatin 2 (TRPM2) cation channel activation.<br />

Journal <strong>of</strong> <strong>Biological</strong> Chemistry 278: 11002-11006.<br />

Xu S & Beech DJ (2001) TrpC1 is a membranespanning<br />

subunit <strong>of</strong> store-operated Ca 2+ channels<br />

in native vascular smooth muscle cells. Circulation<br />

Research 88: 84-87.


Olivier Bernus<br />

MSc, PhD (Ghent, Belgium)<br />

Postdoctoral research, SUNY Upstate Medical University, USA<br />

Tenure-Track Independent Research Fellow (2007-)<br />

Contact: o.bernus@leeds.ac.uk<br />

Three-dimensional organization <strong>of</strong> cardiac<br />

electrical activity during arrhythmias<br />

My research focuses on the<br />

mechanisms underlying life-threatening<br />

cardiac arrhythmias leading to sudden<br />

cardiac death, the largest cause <strong>of</strong><br />

death in the industrialized world.<br />

Every heartbeat is triggered by electrical<br />

waves <strong>of</strong> excitation propagating through<br />

the cardiac muscle from sinus node to<br />

the ventricles. Abnormal propagation<br />

<strong>of</strong> this wave severely compromises<br />

the mechanical function <strong>of</strong> the heart<br />

and represents a major cause <strong>of</strong><br />

arrhythmias. Reentry, during which<br />

a wave <strong>of</strong> excitation repeatedly<br />

activates the cardiac muscle, is such<br />

a type <strong>of</strong> abnormal propagation and<br />

occurs during dangerous arrhythmias<br />

such as fibrillation. My laboratory<br />

utilizes both computational and<br />

experimental techniques to visualize<br />

the propagation <strong>of</strong> electrical waves<br />

through myocardium and understand<br />

the mechanisms underlying reentry<br />

and associated arrhythmias.<br />

There is compelling clinical evidence<br />

that acute myocardial ischemia<br />

occurring after coronary occlusion is<br />

one <strong>of</strong> the most important causes <strong>of</strong><br />

ventricular arrhythmias. Recently, we<br />

discovered a novel mechanism for<br />

arrhythmogenesis during early regional<br />

ischemia using a realistic computational<br />

model <strong>of</strong> ischemic myocardium.<br />

In this model, reentry occurred as a<br />

result <strong>of</strong> calcium-mediated alternating<br />

conduction blocks in the ischemic<br />

border zone. Based on this hypothesis,<br />

we have designed an experimental<br />

model <strong>of</strong> regional ischemia and are<br />

currently investigating arrhythmogenesis<br />

in this model.<br />

Optical imaging using voltage-sensitive<br />

dyes has become a powerful tool to<br />

study electrical propagation in cardiac<br />

tissue. However, poor transparency <strong>of</strong><br />

tissue has enforced surface or subsurface<br />

imaging and prevents the use<br />

<strong>of</strong> conventional optical methods to<br />

visualize the electrical waves through<br />

the thickness <strong>of</strong> the cardiac muscle.<br />

My laboratory works on the application<br />

<strong>of</strong> novel optical tomographical<br />

methods and laser scanning to probe<br />

deeper layers <strong>of</strong> the cardiac muscle<br />

and unravel the three-dimensional<br />

wave patterns underlying cardiac<br />

arrhythmias.<br />

Funding: Research Foundation –<br />

Flanders (Belgium), IWT (Belgium)<br />

Overseas collaborators: Arkady<br />

Pertsov (USA), Sasha Panfilov (The<br />

Netherlands), Henri Verschelde<br />

(Belgium)<br />

More information:<br />

http://users.ugent.be/~obernus<br />

Figure 1: Reentry in a computational model <strong>of</strong> the human<br />

ventricles.<br />

Figure 2: Three-dimensional reconstruction <strong>of</strong> a linear<br />

object using biaxial laser scanning in an experimental<br />

phantom <strong>of</strong> biological tissue.<br />

Representative Publications<br />

Wellner, M, Bernus, O, Mironov, SF, Pertsov<br />

AM (2006) Multiplicative tomography <strong>of</strong><br />

cardiac electrical activity. Phys. Med. Biol.<br />

51:44, 29-4446.<br />

Khait, VD, Bernus, O, Mironov, SF, Pertsov AM<br />

(2006) A Method for the Three-Dimensional<br />

Localization <strong>of</strong> Cardiac Electrical Activity using<br />

Optical Imaging. J. Biomed. Opt. 11:034007.<br />

Bernus, O, Zemlin, CW, Zaritsky, R et al<br />

(2005) Alternating Conduction in the Ischemic<br />

Border Zone as Precursor <strong>of</strong> Reentrant<br />

Arrhythmias: a Simulation Study. Europace 7:<br />

93-104.<br />

Bernus, O, Wilders, R, Zemlin, C et al. (2002)<br />

A computationally efficient electrophysiological<br />

model <strong>of</strong> human ventricular cells, Am. J.<br />

Physiol. - Heart and Circulatory Physiology<br />

282:H2296-H2308.


Karen Birch<br />

BSc (Hons) Movement Science: Liverpool University (1990)<br />

PhD Exercise Physiology: Liverpool John Moores University (1995)<br />

Lecturer and Senior Lecturer in Exercise Science, Manchester Metropolitan<br />

University (1993-2002)<br />

Senior Lecturer in Exercise Physiology, University <strong>of</strong> Leeds (2002-)<br />

Member <strong>of</strong> Physoc, ACSM.<br />

Female reproductive hormones,<br />

exercise and cardiovascular disease<br />

Figure 1: Cardiopulmonary exercise test<br />

My research group comprises a team<br />

<strong>of</strong> researchers with an intense focus<br />

on investigation <strong>of</strong> the interplay<br />

between female reproductive hormone<br />

fluctuation, exercise and cardiovascular<br />

health and function. This work involves<br />

collaboration with the LIGHT, Vascular<br />

Medicine at the LGI and the Academic<br />

Dept. Obstetrics and Gyneacology,<br />

St James’s University Hospital. The<br />

techniques utilised by the team include<br />

cardiopulmonary exercise tests,<br />

echocardiography for assessment <strong>of</strong><br />

left ventricular structure and function,<br />

doppler ultrasonography for assessment<br />

<strong>of</strong> haemodynamics and endothelial<br />

function and applanation tonnometry<br />

for assessment <strong>of</strong> arterial stiffness.<br />

• The hormone oestrogen has potent<br />

effects upon the vasculature that are<br />

seen to influence haemodynamics.<br />

These effects are <strong>of</strong>ten tempered by<br />

the ovarian steroid progesterone.<br />

We have investigated the influence<br />

<strong>of</strong> oestrogen and progesterone<br />

fluctuations throughout the menstrual<br />

cycle and oral contraceptive cycle<br />

upon post-exercise hypotension.<br />

Having indicated the lack <strong>of</strong> effect<br />

<strong>of</strong> exogenous hormones (Birch et<br />

al., 2002: Experimental Physiology)<br />

we have recently been the first<br />

group to highlight that, rather than<br />

increase the magnitude <strong>of</strong> postexercise<br />

hypotension, oestrogen and<br />

progesterone appears to buffer the<br />

hypotension in the late follicular and<br />

mid luteal phases <strong>of</strong> the menstrual<br />

cycle (Esformes et al. 2005: Medicine<br />

and Science in Sports and Exercise<br />

and ACSM Annual Congress, 2005).<br />

The impact <strong>of</strong> fluctuating hormones<br />

upon health and performance in<br />

premenopausal women has been<br />

highlighted further in an invited<br />

clinical review for the British Medical<br />

Journal (Birch, 2005) and a Chapter in<br />

the British Medical Association ABC <strong>of</strong><br />

Sports Medicine.<br />

• Loss <strong>of</strong> the ovarian hormones at<br />

the menopausal transition has<br />

been shown to increase the risk <strong>of</strong><br />

cardiovascular disease and type 2<br />

diabetes in women. This has been<br />

highlighted in a key note, by invitation<br />

by the Physiological Society, at both<br />

the British Association <strong>of</strong> Science<br />

Annual Congress, Dublin, (2005)<br />

and the Association for Science<br />

Education (2005). With funding<br />

from Heart Research UK we have<br />

assessed the impact <strong>of</strong> a six month<br />

exercise training programme upon<br />

risk factors for cardiovascular disease<br />

in postmenopausal women with and<br />

without type 2 diabetes. These results<br />

have been presented at EuroPrevent<br />

(a European Society <strong>of</strong> Cardiology<br />

Congress) in Athens (2006) and Madrid<br />

(2007) and the annual Congress <strong>of</strong><br />

Figure 2: Ultrasound assessment <strong>of</strong> flow<br />

mediated dilation<br />

ACSM (2007) in New Orleans. The most<br />

exciting results <strong>of</strong> these studies were<br />

that exercise training can improve<br />

endothelial function independently <strong>of</strong><br />

changes in any other CVD risk factors<br />

in these women. We have recently<br />

received BHF funding to explore this<br />

finding further by assessing the impact<br />

<strong>of</strong> exercise training upon endothelial<br />

progenitor cell function and number.<br />

Funding: European Commission FP5,<br />

Nuffield Foundation, Heart Research UK,<br />

British Heart Foundation<br />

More information:<br />

http://www.leeds.ac.uk/sports_science/<br />

staff/kb.htm<br />

Representative Publications<br />

Cubbon RM, Murgatroyd SR, Ferguson C,<br />

Bowen TS, Rakobowchuk M, Baliga V, Cannon D,<br />

Rajwani A, Abbas A, Kahn M, Birch,,KM, Porter KE,<br />

Wheatcr<strong>of</strong>t SB, Rossiter HB, Kearney MT (2010)<br />

Human exercise-induced circulating progenitor<br />

cell mobilization is nitric oxide-dependent and is<br />

blunted in South Asian men. Arterioscler Thromb<br />

Vasc Biol, 30(4): 878-884.<br />

Oxborough D, Batterham AM, Shave R, Artis N,<br />

Birch KM, Whyte G, Ainslie PN, George KP (2009)<br />

Interpretation <strong>of</strong> two-dimensional and tissue<br />

Doppler-derived strain (epsilon) and strain rate<br />

data: is there a need to normalize for individual<br />

variability in left ventricular morphology? Eur J<br />

Echocardiogr, 10(5): 677-682.<br />

Esformes JI, Norman F, Sigley J, Birch KM (2006)<br />

The influence <strong>of</strong> menstrual cycle phase upon<br />

postexercise hypotension Med Sci Sports Exerc<br />

38(3): 484-491.<br />

Birch KM (2005) ABC <strong>of</strong> sports and exercise<br />

medicine - Female athlete triad British Medical<br />

Journal, 330: 244-246.


Sarah Calaghan<br />

BSc, PhD (Leeds)<br />

Postdoctoral Research, Dept <strong>of</strong> Biochemistry & Molecular <strong>Biology</strong>, Dept <strong>of</strong> Physiology, University <strong>of</strong> Leeds<br />

British Heart Foundation Research Fellow (2000-2003)<br />

School <strong>of</strong> Biomedical Sciences University Research Fellow (2006-)<br />

<strong>Institute</strong> <strong>of</strong> <strong>Membrane</strong> and <strong>Systems</strong> <strong>Biology</strong><br />

Contact: s.c.calaghan@leeds.ac.uk<br />

Control <strong>of</strong> signalling<br />

in the cardiac cell<br />

The heart pumps blood around<br />

the body, delivering nutrients to<br />

and removing waste products from<br />

every organ. Its function is finely<br />

tuned to respond to the demands <strong>of</strong><br />

the body. My research focuses on<br />

the mechanisms which control the<br />

behaviour <strong>of</strong> individual cardiac muscle<br />

cells in the heart in response to a variety<br />

<strong>of</strong> stimuli. This information can be used<br />

to understand the function <strong>of</strong> the heart<br />

in both health and disease.<br />

The way that the heart functions in<br />

a healthy individual is a result <strong>of</strong> a<br />

balance between the stimulatory<br />

sympathetic nervous system and the<br />

inhibitory parasympathetic system.<br />

These 2 systems work through different<br />

receptors (β-adrenoceptors and<br />

muscarinic receptors), but many <strong>of</strong><br />

the components <strong>of</strong> the downstream<br />

signalling pathways are the same. I am<br />

interested in how cellular signalling<br />

is controlled to allow these receptors<br />

to produce such diverse functional<br />

responses. One structure that<br />

contributes to this is the caveola, which<br />

is a small flask shaped pocket in the<br />

cell membrane (Figure 1). Caveolae can<br />

concentrate or exclude components <strong>of</strong><br />

signalling pathways so as to modulate<br />

both the efficiency and fidelity <strong>of</strong><br />

signal transduction. Our work has<br />

revealed a central role for caveolae in<br />

compartmentalisation <strong>of</strong> cyclic AMP<br />

signalling in the adult cardiac myocyte<br />

following β2 adrenoceptor stimulation.<br />

We are currently exploring the effect <strong>of</strong><br />

drugs (statins) and diet (polyunsaturated<br />

fatty acids) on caveolar organisation,<br />

and the potential impact <strong>of</strong> this on<br />

cAMP-dependent signalling.<br />

The heart possesses a unique intrinsic<br />

ability to regulate its force <strong>of</strong> contraction<br />

in response to circulatory demand. For<br />

example, during exercise, the amount <strong>of</strong><br />

blood returning to the heart increases<br />

and stretches the cardiac muscle.<br />

This acts as a stimulus for increased<br />

contraction, allowing the chambers <strong>of</strong><br />

the heart to expel this greater volume<br />

<strong>of</strong> blood. Some <strong>of</strong> the processes which<br />

link stretch to increased contraction<br />

are not understood. I have identified a<br />

number <strong>of</strong> elements (stretch-activated<br />

channels, the NaH exchanger) which<br />

contribute to the slow phase <strong>of</strong> force<br />

increase following stretch both in single<br />

cardiac myocytes. Recent work from the<br />

laboratory has shown that caveolae are<br />

reservoirs <strong>of</strong> extra membrane recruited<br />

upon stretch in the cardiac myocyte. This<br />

has implications for membrane tension,<br />

stretch-activated channel function<br />

and thereby the mechanotransductive<br />

response <strong>of</strong> the heart.<br />

Figure 1: A caveola in the cell membrane (courtesy <strong>of</strong><br />

Tim Lee, <strong>Faculty</strong> <strong>of</strong> <strong>Biological</strong> Sciences)<br />

Figure 2: A cardiac cell from a model <strong>of</strong> type 1 diabetes<br />

stained to show the microtubular cytoskeleton<br />

Cardiac function is adversely affected by<br />

many different diseases. For example,<br />

cardiovascular complications are a major<br />

cause <strong>of</strong> disability and death in patients<br />

with diabetes. We have identified a<br />

novel change in the microtubular<br />

network (Figure 2), that may contribute<br />

to adverse alterations in cardiac cell<br />

function in a model <strong>of</strong> type 1 diabetes.<br />

Funding: British Heart Foundation,<br />

Medical Research Council<br />

Overseas collaborators:<br />

Bob Harvey (Reno, US), Jean-Yves Le<br />

Guennec (France), Chris Howarth (UAE)<br />

Representative Publications<br />

Kozera L, White E, Calaghan S (2009) Caveolae<br />

act as membrane reserves which limit<br />

mechanosensitive I(Cl,swell) channel activation<br />

during swelling in the rat ventricular myocyte.<br />

PLoS One 4: e8312<br />

Calaghan SC, Kozera L, White E (2008)<br />

Compartmentalisation <strong>of</strong> cAMP-dependent<br />

signalling by caveolae in the adult cardiac<br />

myocyte. J Mol Cell Cardiol 44: 85-92<br />

Shiels H, O’Connell A, Qureshi MA, Howarth FC,<br />

White E, Calaghan S (2006) Stable microtubules<br />

contribute to cardiac dysfunction in the<br />

streptozotocin-induced model <strong>of</strong> type 1 diabetes<br />

in the rat. J Mol Cell Biochem 294: 173-180<br />

Calaghan SC, White E (2004) Activation <strong>of</strong><br />

Na + -H + exchange and stretch-activated channels<br />

underlies the slow inotropic response to stretch<br />

in myocytes and muscle from the rat heart. J<br />

Physiol 559: 205-214


John Colyer<br />

BSc; CNAA; MPhil, London<br />

PhD, Southampton<br />

Post-doctoral work, University <strong>of</strong> Calgary, Canada<br />

British Heart Foundation lecturer, University <strong>of</strong> Leeds (1991-99)<br />

Senior lecturer (1999-2007)<br />

Pr<strong>of</strong>essor <strong>of</strong> Biotechnology, University <strong>of</strong> Leeds (2007-)<br />

Founder <strong>of</strong> Fluorescience Ltd (biotech/drug discovery company) (1998) Badrilla Ltd. (2004)<br />

Natural & Therapeutic Control<br />

<strong>of</strong> Cardiac Function<br />

The performance <strong>of</strong> many biological<br />

events is controlled through the transient<br />

chemical modification <strong>of</strong> components,<br />

or the partnering <strong>of</strong> new components<br />

in a cell. Where and when these events<br />

take place is key to the process <strong>of</strong><br />

control, and errors in these processes<br />

can lead to human disease. In my lab<br />

we are interested in the development <strong>of</strong><br />

technologies which permit observation <strong>of</strong><br />

these short-lived chemical events, and<br />

the application <strong>of</strong> these technologies<br />

to understand normal and abnormal<br />

cardiac performance (Figure 1).<br />

The chemical adaptation <strong>of</strong> a small<br />

number <strong>of</strong> influential proteins changes<br />

the performance <strong>of</strong> the heart to meet<br />

the demands <strong>of</strong> exercise and stress,<br />

but this process fails following cardiac<br />

damage leading to life threatening<br />

loss <strong>of</strong> performance <strong>of</strong> the heart. We<br />

have developed tools to examine these<br />

events by immunoassay, and within<br />

the company Badrilla Ltd., we are<br />

developing quantitative immunoassays<br />

based on novel proprietary calibration<br />

standards (Figure 2).<br />

Figure 2: Calibration standards for quantitative immunoassays<br />

We are also developing novel<br />

experimental strategies for cardiac<br />

therapy. Having identified influential<br />

components in cardiac biology, we<br />

have engineered a strategy in which<br />

the diseased component (a protein)<br />

is removed by molecular intervention<br />

and replaced with a designer version<br />

<strong>of</strong> the component which corrects the<br />

malfunction. This strategy is being<br />

developed with a cardiac protein<br />

component, but has applications<br />

across medicine & biotechnology.<br />

Funding: MRC<br />

Representative Publications<br />

Jones, P.P., Bazzazi, H., Kargacin, G.J.<br />

& Colyer, J. (2006) Inhibition <strong>of</strong> cAMPdependent<br />

protein kinase under conditions<br />

occurring in the cardiac dyad during a Ca2+<br />

transient. Biophysical Journal 91, 433-443<br />

Carter, S., Colyer, J., Sitsapesan, R. (2006)<br />

Maximum phosphorylation <strong>of</strong> the cardiac<br />

ryanodine receptor at serine-2809 by protein<br />

kinase A produces unique modifications to<br />

channel gating and conductance not observed<br />

at lower levels <strong>of</strong> phosphorylation. Circ. Res.<br />

98, 1506-1513<br />

Johnson B.R.G., Bushby, R.J., Colyer, J., &<br />

Evans, S.D. (2006) Self assembly <strong>of</strong> actin<br />

scaffolds at ponticulin containing supported<br />

phospholipids bilayers. Biophysical Journal 90,<br />

L21-23L<br />

Rodriguez, P., Bhogal, M.S. & Colyer, J.,<br />

(2003) Stoichiometric phosphorylation <strong>of</strong><br />

cardiac Ryanodine Receptor on serine-2809<br />

by protein kinase A and calmodulin-dependent<br />

kinase II. J. Biol. Chem. 278, 38593-38600<br />

Figure 1: Use <strong>of</strong> fluorescently labelled Coiled coil<br />

peptides to monitor phosphorylation


Jim Deuchars<br />

BSc Physiology, IIi, Glasgow (1988)<br />

PhD Physiology with Pr<strong>of</strong>. KM Spyer, University <strong>of</strong> London (1992)<br />

Postdoc with Pr<strong>of</strong>. AM Thomson, University <strong>of</strong> London (1992-1997)<br />

Lecturer in Physiology, Leeds, (1997) Pr<strong>of</strong>essor<br />

Contact: J.Deuchars@leeds.ac.uk<br />

Central neuronal circuits influencing cardiovascular<br />

control – from neuronal characteristics to pathway<br />

discovery and function<br />

This research is a team effort, areas<br />

<strong>of</strong> which I lead in conjunction with<br />

Sue Deuchars. We investigate the<br />

organisation and function <strong>of</strong> the parts<br />

<strong>of</strong> the brain and spinal cord that<br />

contribute to control <strong>of</strong> the autonomic<br />

nervous system. This branch <strong>of</strong> the<br />

nervous system undertakes tasks<br />

to keep our body functioning, such<br />

as control <strong>of</strong> blood pressure, heart<br />

rate, breathing and digestion. The<br />

current team comprises postdoctoral<br />

and postgraduate researchers who<br />

use CNS slice electrophysiology,<br />

neuronal tracing, molecular biology,<br />

immunohistochemistry and light<br />

and electron microscopy. This<br />

interdisciplinary approach allows us to<br />

examine issues from several angles,<br />

leading to us being able to:<br />

l Identify new CNS regions that may<br />

be involved in autonomic control, for<br />

example a nucleus in the brainstem<br />

that receives afferent input from<br />

neck muscle proprioceptors and<br />

which projects to the nucleus tractus<br />

solitarius, a region pivotal in central<br />

autonomic neurocircuitry (Edwards<br />

et al., 2007., J Neurosci. 2007<br />

27(31):8324-33).<br />

l provide evidence that the major<br />

group <strong>of</strong> inhibitory neurotransmitter<br />

receptors, GABA receptors, can be<br />

made up components from 2 (A, C)<br />

<strong>of</strong> the 3 (A,B,C) sub-types (Milligan et<br />

al., 2004, J. Neurosci, 24:9241-50).<br />

l reveal the distribution and function<br />

<strong>of</strong> ion channels contributing to the<br />

properties <strong>of</strong> specific groups <strong>of</strong> nerve<br />

cells involved in autonomic neuronal<br />

circuits – both in the brainstem<br />

(Dallas et al., 2005, J.Physiol.<br />

562:655-72) and the spinal cord<br />

(Deuchars et al., 2001, Neurosci.,<br />

106, 433-446). In each area these<br />

channels appear to mark a cell type<br />

with specific roles in influencing<br />

autonomic nervous activity. In one<br />

study (Dallas et al., 2005), we applied<br />

antibodies as ion channel specific<br />

modulators to identify the specific<br />

channel proteins contributing to<br />

neuronal behaviour (see Figure 1).<br />

Funding: Wellcome Trust; British Heart<br />

Foundation; Government agencies<br />

MRC, BBSRC.<br />

More information:<br />

http://www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.<br />

php?staff=JDeu<br />

Figure 1: A summary <strong>of</strong> the many different types <strong>of</strong><br />

neuronal proteins which antibodies have been used to<br />

target functionally, reviewed in Dallas, Deuchars and<br />

Deuchars (2005), J. Neurosci. Meths. 146(2):133-48<br />

Figure 2: the Kv3.3 potassium channel subunit (red)<br />

is localised to presynaptic terminals with SV2 (yellow is<br />

co-localised).<br />

Representative Publications<br />

Edwards IJ, Dallas ML, Poole SL, Milligan CJ,<br />

Yanagawa Y, Szabo G, Erdelyi F, Deuchars<br />

SA, Deuchars J.(2007). The neurochemically<br />

diverse intermedius nucleus <strong>of</strong> the medulla<br />

as a source <strong>of</strong> excitatory and inhibitory<br />

synaptic input to the nucleus tractus solitarii.J<br />

Neurosci. 2007 Aug 1;27(31):8324-33<br />

Dallas, M.L., Atkinson, L., Milligan, C.J.,<br />

Morris, N.P., Lewis, D.I., Deuchars, S.A. and<br />

Deuchars, J. (2005). Localisation and function<br />

<strong>of</strong> the Kv3.1b subunit in the rat medulla<br />

oblongata: focus on the nucleus tractus<br />

solitarius. Journal <strong>of</strong> Physiology, 562(Pt<br />

3):655-72<br />

Deuchars, S.A., Milligan, C.J., Stornetta, R.L.<br />

and Deuchars, J. (2005). GABAergic neurones<br />

in the central region <strong>of</strong> the spinal cord: a novel<br />

substrate for sympathetic inhibition. Journal <strong>of</strong><br />

Neuroscience, 25(5):1063-70<br />

Milligan, C.J.; Buckley, N.J.; Garret, M.,<br />

Deuchars, J. and Deuchars, S.A. (2004).<br />

Evidence for inhibition mediated by coassembly<br />

<strong>of</strong> GABAA and GABAC receptor<br />

subunits in native central neurones. Journal<br />

<strong>of</strong> Neuroscience


Susan Deuchars<br />

BSc (Cardiff)<br />

PhD (University <strong>of</strong> London)<br />

Independent Research Fellow (1997–2005)<br />

Academic Fellow (2005-, 60% FT)<br />

Editorial Board Autonomic Neuroscience: Basic and Clinical<br />

Convenor for CRAC Special Interest Group, The Physiological Society (2001-2007)<br />

Contact: s.a.deuchars@leeds.ac.uk<br />

Unravelling autonomic circuits<br />

in brainstem and spinal cord<br />

The sympathetic and parasympathetic<br />

branches <strong>of</strong> the autonomic nervous<br />

system control most <strong>of</strong> the essential<br />

homeostatic processes. Many<br />

clinical conditions are associated<br />

with abnormal autonomic function,<br />

such as hypertension and associated<br />

cardiovascular problems. One underexplored<br />

avenue for treatment is the<br />

manipulation <strong>of</strong> areas <strong>of</strong> the central<br />

nervous system involved in sympathetic<br />

and thus cardiovascular control.<br />

I lead an enthusiastic team in<br />

conjunction with Jim Deuchars to<br />

investigate properties <strong>of</strong> neuronal<br />

circuits that underlie control <strong>of</strong> the<br />

cardiovascular system.<br />

Figure 1: Spinal cord interneuron<br />

One focus <strong>of</strong> our research is<br />

characterizing the role <strong>of</strong> interneurons<br />

(small local cells in the spinal cord)<br />

in the sympathetic circuits: where<br />

they are located, which neurons link<br />

to these cells, what neurotransmitters<br />

they contain and how they talk to other<br />

cells. We have discovered a novel group<br />

<strong>of</strong> interneurons that directly inhibit<br />

sympathetic neuronal activity and may<br />

be important in mediating stress-related<br />

changes in sympathetic outflow.<br />

We have also provided the first<br />

characterization <strong>of</strong> other interneurons<br />

involved in sympathetic control<br />

and described a role for a specific<br />

potassium channel in shaping their<br />

firing properties. These ion channels<br />

are not present in other sympathetic<br />

neurons and may help to shape the<br />

pattern <strong>of</strong> outflow from the spinal cord.<br />

Since sympathetic outflow may be<br />

severely compromised during stressful<br />

events such as ischaemia, it is vital<br />

to understand how sympathetic<br />

activity may be regulated by specific<br />

modulators (such as adenosine) that<br />

are released during these events.<br />

We have highlighted how activation<br />

<strong>of</strong> functionally diverse adenosine<br />

receptors may act in a novel synergistic<br />

way to cause an overall decrease in<br />

sympathetic preganglionic activity.<br />

This mechanism may be crucial in<br />

reducing potentially dangerous levels <strong>of</strong><br />

excitability in these neurons, which are<br />

a key component in the maintenance <strong>of</strong><br />

cardiovascular homeostasis.<br />

Funding: Wellcome Trust, British Heart<br />

Foundation.<br />

Overseas collaborators: Ida Llewllyn-<br />

Smith (Flinders, Adelaide), Ruth<br />

Stornetta (University <strong>of</strong> West Virginia)<br />

More information:<br />

www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.<br />

php?staff=SAD<br />

Figure 2: Specific ion channels in interneurons enable<br />

them to fire at a fast frequency<br />

Representative Publications<br />

Deuchars, S.A. (2007) Multi-tasking in the<br />

spinal cord - do “sympathetic” interneurones<br />

work harder than we give them credit for?<br />

Journal <strong>of</strong> Physiology 580(Pt 3):723-9.<br />

Deuchars, SA, Milligan, CJ, Stornetta, RL &<br />

Deuchars, J (2005) GABAergic neurons in<br />

the central region <strong>of</strong> the spinal cord: a novel<br />

substrate for sympathetic inhibition. Journal <strong>of</strong><br />

Neuroscience 25: 1063–1070.<br />

Brooke, RE, Deuchars, J & Deuchars,<br />

SA (2004) Input specific modulation <strong>of</strong><br />

neurotransmitter release in the lateral horn<br />

<strong>of</strong> the spinal cord via adenosine receptors.<br />

Journal <strong>of</strong> Neuroscience 24: 127–137.<br />

Deuchars, SA, Brooke, RE, Frater, B<br />

& Deuchars, J (2001a) Properties <strong>of</strong><br />

interneurones in the intermediolateral<br />

cell column <strong>of</strong> the rat spinal cord: role <strong>of</strong><br />

the potassium channel subunit Kv3.1.<br />

Neuroscience 106: 433–446.<br />

Deuchars, SA, Brooke, RE & Deuchars, J<br />

(2001b) Adenosine A1 receptors reduce<br />

release from excitatory but not inhibitory<br />

synaptic inputs onto lateral horn neurons.<br />

Journal <strong>of</strong> Neuroscience 21: 6308–6320.


Keith Dilly<br />

BSc (Coventry University)<br />

MPhil (Liverpool University)<br />

PhD (University <strong>of</strong> Maryland, MD, USA)<br />

Postdoctoral Research, University <strong>of</strong> Maryland Biotechnology <strong>Institute</strong>, MD, USA<br />

Postdoctoral Research, Columbia University, NY, USA<br />

Postdoctoral Research, University <strong>of</strong> Washington, WA, USA<br />

Tenure Track Independent Research Fellow, University <strong>of</strong> Leeds<br />

Contact: k.w.dilly@leeds.ac.uk<br />

Calcium signaling<br />

in the heart<br />

My research is focused on<br />

understanding the electrical activity and<br />

mechanical function <strong>of</strong> the heart.<br />

To work as an efficient pump the<br />

heart must beat in a coordinated<br />

fashion. Cardiac muscle contraction<br />

is stimulated by electrical activity<br />

<strong>of</strong> the cardiac action potential.<br />

This complex pathway, known as<br />

excitation-contraction (E-C) coupling<br />

can be disrupted resulting in reduced<br />

pumping efficiency <strong>of</strong> the heart and<br />

heart failure. My research focuses on<br />

how electrical activity and mechanical<br />

function <strong>of</strong> the heart are coordinated<br />

and regulated – how changes in cardiac<br />

calcium signaling under normal and<br />

pathophysiological conditions may result<br />

in heart failure and how molecular<br />

mechanisms may prevent such defects.<br />

Furthering our understanding may<br />

provide useful therapeutic targets for<br />

the treatment <strong>of</strong> heart failure.<br />

Figure 1: Greater Ca 2+ signaling in Endo than in Epi<br />

cardiac myocytes<br />

Figure 2: Intimate localization <strong>of</strong> KCNQ1 and β 2<br />

-AR<br />

shown by acceptor bleaching FRET.<br />

We recently discovered regional<br />

differences in E-C coupling and calcium<br />

signalling in the heart. We also showed<br />

these regional differences in calcium<br />

signalling and E-C coupling result in<br />

differential activation <strong>of</strong> the calcineurin-<br />

NFAT pathway. This in turn causes<br />

differential expression <strong>of</strong> a cardiac<br />

potassium channel gene involved<br />

in electrical excitability <strong>of</strong> cardiac<br />

tissue. We are intent on discovering<br />

the mechanisms responsible for<br />

controlling this excitation-transcription<br />

coupling pathway, which may prove to<br />

be a ubiquitous signalling pathway <strong>of</strong><br />

excitable tissues. (Figure 1).<br />

Sympathetic nervous system-mediated<br />

control <strong>of</strong> cardiac function results<br />

from activation <strong>of</strong> the β-adrenergic<br />

signalling pathway and numerous<br />

downstream effector molecules. Cellular<br />

compartmentalization <strong>of</strong> the response<br />

to adrenergic signalling is in part the<br />

result <strong>of</strong> localized A-kinase anchoring<br />

proteins. Recently we demonstrated<br />

localization <strong>of</strong> adrenergic signalling<br />

to specific cardiac potassium channels.<br />

(Figure 2). We are interested in further<br />

understanding localization <strong>of</strong><br />

protein kinase A signalling within<br />

cardiac muscle.<br />

We recently found a rare fatal heart<br />

condition (LQT 4) can be caused by<br />

disruption <strong>of</strong> a protein (ankyrin-B) that<br />

anchors ion channels within cardiac<br />

cells. Mutation <strong>of</strong> ankyrin-B results in<br />

disrupted sub-cellular organization,<br />

altered cardiac calcium signaling and<br />

susceptibility to arrhythmia and sudden<br />

death with exercise or β-adrenergic<br />

stimulation. (Figure 3).<br />

Figure 3: Steady state action potentials recorded from<br />

AnkB (+/-) ventricular myocytes.<br />

Representative Publications<br />

Rossow, C.F., K.W. Dilly and L.F. Santana.<br />

Differential calcineurin/NFATc3 activity<br />

underlies the mouse Ito transmural gradient.<br />

(2006). Circ Res. May 26;98(10):306-13.<br />

Dilly, K.W., Charles F. Rossow, James S.<br />

Meabon, Jennifer L. Cabarrus and Luis F.<br />

Santana. Mechanisms underling variations in<br />

excitation-contraction coupling across the left<br />

ventricular free wall. (2006). J.Physiol. Apr<br />

1;572(pt1):227-41.<br />

Dilly, K.W., Kurokawa J., Terrenoire C.,<br />

Reiken S, Lederer W.J., Marks A.R. and<br />

Kass R.S. Overexpression <strong>of</strong> β2-adrenergic<br />

receptors cAMP-dependent protein kinase<br />

phosphorylates and receptor/channel colocalization.<br />

(2004). J.Biol.Chem. Sep 24;<br />

279(39): 40778-97.<br />

Mohler, Peter J., Jean-Jacques Schott,<br />

Anthony O. Gramolini, Keith W. Dilly, Silvia<br />

Guatimosim, William H. duBell, Long-Sheng<br />

Song, Karine Haurogné, Florence Kyndt,<br />

Mervat E. Ali, Terry B. Rogers, W. J. Lederer,<br />

Denis Escande, Herve Le Marec, Vann Bennett.<br />

Ankyrin-B mutation causes type 4 long QT<br />

cardiac arrhythmia and sudden cardiac death.<br />

(2003). Nature. Feb 6; 421(6923): 634-9.


Dan Donnelly<br />

BSc <strong>Biological</strong> Chemistry, University <strong>of</strong> Leicester (1988)<br />

PhD, University <strong>of</strong> London 1992, supervisor Pr<strong>of</strong> TL Blundell<br />

Post doc with Pr<strong>of</strong> J.B.C. Findlay (1991-1995)<br />

Lecturer (1995-2004) & Senior Lecturer (2004-), University <strong>of</strong> Leeds<br />

Contact: d.donnelly@leeds.ac.uk<br />

G Protein-Coupled<br />

Receptors<br />

My research group studies the structure<br />

& function <strong>of</strong> G protein-coupled<br />

receptors - one <strong>of</strong> the most diverse<br />

and ubiquitous families <strong>of</strong> integral<br />

membrane proteins. GPCRs play a<br />

pivotal role in many cellular signalling<br />

pathways and are prime targets for<br />

the development <strong>of</strong> therapeutic agents<br />

designed to either block or activate the<br />

receptors. The aim <strong>of</strong> this laboratory is<br />

to elucidate the mechanism by which<br />

these receptors bind their ligands<br />

and transduce the signal across the<br />

plasma membrane.<br />

The control <strong>of</strong> the body’s blood sugar<br />

level requires keeping an intricate<br />

balance between the levels and<br />

actions <strong>of</strong> the two opposing pancreatic<br />

hormones, insulin and glucagon. While<br />

low glucose levels result in glucagon<br />

secretion from pancreatic alpha cells, in<br />

the high glucose situation the action <strong>of</strong><br />

glucose on pancreatic beta cells results<br />

in increased plasma insulin levels.<br />

However, high blood glucose levels are<br />

not solely responsible for increased<br />

insulin secretion. Two hormones,<br />

glucagon-like peptide-1 (GLP-1) and<br />

glucose-dependent-insulinotropic<br />

polypeptide (GIP), are responsible<br />

sensing food intake and consequently<br />

sensitizing the pancreatic beta cells’<br />

insulin secretory system to glucose.<br />

Using truncated and mutated receptors<br />

alongside modified peptide ligands, our<br />

group has defined a two-stage model for<br />

peptide binding at the GLP-1 receptor<br />

(Al-Sabah & Donnelly, 2003; Lopez de<br />

Maturana et al. 2003).<br />

The GLP-1 work in my laboratory<br />

is currently funded by an Industrial<br />

Partnership award from BBSRC &<br />

AstraZeneca, part <strong>of</strong> which involves<br />

the design & synthesis <strong>of</strong> small<br />

molecules ligands (Dr. Colin Fishwick,<br />

School <strong>of</strong> Chemistry).<br />

As part <strong>of</strong> a collaboration with GSK, our<br />

group also study the calcitonin receptorlike<br />

receptor (e.g. Miller et al., 2010) and<br />

the receptors for parathyroid hormone<br />

(e.g. Mann et al., 2008).<br />

Current Funding: BBSRC; GSK,<br />

AstraZeneca<br />

Past Funding: Novo Nordisk, Knoll, Royal<br />

Society, BBSRC, Wellcome<br />

Trust, British Heart Foundation<br />

and Diabetes UK.<br />

More information:<br />

www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.<br />

php?staff=DD<br />

Figure 1: Schematic figure <strong>of</strong> how GLP-1 bind to its<br />

receptor (left) and a competition binding experiment<br />

using three different ligands at the GLP-1 receptor<br />

expressed in HEK-293 cells (right).<br />

Representative Publications<br />

Mann RJ, Nasr NE, Sinfield JK, Paci E, Donnelly<br />

D (2010) The major determinant <strong>of</strong> exendin-4/<br />

GLP-1 differential affinity at the rat GLP-1<br />

receptor N-terminal domain is a hydrogen bond<br />

from SER-32 <strong>of</strong> exendin-4. Brit. J. Pharmacol doi:<br />

10.1111/j.1476-5381.2010.00834.x<br />

Miller PS, Barwell J, Poyner DR, Wigglesworth<br />

MJ, Garland SL, Donnelly D (2010) Non-peptidic<br />

antagonists <strong>of</strong> the CGRP receptor, BIBN4096BS<br />

and MK-0974, interact with the calcitonin<br />

receptor-like receptor via methionine-42 and<br />

RAMP1 via tryptophan-74. Biochem Biophys Res<br />

Commun 391(1): 437-442<br />

Mann R, Wigglesworth MJ, Donnelly D (2008)<br />

Ligand-receptor interactions at the parathyroid<br />

hormone receptors: subtype binding selectivity is<br />

mediated via an interaction between residue 23<br />

on the ligand and residue 41 on the receptor. Mol<br />

Pharmacol 74(3): 605-613<br />

Al-Sabah S, Donnelly D (2003) A model for<br />

receptor-peptide binding at the glucagon-like<br />

peptide-1 (GLP-1) receptor through the analysis<br />

<strong>of</strong> truncated ligands and receptors British Journal<br />

<strong>of</strong> Pharmacology 140: 339-346<br />

Lopez de Maturana R, Willshaw A, Kuntzsch<br />

A, Rudolph R, Donnelly D (2003) The isolated<br />

N-terminal domain <strong>of</strong> the glucagon-like<br />

peptide-1 receptor binds exendin peptides<br />

with much higher affinity than GLP-1 Journal <strong>of</strong><br />

<strong>Biological</strong> Chemistry 278: 10195-10200


John Findlay<br />

B.Sc. (1st Hons.): Biochemistry, University <strong>of</strong> Aberdeen (1968)<br />

Ph.D: Biochemistry, University <strong>of</strong> Leeds (1972)<br />

Post-doctoral: Harvard University, USA<br />

Pr<strong>of</strong>essor <strong>of</strong> Biochemistry: University <strong>of</strong> Leeds (1990-)<br />

Contact: j.b.c.findlay@leeds.ac.uk<br />

The <strong>Membrane</strong> Protein<br />

and Proteomics Group<br />

The main interest <strong>of</strong> this laboratory is to<br />

examine the structure and mechanism<br />

<strong>of</strong> action <strong>of</strong> membrane proteins.<br />

Research focuses on receptor systems<br />

(G protein coupled receptors (GPCRs)<br />

and membrane receptors for lipocalins),<br />

ion channels (proton-transporting<br />

channels and ligand-regulated Cachannel),<br />

the proteomics <strong>of</strong> mast<br />

cell and stem cell differentiation<br />

and bioinfomatics. There are also<br />

investigations into mutations in GPCRs<br />

and proteins <strong>of</strong> the visual system<br />

which give rise to human disease.<br />

The principle techniques used,<br />

dependent on the exact project, include<br />

protein chemistry, electrophysiology,<br />

molecular biology, protein mutation<br />

and expression, general membranology<br />

and biophysical analysis (NMR, X-<br />

ray, EM). There are a number <strong>of</strong><br />

ongoing collaborations with other<br />

groups in Leeds, elsewhere in the UK<br />

and abroad. The laboratory houses<br />

a protein chemistry facility which<br />

includes automated protein sequencing<br />

and robotic proteomics utilizing mass<br />

spectrometry.<br />

Current projects include:<br />

l Studies <strong>of</strong> the Structure:Function<br />

<strong>of</strong> G protein coupled receptors<br />

including aspects <strong>of</strong> their folding,<br />

assembly and quarternary structure.<br />

l The development <strong>of</strong> new biosensor<br />

and assay systems by exploiting<br />

native and novel GPCRs expressed in<br />

reporting systems <strong>of</strong> various kinds.<br />

This project will not only develop<br />

novel sensor / assay systems but will<br />

reveal how novel specificities arise in<br />

GPCR structures<br />

l<br />

l<br />

l<br />

l<br />

Structure:Function <strong>of</strong> lipocalin<br />

receptors, their expression,<br />

reconstitution and functional<br />

characteristics.<br />

The role and mechanism <strong>of</strong> action <strong>of</strong><br />

the RBP-receptor system and its role<br />

in Type2 Diabetes<br />

The proteomics <strong>of</strong> embryonic and<br />

adult stem cells and the pathways<br />

involved in differentiation to mature<br />

cell types.<br />

Drug discovery projects to address<br />

insulin resistance and genetic<br />

disease in membrane proteins<br />

Funding: Research supported by<br />

BBSRC, Wellcome Trust, EC, MRC,<br />

Government Depts.<br />

More Information:<br />

www.fbs.leeds.ac.uk/staff/findlay/<br />

Figure 1: A model <strong>of</strong> the proposed role <strong>of</strong> RBP, CRBP<br />

and their receptor in the uptake <strong>of</strong> retinol (vitamin A) into<br />

the cell<br />

Representative Publications<br />

Wrigley J D J, Ahmed T, Nevett C L, Findlay<br />

J B C. Peripherin/rds influences membrane<br />

vesicle morphology. J Biol Chem , Vol 275,<br />

No18, 13191-13194 2000.<br />

Bhogal N, Blaney FE, Ingley PM, Rees and<br />

Findlay JBC. The proximity <strong>of</strong> the extreme<br />

N-terminus <strong>of</strong> the NK2 tachykinin receptro to<br />

Cys167 in the putative fourth transmembrane<br />

helix. Biochemistry, 43, 3027-3038, 2004<br />

Blades MJ, Ison JC, Ranasinghe R, Findlay JBC.<br />

Automatic generation and evaluation <strong>of</strong> sparse<br />

protein signatures for families <strong>of</strong> protein structural<br />

domains. Protein Science 14,13-23, 2005<br />

Clare DK, Orlova EV, Finbow MA, et al. An<br />

expanded and flexible form <strong>of</strong> the vacuolar<br />

ATPase membrane sector. Structure, 14 (7),<br />

1149-1156 (2006)<br />

Redondo C, Vourapolou M, Evans J and Findlay<br />

JBC. The identification <strong>of</strong> the Retinol Binding<br />

Protein interaction site and functional state <strong>of</strong><br />

RBPs for the membrane receptor. Invited by<br />

FASEB J. (2007). In Press.


Nikita Gamper<br />

MS (St. Petersburg State University)<br />

PhD (<strong>Institute</strong> <strong>of</strong> Evolutionary Physiology and Biochemistry, St. Petersburg)<br />

Postdoctoral research, Tübingen University, Germany<br />

and UT Health Science Center at San Antonio, TX, USA<br />

Lecturer in Neuroscience (2005-)<br />

Contact: n.gamper@leeds.ac.uk<br />

Ion channels and regulation<br />

<strong>of</strong> cellular excitability<br />

Separation <strong>of</strong> electrical charges on the<br />

cell membrane is necessary for cell-tocell<br />

communication. Such separation<br />

is achieved by a coordinated work <strong>of</strong><br />

different ion channels, transporters and<br />

pumps. Accordingly, dysfunction <strong>of</strong><br />

ion channels causes human disease,<br />

including epilepsy and sudden<br />

cardiac death. We use cutting-edge<br />

biophysical, biochemical, molecular<br />

and cell-biological approaches to study<br />

ion channels and their involvement in<br />

regulation <strong>of</strong> cellular excitability.<br />

One focus <strong>of</strong> our research is the Kv7<br />

K + channels that control neuronal<br />

excitability and are involved in cardiac<br />

arrhythmias, epilepsy and deafness.<br />

In one project we recently showed the<br />

oxidative modification and upregulation<br />

<strong>of</strong> neuronal M channels by reactive<br />

oxygen species (ROS). Since ROS are<br />

produced during hypoxia or ischaemia<br />

in the brain, oxidative modification <strong>of</strong> M<br />

channels represents a mechanism for<br />

‘neuronal silencing’ in the precarious<br />

time when neurons may die. We study<br />

oxidative modification using cloned Kv7<br />

channels expressed in an immortalized<br />

cell line and native M channels<br />

in cultured primary neurons and<br />

organotypic hippocampal slice cultures.<br />

Other Kv7-related projects are focused<br />

on the regulation <strong>of</strong> channel trafficking,<br />

assembly at the plasma membrane and<br />

retrieval for degradation. We also study<br />

regulation <strong>of</strong> Kv7 channels by their<br />

auxiliary subunits.<br />

We are also interested in the regulation<br />

<strong>of</strong> neuronal ion channels by G-proteincoupled<br />

receptors (GPCRs), and how the<br />

specificity <strong>of</strong> GPCR-triggered signalling<br />

is achieved in a single neuron. The<br />

project is based on the recent discovery<br />

<strong>of</strong> signalling specificity among Gq/11-<br />

coupled receptors in the regulation <strong>of</strong><br />

neuronal ion channels. Although coupled<br />

to similar signalling pathways, different<br />

receptor types have different effects.<br />

Recently we proposed a hypothesis<br />

<strong>of</strong> receptor-specific phospholipid<br />

signals (Figure 1) to account for such<br />

remarkable specificity, but further work<br />

is needed. We plan to study receptorspecific<br />

regulation <strong>of</strong> K + and Ca 2+<br />

channels in sensory neurons, where<br />

such regulation is critical for pain<br />

sensation. Figure 2 shows measurement<br />

<strong>of</strong> bradykinin receptor activation by<br />

the translocation <strong>of</strong> a GFP-tagged<br />

membrane-localized PIP 2<br />

-sensitive<br />

probe, delivered to sensory neurons<br />

<strong>of</strong> trigeminal ganglia by a biolistic<br />

‘Gene Gun’.<br />

Funding: AHA<br />

Figure: 2<br />

Representative Publications<br />

Gamper, N, Li, Y & Shapiro, SM (2005)<br />

Structural requirements for subunit-specific<br />

modulation <strong>of</strong> KCNQ K + channels by Ca 2+ /<br />

Calmodulin. Molecular <strong>Biology</strong> <strong>of</strong> the Cell 16:<br />

3538–3551.<br />

Gamper, N, Reznikov, V, Yamada, Y, Yang, J<br />

& Shapiro, MS (2004) PIP 2<br />

signals underlie<br />

receptor-specific G q/11<br />

-mediated modulation <strong>of</strong><br />

N-type Ca 2+ channels. Journal <strong>of</strong> Neuroscience<br />

24: 10980–10992 (see also subsequent<br />

review by Delmas, Coste, Gamper & Shapiro<br />

(2005) Neuron 47: 179–182).<br />

Gamper, N & Shapiro, MS (2003) Calmodulin<br />

mediates Ca 2+- dependent modulation <strong>of</strong><br />

KCNQ2/3 potassium channels. Journal <strong>of</strong><br />

General Physiology 122: 17–31.<br />

Huber, SM, Uhlemann, A-C, Gamper, NL,<br />

Duranton, C, Kremsner, PG & Lang, F (2002)<br />

Plasmodium falciparum activates endogenous<br />

Cl - channels <strong>of</strong> human erythrocytes by<br />

membrane oxidation. EMBO Journal 21:<br />

22–30.<br />

Figure: 1


Michael Harrison<br />

BSc Hons, PhD (Leeds)<br />

Lecturer in Biochemistry<br />

Contact: M.A.Harrison@leeds.ac.uk<br />

Structure and function<br />

<strong>of</strong> proton pumps<br />

The current focus <strong>of</strong> my work is on the<br />

vacuolar H + -ATPase, a large protein<br />

complex that uses energy from ATP<br />

to pump protons across biological<br />

membranes. This ‘acid pump’ is found in<br />

virtually all eukaryotic cells, playing key<br />

roles in the function <strong>of</strong> endomembranes.<br />

In osteoclastic bone cells, some kidney<br />

epithelial and some tumour cells, it is<br />

also found at the plasma membrane,<br />

where acts to pump acid out <strong>of</strong> the cell.<br />

Loss-<strong>of</strong>-function mutations in the genes<br />

encoding subunits <strong>of</strong> the V-ATPase<br />

can result in kidney disease, hereditary<br />

deafness or the bone thickening disease<br />

osteopetrosis, and because <strong>of</strong> its<br />

involvement in the processes <strong>of</strong> bone<br />

resorption and tumour metastasis, the<br />

V-ATPase has also attracted attention as<br />

a possible drug target.<br />

My interest in this key protein covers<br />

several areas: in structural biology, my<br />

lab is characterising the structures <strong>of</strong> the<br />

individual polypeptides that make up the<br />

V-ATPase and the contacts, both static<br />

and dynamic, that they make with each<br />

other. This is allied to advanced methods<br />

in electron microscopy which can provide<br />

detailed images <strong>of</strong> individual V-ATPase<br />

molecules trapped in different states<br />

(see picture). We are also asking questions<br />

about the site <strong>of</strong> binding and mechanism<br />

<strong>of</strong> action <strong>of</strong> V-ATPase inhibitors, answers<br />

to which may help in the design <strong>of</strong> new,<br />

more specific inhibitors with therapeutic<br />

potential.<br />

In the area <strong>of</strong> cell biology, work in the lab<br />

is focused on two questions: Firstly, how<br />

is the V-ATPase regulated in response to<br />

physiological demands? Secondly, how is<br />

location <strong>of</strong> the V-ATPase within the cell<br />

controlled? Relocation to the plasma<br />

membrane occurs during maturation<br />

<strong>of</strong> the osteoclasts and in some forms <strong>of</strong><br />

tumour cell in response to extracellular<br />

signals, requiring changes in the<br />

interactions between V-ATPase and the<br />

cytoskeleton. The signalling mechanisms<br />

that control this process remain<br />

uncertain, but understanding them may<br />

be a crucial factor in the design <strong>of</strong> new<br />

drugs that prevent bone degeneration.<br />

Figure 2 (clockwise from top right):<br />

Osteoclasts dissolve bone as part <strong>of</strong> the<br />

normal cycle <strong>of</strong> skeleton repair. Cultured<br />

in the laboratory, they will attack the<br />

surface <strong>of</strong> bone wafers, providing a bone<br />

diseases model. Osteoclasts dissolve<br />

bone by forming an ‘acid bath’ on its<br />

surface – this requires the activity <strong>of</strong> the<br />

V-ATPase. Both natural and synthetic<br />

compounds inhibit the V-ATPase by<br />

binding to a specific region at the<br />

interface between two key membrane<br />

subunits. As a consequence, such<br />

compounds can act to block the process<br />

<strong>of</strong> bone resorption.<br />

Funding: Work in the lab is currently<br />

supported by BBSRC, and in the past by<br />

the European Union and Wellcome Trust.<br />

Figure 1<br />

Figure 2<br />

Representative Publications<br />

Jones RPO, Durose LJ, Phillips C, Keen JN,<br />

Findlay JBC & Harrison MA (2010) A site-directed<br />

cross-linking approach to the characterization <strong>of</strong><br />

subunit E-subunit G contacts in the vacuolar H + -<br />

ATPase stator. Mol. Memb. Biol. in press<br />

Muench SP, Huss M, Song CF, Phillips C,<br />

Wieczorek H, Trinick J & Harrison MA (2009)<br />

Cryo-electron microscopy <strong>of</strong> the vacuolar ATPase<br />

motor reveals its mechanical and regulatory<br />

complexity. J. Mol. Biol. 386: 989-999<br />

Kóta Z, Páli, T, Dixon N, Kee T, Harrison M, Findlay<br />

JBC, Finbow ME & Marsh D (2008) Incorporation<br />

<strong>of</strong> transmembrane peptides from the vacuolar<br />

H + -ATPase in phospholipid membranes: spinlabel<br />

electron paramagnetic resonance and<br />

polarised infrared spectroscopy. Biochemistry 47:<br />

3937-3949<br />

Dixon N, Pali T, Kee TP, Ball SK, Harrison MA,<br />

Findlay JBC, Nyman J, Väänänen K, Finbow ME<br />

& Marsh D (2008) Interaction <strong>of</strong> spin-labelled<br />

inhibitors <strong>of</strong> the vacuolar H + -ATPase with the<br />

transmembrane V o<br />

-sector. Biophys. J. 94: 506–514<br />

Duarte AMS, Wolfs CJAM, Van Nuland NAJ,<br />

Harrison MA, Findlay JBC, Van Mierlo CPM &<br />

Hemminga MA (2007) Structure and localization<br />

<strong>of</strong> an essential transmembrane segment <strong>of</strong> the<br />

proton translocation channel <strong>of</strong> yeast H + -V-<br />

ATPase. Biophys. Biochim. Acta (Biomembranes)<br />

1768: 218-227<br />

Clare DK, Orlova EV, Finbow ME, Harrison MA,<br />

Findlay JBC & Saibil HR (2006) An expanded and<br />

flexible form <strong>of</strong> the vacuolar ATPase membrane<br />

sector. Structure 14: 1149-1156


Simon Harrison<br />

BSc (Leeds), PhD (Glasgow)<br />

Postdoctoral work with Pr<strong>of</strong> D Bers, University <strong>of</strong> California (1986-1989)<br />

Convenor <strong>of</strong> HCM special interest group <strong>of</strong> the Physiological Society (2003-)<br />

Senior Lecturer (1999-)<br />

Contact: s.m.harrison@leeds.ac.uk<br />

Heart function in health<br />

and disease<br />

My research interests lie in<br />

understanding the mechanisms <strong>of</strong><br />

normal excitation-contraction coupling<br />

in the heart, how these processes are<br />

regulated and how a variety <strong>of</strong> ‘disease<br />

states’ (hypertrophy, sepsis, etc) affect<br />

the strength <strong>of</strong> contraction <strong>of</strong> the heart.<br />

High blood pressure or ‘hypertension’<br />

affects 10 million people in the<br />

UK (http://en.wikipedia.org/wiki/<br />

Hypertension). In hypertensive patients<br />

the heart has to work harder to pump<br />

blood around the body and this<br />

causes the walls <strong>of</strong> the heart to thicken<br />

(hypertrophy). Eventually hypertrophy<br />

can lead to heart failure and associated<br />

with this progression are changes at<br />

the cellular level in the way heart cells<br />

contract. We have been studying the<br />

mechanisms associated with altered<br />

contraction and calcium regulation in<br />

normal and hypertrophied heart cells. In<br />

collaboration with Pr<strong>of</strong> Ed White’s group<br />

we also compare ‘bad’ hypertrophy<br />

(above) with ‘good’ hypertrophy which<br />

occurs in athletes involved in endurance<br />

training. We aim to understand why<br />

bad hypertrophy leads to heart failure<br />

whereas good hypertrophy does not.<br />

Sepsis (http://en.wikipedia.org/wiki/<br />

sepsis) is a potentially life-threatening<br />

condition associated with the release<br />

<strong>of</strong> inflammatory cytokines like tumour<br />

necrosis factor (TNF). These cytokines<br />

have direct inhibitory effects on the<br />

heart and contribute to cardiovascular<br />

complications. We have shown that<br />

combinations <strong>of</strong> TNF and interleukin-1ß<br />

dramatically increase the leakiness <strong>of</strong> the<br />

internal store <strong>of</strong> calcium so that it cannot<br />

contribute properly during the heart<br />

beat. In the right hand panel <strong>of</strong> Figure 1<br />

Figure 1<br />

the increased number <strong>of</strong> bright spots<br />

(‘sparks’) compared to the left hand<br />

panel represents increased leak <strong>of</strong><br />

calcium from the store induced by these<br />

cytokines (see 2010 paper Cell Calcium).<br />

One consequence <strong>of</strong> severe sepsis is<br />

cell necrosis or death, and the contents<br />

<strong>of</strong> dying cells can be released into the<br />

vicinity <strong>of</strong> healthy cells. We are currently<br />

investigating the effects <strong>of</strong> histones (the<br />

normal function <strong>of</strong> which is to organise<br />

DNA in the nucleus) on cardiac function.<br />

Our initial experiments suggest that<br />

histones H3 and H4 induce very<br />

deleterious effects on the regulation<br />

<strong>of</strong> cytosolic calcium and therefore<br />

contractility in isolated ventricular<br />

myocytes. We aim to characterise their<br />

effects and identify a novel means <strong>of</strong><br />

inhibiting these proteins which may<br />

prove therapeutically useful in patients<br />

suffering from severe sepsis.<br />

Funding sources for these projects<br />

include: The British Heart Foundation,<br />

Wellcome Trust, Royal Society and the<br />

Medical Research Council.<br />

Representative Publications<br />

Duncan DJ, Yang Z, Hopkins PM, Steele DS &<br />

Harrison SM (2010) TNF-α and IL-1β increase<br />

Ca2+ leak from the sarcoplasmic reticulum and<br />

susceptibility to arrhythmia in rat ventricular<br />

myocytes. Cell Calcium 47: 378-386<br />

Stones R, Billeter R, Zhang H, Harrison SM &<br />

White E (2009) The role <strong>of</strong> transient outward<br />

K+ current in electrical remodelling induced by<br />

voluntary exercise in female rat hearts. Basic<br />

Research in Cardiology 104: 643-652.<br />

Stones R, Natali A, Billeter R, Harrison SM & White<br />

E (2008) Voluntary exercise-induced changes<br />

in β2-adrenoceptor signalling in rat ventricular<br />

myocytes. Experimental Physiology 93: 1065-<br />

1075<br />

Fowler MR, Naz JR, Graham MD, Orchard CH &<br />

Harrison SM (2007) Age and hypertrophy alter<br />

the contribution <strong>of</strong> sarcoplasmic reticulum and<br />

Na+/Ca2+ exchange to Ca2+ removal in rat left<br />

ventricular myocytes. Journal <strong>of</strong> Molecular and<br />

Cellular Cardiology 42: 582-589


Peter Henderson<br />

BSc, PhD (Bristol); MA, ScD (Cambridge)<br />

Postdoctoral research, University <strong>of</strong> Madison, USA, Lecturer, University <strong>of</strong> Leicester (1973-75)<br />

Lecturer, University <strong>of</strong> Cambridge (1975-90), Visiting Research Pr<strong>of</strong>essor, Jichi, Japan (1982-83)<br />

Reader, University <strong>of</strong> Cambridge (1990-92), Pr<strong>of</strong>essor <strong>of</strong> Biochemistry and Molecular <strong>Biology</strong><br />

University <strong>of</strong> Leeds (1992-), Canadian Commonwealth Research Fellow (1993), Dean <strong>of</strong> Research<br />

<strong>Faculty</strong> <strong>of</strong> <strong>Biological</strong> Sciences (1998-2002), Leverhulme Senior Research Fellow (2003-05)<br />

Scientific Director, European <strong>Membrane</strong> Protein consortium (EMeP) (2005-)<br />

Contact: p.j.f.henderson@leeds.ac.uk<br />

<strong>Membrane</strong> transport<br />

and antibiotic action<br />

I am interested in how cells transport<br />

nutrients, wastes and antibiotics<br />

across the essentially impermeable<br />

cell membrane.<br />

All cells are bounded by a lipid<br />

bilayer membrane that is inherently<br />

impermeable to the majority <strong>of</strong><br />

hydrophilic solutes required for cell<br />

nutrition and to many <strong>of</strong> the waste<br />

products and toxins that must be<br />

excreted. Accordingly, the membrane<br />

contains proteins, the sole function <strong>of</strong><br />

which is to catalyse the translocation <strong>of</strong><br />

substrates through the membrane. The<br />

structure-activity relationships <strong>of</strong> these<br />

proteins are difficult to elucidate because<br />

they are <strong>of</strong> low natural abundance in the<br />

membrane; they are very hydrophobic;<br />

and, even when purified, they are very<br />

difficult to crystallize, which is just the<br />

beginning <strong>of</strong> determining their molecular<br />

mechanism <strong>of</strong> operation.<br />

As approximately 5-15% <strong>of</strong> all proteins,<br />

revealed by the current efforts in genome<br />

sequencing, are membrane transport<br />

proteins vital for the capture <strong>of</strong> nutrients<br />

(Figure 1), and hence the first stage<br />

in cell growth, there is an urgent need<br />

in the new millennium to determine<br />

their structures. Their additional roles<br />

in antibiotic resistance, toxin secretion,<br />

respiration, and ATP synthesis in bacteria<br />

(Figure 1), and neurotransmission,<br />

kidney function, intestinal absorption,<br />

tumour growth and other diverse cell<br />

functions in man presage a major<br />

investigative effort to elucidate their<br />

molecular mechanisms <strong>of</strong> action.<br />

We concentrate on bacterial transport<br />

proteins homologous to human<br />

transporters, and employ recombinant<br />

DNA methods to amplify their expression<br />

and genetically engineer ‘tags’ to<br />

facilitate purification and make mutants<br />

that illuminate their activities. A wide<br />

range <strong>of</strong> physical techniques are then<br />

applied, including fluorimetry, circular<br />

dichroism, mass spectrometry, EPR,<br />

X-ray crystallography and NMR.<br />

We have cloned over 50 transport<br />

proteins. Many, especially transporters<br />

<strong>of</strong> antibiotics and sugars, have been<br />

purified. Several form 3d crystals and<br />

diffract, and structures should result<br />

soon. We attract collaborators from<br />

abroad, and have well funded projects<br />

with scientists in Europe and in Glasgow,<br />

London and Southampton.<br />

Funding: BBSRC, EPSRC, EU,<br />

Wellcome Trust, Novartis<br />

Overseas collaborators in Belgium,<br />

France, Japan, Germany, Holland,<br />

Norway, Portugal, Sweden.<br />

Figure 1: General scheme <strong>of</strong> bacterial transport reactions<br />

Representative Publications<br />

Suzuki, S and Henderson, P.J.F. (2006)<br />

“The hydantoin transport protein from<br />

Microbacterium liquefaciens” J. Bacteriol.<br />

188, 3329-3336.<br />

Clough, J., Saidijam, M., Bettaney K.E.,<br />

Szakonyi, G., Meuller, J., Suzuki, S., Bacon,<br />

M., Barksby, E., Ward, A., Gunn-Moore, F.,<br />

O’Reilly, J., Rutherford, N.G., Bill, R.M.<br />

and Henderson, P.J.F. (2006) “Prokaryote<br />

membrane transport proteins: amplified<br />

expression and purification”. Structural<br />

genomics <strong>of</strong> membrane proteins (ed.<br />

Lundstrom, K.) pp.21-42. CRC Press, USA.<br />

Saidijam, M., Benedetti, G., Ren, Q., Zhiqiang,<br />

X., Hoyle, C.J., Palmer, S.L., Ward, A.,<br />

Bettaney, K.E., Szakonyi, G., Meuller, J.,<br />

Morrison, S., Pos, M.K., Butaye, P, Walravens,<br />

K., Langton, K., Herbert, R.B., Skurray, R.A.,<br />

Paulsen, I.T., O’Reilly, J., Rutherford, N.G.,<br />

Brown, M.H., Bill, R.M. and Henderson, P.J.F.<br />

(2006) “Microbial Drug Efflux Proteins <strong>of</strong> the<br />

Major Facilitator Superfamily”, in Current Drug<br />

targets 7, pp. 793-811.<br />

Patching, S.G., Herbert, R.B., O’Reilly, J.,<br />

Brough, A.R. and Henderson, P.J.F. (2004)<br />

“Low 13C-background for NMR-based studies<br />

<strong>of</strong> ligand binding using 13C-depleted glucose<br />

as carbon source for microbial growth”. J. Am.<br />

Chem. Soc. 126, 86-87.


Arun Holden<br />

BA (Oxford, UK)<br />

PhD (Alberta, Canada) has been at Leeds since 1971, and is currently Pr<strong>of</strong>essor <strong>of</strong> Computational <strong>Biology</strong>,<br />

and Deputy Director, Centre for Nonlinear Studies, and is an editor <strong>of</strong> several nonlinear science journals<br />

and two book series on Nonlinear Science.<br />

Contact: arun@cbiol.leeds.ac.uk<br />

Computational<br />

<strong>Biology</strong><br />

(a) The Computational <strong>Biology</strong> laboratory<br />

constructs detailed computer models<br />

<strong>of</strong> muscular organs - the beating heart,<br />

and the pregnant uterus - that are based<br />

on data from membrane, cell and tissue<br />

experiments, and from clinical data.<br />

These models are validated by their<br />

prediction <strong>of</strong> normal organ behaviour,<br />

and abnormal behaviours seen in<br />

disease. They are applied to dissect,<br />

in space and time, physiological and<br />

pathological mechanism, to prescreen<br />

drugs, and to contribute to the drug<br />

design and discovery process, and the<br />

design <strong>of</strong> physical interventions.<br />

(b) Most <strong>of</strong> our research has been in the<br />

construction <strong>of</strong> cardiac virtual tissues,<br />

and these are now being applied to<br />

biomedical problems. Application areas<br />

include the normal pacemaking <strong>of</strong> the<br />

heart and its pharmacological control;<br />

the genetic engineering <strong>of</strong> pacemaking<br />

activity in the ventricles, as an alternative<br />

to implanted electronic pacemakers;<br />

the initiation and control <strong>of</strong> re-entrant<br />

arrhythmias in the atria and ventricles;<br />

the effects <strong>of</strong> drugs on propagation<br />

phenomena within the heart; and the<br />

effects <strong>of</strong> electrical and very large<br />

magnetic fields on the behaviour <strong>of</strong> the<br />

in situ heart. The same methodology, <strong>of</strong><br />

developing and applying virtual tissues,<br />

is being applied to model uterine activity<br />

and possible mechanisms in premature<br />

and normal labour.<br />

(c) An important new development has<br />

been the extraction <strong>of</strong> structural data,<br />

(geometry, cell orientation) from diffusion<br />

tensor magnetic resonance imaging <strong>of</strong><br />

tissues; this allows the reconstruction <strong>of</strong><br />

a 3-D detailed digital map <strong>of</strong> an organ in<br />

a few days, rather than the months-years<br />

necessary for histological reconstruction.<br />

Funding: Computational <strong>Biology</strong> Leeds<br />

is funded by project, programme and<br />

network grants from the BBSRC, EPSRC,<br />

MRC, BHF, EU and Sun Microsystems<br />

Figure 1: Reconstructed geometry <strong>of</strong> gravid human<br />

uterus, side view<br />

Figure 2: Simulation <strong>of</strong> electrical activity producing a<br />

contraction in a near full term uterus, front view<br />

Figues 3 & 4: Reconstructed ventricular geometry,<br />

and visualsiation <strong>of</strong> electrical activity during<br />

simulated fibrillation<br />

Figure 4<br />

Representative Publications<br />

Tong WC, Holden AV. (2005) Induced<br />

pacemaker activity in virtual mammalian<br />

ventricular cells, Lecture Notes in Computer<br />

Science, 3504: 226-235<br />

Zhang HG, Garratt CJ, Zhu JJ, et al. (2005)<br />

Role <strong>of</strong> up-regulation <strong>of</strong> I-K1 in action<br />

potential shortening associated with atrial<br />

fibrillation in humans. Cardiovascular Research<br />

66 (3): 493-502<br />

Holden AV. (2005) The sensitivity <strong>of</strong> the<br />

heart to static magnetic fields. Progress in<br />

Biophysics and Molecular biology, 87:<br />

289-320<br />

Aslanidi OV, Clayton RH, Lambert JL,<br />

Holden AV. (2005) Dynamical and cellular<br />

electrophysiological mechanisms <strong>of</strong> ECG<br />

changes during ischaemia. J.Theoretical<br />

biology 237: 369-381.


Ronaldo Ichiyama<br />

BSc Lic Universidade Estadual de Campinas (Brazil)<br />

MSc, PhD University <strong>of</strong> Illinois at Urbana-Champaign (USA)<br />

Postdoctoral Fellow University <strong>of</strong> California Los Angeles (USA)<br />

Contact: r.m.ichiyama@leeds.ac.uk<br />

More information: http://www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.php?tag=Ichiyama_RM<br />

Structural and Functional Plasticity in the CNS activity<br />

dependent plasticity in the spinal cord<br />

Neural Control <strong>of</strong> Movement<br />

The mechanisms related to the neural<br />

control <strong>of</strong> movement in mammalian<br />

systems are largely unanswered, simply<br />

because the intricacies <strong>of</strong> interneuronal<br />

communication and computations<br />

necessary to produce coordinated,<br />

voluntary movements are <strong>of</strong> such<br />

complexity that we have only begun<br />

to understand them. Relative to the<br />

overwhelming complexity <strong>of</strong> the<br />

supraspinal control <strong>of</strong> movement, the<br />

spinal neural circuits provide a simpler<br />

model to investigate motor control<br />

issues. This does not, however, imply<br />

that spinal control <strong>of</strong> movement in<br />

mammalian species is a simple model.<br />

My research has focused on the changes<br />

within the spinal cord that occur after<br />

a complete spinal cord transection and<br />

on the activity-dependent plasticity<br />

in the spinal neural circuits associated<br />

with locomotor training after the spinal<br />

cord injury. We have used behavioural,<br />

immunohistochemical, electron<br />

microscopic and electrophysiologic<br />

methods to investigate those issues.<br />

The combined results strongly suggest<br />

that the biochemical, structural, and<br />

electrophysiological properties <strong>of</strong><br />

motoneurons change dramatically<br />

within the spinal cord isolated from the<br />

brain, which are altered by locomotor<br />

training. Understanding these processes<br />

in detail will provide critical insight into<br />

the mechanisms involved in the control<br />

and learning <strong>of</strong> movements within<br />

spinal cord neural circuits.<br />

Neural Control <strong>of</strong> Cardiorespiratory<br />

Function in Exercise<br />

Another interest in my lab is the neural<br />

control <strong>of</strong> cardiovascular function<br />

related to movement production and<br />

exercise. During muscular activity, two<br />

basic mechanisms control cardiovascular<br />

adjustments: a central command and<br />

a reflex arising from the contracting<br />

muscles, which is dependent on<br />

medullary centers. I have investigated<br />

issues related to both <strong>of</strong> those<br />

mechanisms. We have implicated higher<br />

cortical centers (insular cortex) in the<br />

central command circuitry. In addition,<br />

I have demonstrated that specific<br />

locomotor and cardiovascular control<br />

areas in the brain change with exercise<br />

training. The posterior hypothalamus<br />

nucleus, the mesencephalic locomotor<br />

region, periaqueductal grey, nucleus<br />

<strong>of</strong> the tractus solitarius and the<br />

rostral ventrolateral medulla showed<br />

diminished, possibly more efficient,<br />

activation pr<strong>of</strong>iles (cfos) in exercised<br />

than in non-exercised rats in response to<br />

a single bout <strong>of</strong> controlled exercise.<br />

Spinal Cord Injury, Neural<br />

Regeneration and Functional<br />

Recovery<br />

After a spinal cord injury, the remaining<br />

unaffected spinal tissue changes to<br />

a great extent. A successful neural<br />

regenerative strategy will have to<br />

overcome not only all the obstacles<br />

that the injury site itself presents (glial<br />

scaring, physical gap, etc.) but also<br />

a new environment that has formed<br />

below the level <strong>of</strong> the lesion. We<br />

hypothesize that the beneficial effects<br />

<strong>of</strong> locomotor training will potentiate the<br />

effects <strong>of</strong> regenerative strategies. We<br />

have combined locomotor training with<br />

different potential neural regenerative<br />

strategies, with both positive and<br />

surprising results. Given the nature <strong>of</strong><br />

spinal cord injuries, we firmly believe<br />

that only a combination strategy will be<br />

effective in regenerating and forming<br />

functional synaptic reconnections after<br />

an injury.<br />

We have developed a technique to<br />

epidurally stimulate the spinal cord,<br />

which produces alternating and<br />

coordinated steps in completely<br />

spinalized adult rats. This technique<br />

allows us to study the control <strong>of</strong><br />

locomotion in an in vivo adult<br />

mammalian preparation, which was not<br />

possible previously. We have used this<br />

technique to investigate reflex control<br />

mechanisms in the intact spinal cord and<br />

also after a complete spinal transection.<br />

Motoneurones (green) activated (red nuclei) during locomotion,<br />

visualized by immun<strong>of</strong>luorescence techniques.<br />

Representative Publications<br />

Maier IC*, Ichiyama RM*, Courtine G* et al. (2009)<br />

Differential effects <strong>of</strong> anti-Nogo –A antibody<br />

treatment and treadmill training in rats with<br />

incomplete spinal cord injury. Brain 132:1426-40<br />

*these authors contributed equally<br />

Ichiyama R et al. (2009) Enhanced motor function<br />

by training in spinal cord contused rats following<br />

radiation therapy. PLoS One 4(8): e6862<br />

Courtine G et al. (2009) Transformation <strong>of</strong><br />

nonfunctional spinal circuits into functional<br />

states after the loss <strong>of</strong> brain input. Nature<br />

Neuroscience 12(10): 1333-1342<br />

Ichiyama RM et al. (2008) Step Training Reinforces<br />

Specific Spinal Locomotor Circuitry in Adult<br />

Spinal Rats. Journal <strong>of</strong> Neuroscience 28(29):7370-<br />

7375<br />

Ichiyama RM et al. (2008) Dose Dependence <strong>of</strong><br />

the 5-HT Agonist Quipazine in Facilitating Spinal<br />

Stepping in the Rat with Epidural Stimulation.<br />

Neuroscience Letters 438 (3): 281-285


Lars Jeuken<br />

BSc/MSc, Utrecht University, the Netherlands (1990-1995)<br />

PhD, Leiden University, the Netherlands (Promotor: Pr<strong>of</strong>. G.W. Canters) (1995-1999)<br />

Postdoctoral research, University <strong>of</strong> Oxford, UK (Supervisor: Pr<strong>of</strong>. F.A. Armstrong) (1999-2002)<br />

Postdoctoral research, (Supervisor: Pr<strong>of</strong>. S.D. Evans) (2002), BBSRC David Phillips Research Fellow, University <strong>of</strong> Leeds,<br />

UK (2002-2007)<br />

Senior Lecturer, University <strong>of</strong> Leeds, UK (2007-)<br />

Contact: l.j.c.jeuken@leeds.ac.uk<br />

Transmembrane charge transport<br />

and biological electron transfer<br />

The lab is active in the development <strong>of</strong><br />

new tools to elucidate charge transport<br />

mechanisms <strong>of</strong> membrane proteins and<br />

enzymes. Charge transport through<br />

membranes is an ubiquitous and key<br />

process in – for instance - metabolism,<br />

photosynthesis, active membrane<br />

transport and signal transduction.<br />

The ultimate aim is to understand the<br />

charge transport activities <strong>of</strong> membrane<br />

proteins in mechanistic and – where<br />

structural information is available -<br />

molecular detail. A method developed<br />

at the University <strong>of</strong> Leeds allows a<br />

biological membrane (containing the<br />

protein <strong>of</strong> interest) to be ‘tethered’<br />

to a solid surface which can act as<br />

an electrode (i.e. solid supported<br />

membranes). Using electrochemical<br />

systems based on this technique we<br />

are trying to create a generation <strong>of</strong><br />

electrodes that are widely applicable to<br />

a range <strong>of</strong> membrane proteins.<br />

We are also interested in the<br />

mechanisms <strong>of</strong> action <strong>of</strong> redox proteins<br />

and enzymes, both membrane and<br />

glubular. Redox proteins function<br />

in transporting electrons between<br />

proteins and oxidase and reduce a<br />

wide range <strong>of</strong> substrates. By optimising<br />

electrode design and combining<br />

electrochemistry with fluorescence<br />

techniques we aim to obtain functional<br />

information about reaction mechanisms<br />

for transmembrane enzymes, like<br />

cytochrome c oxidase, and globular<br />

enzymes, like the copper-containing<br />

nitrite reductase.<br />

Objectives<br />

One objective is to design and create<br />

novel electrodes that can be used<br />

to study a wide range <strong>of</strong> membrane<br />

proteins. In order to do this we need to<br />

understand the structural properties <strong>of</strong><br />

electrodes that can electrically interact<br />

with the membrane proteins. By<br />

thorough surface characterisation using<br />

microscopy and spectroscopy we hope<br />

to improve existing electrode designs.<br />

Secondly, we use these new tools to<br />

study catalytic mechanisms <strong>of</strong> redoxactive<br />

and transport proteins in their<br />

native environment.<br />

More information: http://www.fbs.leeds.<br />

ac.uk/staff/pr<strong>of</strong>ile.php?tag=Jeuken_LJC<br />

Representative Publications<br />

Kendall JKR, Johnson BRG, Symonds PH,<br />

Imperato G, Bushby RJ, Gwyer JD, van Berkel<br />

C, Evans SD, Jeuken LJC (2010) Effect <strong>of</strong> the<br />

Structure <strong>of</strong> Cholesterol-Based Tethered Bilayer<br />

Lipid <strong>Membrane</strong>s on Ionophore Activity.<br />

ChemPhysChem 11: 2191-2198. DOI: 10.1002/<br />

cphc.200900917<br />

Jeuken LJC (2009) Electrodes for integral<br />

membrane enzymes. Nat. Prod. Rep. 26: 1234-<br />

1240. DOI:10.1039/B903252E<br />

Weiss SA, Bushby RJ, Evans SD, Henderson<br />

PJF, Jeuken LJC (2009) Characterisation <strong>of</strong><br />

cytochrome bo3 activity in a native-like surfacetethered<br />

membrane. Biochem. J. 417: 555-560.<br />

DOI:10.1042/BJ20081345<br />

Jeuken LJC, Weiss SA, Henderson PJF, Evans SD,<br />

Bushby RJ (2008) Impedance Spectroscopy <strong>of</strong><br />

Bacterial <strong>Membrane</strong>s: Coenzyme-Q Diffusion in a<br />

Finite Diffusion Layer. Anal. Chem. 80: 9084-9090.<br />

DOI:10.1021/ac8015856<br />

Wijma HJ, Jeuken LJC, Verbeet MP, Armstrong<br />

FA, Canters GW (2007) Protein Film Voltammetry<br />

<strong>of</strong> Copper-containing Nitrite Reductase reveals<br />

Reversible Inactivation. J. Am. Chem. Soc. 129:<br />

8557-8565. DOI: 10.1021/ja071274q<br />

Jeuken LJC, Connell SD, Henderson PJF, Gennis<br />

RB, Evans SD, Bushby RJ (2006) Redox Enzymes<br />

in Tethered <strong>Membrane</strong>s. J. Am. Chem. Soc. 128:<br />

1711-1716


Lin-Hua Jiang<br />

BSc and MSc, East China Normal University, P.R. China<br />

PhD with D Wray, University <strong>of</strong> Leeds<br />

Postdoc with RA North, University <strong>of</strong> Sheffield<br />

Wellcome Trust University Award (2004)<br />

Contact: L.H.Jiang@leeds.ac.uk<br />

Structure, function and regulation<br />

<strong>of</strong> ion channels<br />

Our research aims to understand<br />

molecular mechanisms determining the<br />

expression, function and regulation <strong>of</strong><br />

Ca2+ permeable ion channels and their<br />

roles in cellular Ca2+ signalling, with<br />

particular interest in ATP-gated P2X<br />

receptors and TRP channels.<br />

P2X receptors are homo/hetero-trimers<br />

and function as extracellular ATPgated<br />

channels with multifarious roles,<br />

including fertility, pain sensation, taste<br />

and immune response. We are working<br />

at two leading forefronts. One is to<br />

understand where the ATP-binding<br />

site is, how ATP binding is coupled to<br />

channel opening and how the open<br />

pore permeates cations and the other is<br />

to elucidate the molecular or structural<br />

basis specifying interactions <strong>of</strong> P2X7<br />

receptors with selective antagonists<br />

and modulators so as to search for<br />

specific and potent human P2X7<br />

receptor antagonists for therapeutic<br />

use in rheumatoid arthritis and other<br />

inflammatory diseases.<br />

Figure 1<br />

TRPM2 is a melastatin-related member<br />

<strong>of</strong> transient receptor potential channel<br />

superfamily. TRPM2 channel is homotetrameric<br />

assembly and opens upon<br />

binding <strong>of</strong> intracellular ADP-ribose.<br />

TRPM2 channel activation also occurs<br />

in response to oxidative stress, and<br />

thus TRPM2 channel act as a cellular<br />

redox potential sensor. Important<br />

physiological and particularly<br />

pathophysiological roles have emerged,<br />

extending from insulin release, cytokine<br />

production, endothelial barrier<br />

dysfunction, to neuronal destruction.<br />

In collaboration with Pr<strong>of</strong>. DJ Beech<br />

and other colleagues at Leeds, we are<br />

studying the molecular mechanisms<br />

controlling TRPM2 channel expression,<br />

assembly, function and regulation and<br />

its role in the pathogenesis <strong>of</strong> oxidative<br />

stress-related diseased conditions<br />

such as neurodegenerative and<br />

cardiovascular diseases.<br />

Funding: BBSRC, Wellcome Trust, Royal<br />

Society and Alzheimer’s Research Trust.<br />

Representative Publications<br />

Browne LE, Jiang L-H and North RA (2010) New<br />

structure enlivens interest in P2X receptors.<br />

Trends Pharmacol. Sci. 31: 229-237<br />

Roger S, Mei Z, Baldwin JM, Bradley H, Dong L,<br />

Baldwin SA, Surprenant A and Jiang L-H (2010)<br />

Single nucleotide polymorphisms that were<br />

identified in affective mood disorders affect ATPactivated<br />

P2X7 receptor functions. J. Psychiatr.<br />

Res. 44: 347-355<br />

Milligan CJ, Li J, Sukumar P, Majeed Y, Dallas<br />

ML, English A, Emery P, Porter KE, Smith AM,<br />

McFadzean I, Beccano-Kelly D, Bahnasi Y, Cheong<br />

A. Naylor J, Zeng F, Liu X, Gamper N, Jiang L-H,<br />

Pearson HA, Peers C, Robertson B and Beech DJ<br />

(2009) Robotic multi-well planar patch-clamp for<br />

native and primary mammalian cells. Nat. Protoc.<br />

4: 244-255<br />

Liu X, Ma W, Surprenant A and Jiang L-H (2009)<br />

Identification <strong>of</strong> extracellular domain residues <strong>of</strong><br />

rat P2X7 receptor involved in functional inhibition<br />

by acidic pH. Br. J. Pharmacol. 156: 139-142<br />

Li J, Sukumar P, Milligan CJ, Kumar B, Ma Z,<br />

Munsch MC, Jiang L-H, Porter KE and Beech BJ<br />

(2008) Interactions, functions and independence<br />

<strong>of</strong> plasma membrane STIM1 and TRPC1 in<br />

vascular smooth muscle cells. Circ. Res. 103:<br />

e97-104<br />

Xia R, Mei Z, Mao H, Yang W, Dong L, Bradley H,<br />

Beech DJ and Jiang L-H (2008) Identification <strong>of</strong><br />

pore residues engaged in determining divalent<br />

cationic permeation in transient receptor<br />

potential melastatin subtype channel. J. Biol.<br />

Chem. 283: 27426-27432<br />

Xu SZ, Sukumar P, Zeng F, Li J, Jairaman A,<br />

English A, Naylor J, Ciurtin C, Majeed Y, Milligan<br />

CJ, Bahnasi YM, Al-Shawaf E, Porter KE, Jiang<br />

L-H, Emery P, Sivaprasadarao A and Beech DJ<br />

(2008) TRPC channel activation by extracellular<br />

thioredoxin. Nature 451: 69-72<br />

Liu X, Surprenant A, Mao HJ, Roger S, Xia R,<br />

Bradley H and Jiang L-H (2008) Identification <strong>of</strong><br />

key residues coordinating functional inhibition<br />

<strong>of</strong> P2X7 receptors by zinc and copper. Mol.<br />

Pharmacol. 73: 252-259


Anne King<br />

BSc (Aberdeen)<br />

PhD (Southampton)<br />

MRC Post-doctoral Research Fellowship, St Bartholomews HMC, London<br />

Wellcome Trust Research Fellow, University College London<br />

Lecturer & Senior Lecturer, University <strong>of</strong> Leeds<br />

Reader, <strong>Institute</strong> <strong>Membrane</strong> & <strong>Systems</strong> <strong>Biology</strong> Affiliations, Neuroscience, Integrative & <strong>Membrane</strong> <strong>Biology</strong><br />

Contact: a.e.king@leeds.ac.uk<br />

Spinal Mechanisms <strong>of</strong> Somatosensation,<br />

Pain and Analgesia<br />

The focus <strong>of</strong> our work is modulation<br />

<strong>of</strong> somatosensory processing in the<br />

spinal dorsal horn with an emphasis<br />

on synaptic transmission between<br />

peripheral sensory afferents and<br />

target spinal neurons that underpin<br />

nociception (pain). From a clinical<br />

perspective, the effective treatment <strong>of</strong><br />

chronic debilitating pain represents a<br />

huge scientific challenge. An important<br />

aspect <strong>of</strong> our research, undertaken<br />

with industrial collaborators, is to<br />

identify novel targets for development<br />

<strong>of</strong> new-generation analgesics. Our work<br />

provides an understanding <strong>of</strong> the basic<br />

neurobiology and neuropharmacology<br />

<strong>of</strong> pain at the level <strong>of</strong> spinal cord and<br />

periphery, building a platform for drug<br />

discovery and therapeutic advances for<br />

clinical pain management. Current work<br />

includes the following.<br />

(1) Adenosine and nucleoside<br />

transporters in spinal cord: the<br />

nucleoside adenosine has potent antinociceptive<br />

actions that may contribute<br />

to the body’s endogenous analgesic<br />

(pain-relieving) system. Its intracellular<br />

and extracellular distribution is critically<br />

dependent on CNT and ENT transporter<br />

proteins. We are currently defining the<br />

function and distribution <strong>of</strong> ENT and<br />

CNT nucleoside transporter subtypes<br />

in normal spinal cord and in models <strong>of</strong><br />

persistent (inflammatory) pain.<br />

(2) Rhythmicity in spinal dorsal horn<br />

sensory processing: rhythmicity<br />

underpins many basic central nervous<br />

system functions, such as sensory<br />

binding and motor output. In the dorsal<br />

horn, we are characterizing synaptic<br />

(GABA-ergic) and non-synaptic (gapjunction)<br />

mechanisms <strong>of</strong> synchronized<br />

dorsal-horn ensemble activity in vitro<br />

and determining novel modulatory<br />

influences <strong>of</strong> analgesic drug families<br />

(3) The trigeminal system and or<strong>of</strong>acial<br />

pain: the mechanisms <strong>of</strong> usedependent<br />

synaptic plasticity in the<br />

trigeminal system that occur as a<br />

consequence <strong>of</strong> prolonged neuronal<br />

activation (e.g. chronic dental and facial<br />

pain). In this joint project with Pr<strong>of</strong>essor<br />

Boissonade (Sheffield) the aim is to<br />

investigate stimulation–transcription<br />

coupling and the signalling cascades<br />

that are activated following prolonged<br />

nociceptive afferent input. We are<br />

examining these mechanisms in the<br />

trigeminal nucleus using molecular<br />

markers (Fos, pERK, CREB) <strong>of</strong> neuronal<br />

activation and plasticity.<br />

Figure 1 Schematic model for regulation<br />

<strong>of</strong> ‘pain signaling’ by ENT1 nucleoside<br />

transporters in spinal dorsal horn. In<br />

substantia gelatinosa (SG), glutamate<br />

(GLU) excites ‘nociceptive’ neurons<br />

that signal ‘pain’ inputs. Adenosine<br />

(ADO) elicits anti-nociception via presynaptic<br />

receptors (A1R), an effect<br />

that is mimicked by blocking the ENT1<br />

transporter. The opioid receptor (MOR)<br />

induces release <strong>of</strong> ADO via modulation<br />

<strong>of</strong> ENT1.<br />

Funding: Wellcome Trust, BBSRC,<br />

GlaxoSmithKline.<br />

More information:<br />

www.fbs.leeds.ac.uk/staff/king/<br />

Figure 1<br />

Representative Publications<br />

Governo, RJM, Deuchars, J, Baldwin, SA &<br />

King, AE (2005) Localisation <strong>of</strong> the NBMPRsensitive<br />

equilibrative nucleoside transporter<br />

ENT1 in the rat dorsal root ganglia and lumbar<br />

spinal cord. Brain Research 1059: 129–138.<br />

Asghar, AU, Cilia La Corte, PF, LeBeau,<br />

FE et al. (2005) Oscillatory activity within<br />

rat substantia gelatinosa in vitro: a role for<br />

chemical and electrical neurotransmission.<br />

Journal <strong>of</strong> Physiology (London) 562: 183–198.<br />

Worsley, MA, Todd, AJ & King, AE (2005)<br />

Serotoninergic-mediated inhibition <strong>of</strong><br />

substance P-sensitive deep dorsal horn<br />

neurons: a combined electrophysiological and<br />

morphological study in vitro. Experimental<br />

Brain Research 160: 360–367.<br />

Ackley, MA & King, AE (2005) Adenosine<br />

contributes to μ-opioid synaptic inhibition in<br />

rat substantia gelatinosa in vitro. Neuroscience<br />

Letters 376: 102–106.


Matthew Lancaster<br />

BSc (Leeds)<br />

PhD (Liverpool)<br />

Postdoctoral research at Leeds<br />

Lecturer in Exercise Physiology (2003-)<br />

Contact: m.k.lancaster@leeds.ac.uk<br />

Cardiac Ageing Leads to<br />

Cardiac Problems<br />

My group’s research is directed towards<br />

understanding what happens to our<br />

hearts as we age. Age is the single<br />

biggest risk factor for developing cardiac<br />

problems but unlike several other known<br />

risk factors for cardiac problems such<br />

as diet and blood pressure ageing is<br />

currently unavoidable, unfortunately<br />

you can’t just give up ageing! By<br />

studying ageing <strong>of</strong> the heart though<br />

we are seeking to identify the specific<br />

mechanisms whereby ageing renders the<br />

heart susceptible to problems and assess<br />

the potential for suitable interventions to<br />

optimise ageing <strong>of</strong> the heart.<br />

Two major issues affecting the ageing<br />

heart are a steadily reducing maximal<br />

heart rate associating with a reduction<br />

in maximal functional capacity and<br />

accompanying this an increasing<br />

susceptibility to cardiac arrhythmias.<br />

These heart rhythm control problems<br />

can lead to an increasing intolerance to<br />

exercise/exertion and also mean the large<br />

majority <strong>of</strong> artificial pacemaker implants<br />

are performed in patients over the age<br />

<strong>of</strong> 65 because their normal cardiac<br />

pacemaker appears to have begun to fail.<br />

Every beat <strong>of</strong> the heart begins in a cell<br />

such as that shown in figure 1. We have<br />

shown that with ageing the electrical<br />

coupling <strong>of</strong> these cells to the rest <strong>of</strong> the<br />

heart begins to deteriorate and they<br />

also begin to change their expression <strong>of</strong><br />

several ion channels that are normally<br />

responsible for their electrical activity.<br />

Disconnected and dysfunctional this is<br />

why we believe the pacemaker <strong>of</strong> the<br />

heart begins to fail with age. Knowledge<br />

<strong>of</strong> this is already beginning to raise<br />

questions regarding how we treat cardiac<br />

conditions in patients with an aged heart<br />

but we are also expanding this work<br />

investigating the effects <strong>of</strong> restoring<br />

connections and modifying excitability in<br />

an attempt to restore youthful function.<br />

Of course it’s unfortunately not just the<br />

cardiac pacemaker that ages we have<br />

found plenty <strong>of</strong> ageing related changes<br />

happening elsewhere in the heart. Some<br />

<strong>of</strong> these such as changes in intracellular<br />

calcium regulation could predispose to<br />

arrhythmias and damage in the event <strong>of</strong><br />

a heart-attack. We have been looking at<br />

how to limit or reverse age-associated<br />

problems using exercise, acute or<br />

life-long. The effects have been pretty<br />

interesting with exercise affecting the<br />

aged heart more than the young heart in<br />

terms <strong>of</strong> changes in gene expression but<br />

unfortunately not all these changes may<br />

be beneficial and regrettably exercise<br />

does not prove to reverse ageing <strong>of</strong> the<br />

heart. Continuation <strong>of</strong> this work will<br />

however <strong>of</strong>fer further insight and how to<br />

prevent ageing <strong>of</strong> the heart turning into<br />

failing <strong>of</strong> the heart.<br />

Funding: Wellcome Trust and the<br />

Strategic Promotion <strong>of</strong> Ageing Research<br />

Capacity, a BBSRC/EPSRC joint venture<br />

Overseas collaborators: Haruo Honjo<br />

(Japan)<br />

More information:<br />

www.fbs.leeds.ac.uk/staff/lancaster/<br />

Figure 1: A typical pacemaker cell.<br />

Representative Publications<br />

Jones SA, Yamamoto M, Tellez JO, Billeter-Clark<br />

R, Boyett MR, Honjo H & Lancaster MK (2008)<br />

Distinguishing properties <strong>of</strong> cells from the<br />

myocardial sleeves <strong>of</strong> the pulmonary veins; a<br />

comparison <strong>of</strong> normal and abnormal pacemakers.<br />

Circulation: Arrhythmia & Electrophysiology 1:<br />

39-48<br />

Jones SA, Boyett MR., Lancaster MK. (2007)<br />

Declining into failure: The age-dependent loss <strong>of</strong><br />

the L-type calcium channel within the sinoatrial<br />

node. Circulation 115: 1183-90<br />

Lancaster MK, Jones SA, Harrison SM & Boyett MR<br />

(2004) Intracellular Ca 2+ and pacemaking within<br />

the rabbit sinoatrial node: heterogeneity <strong>of</strong> role<br />

and control. Journal <strong>of</strong> Physiology 556: 481–494<br />

Jones SA, Lancaster MK & Boyett MR (2004)<br />

Ageing-related changes <strong>of</strong> connexins and<br />

conduction within the sinoatrial node. Journal <strong>of</strong><br />

Physiology 560: 429–437<br />

Honjo H, Inada S, Lancaster MK et al. (2003)<br />

Sarcoplasmic reticulum Ca 2+ release is not a<br />

dominating factor in sinoatrial node pacemaker<br />

activity. Circulation Research 92: 41–44


Jonathan Lippiat<br />

BSc (Leicester)<br />

PhD (Leicester)<br />

Postdoctoral research at Oxford<br />

Junior Research Fellow, Linacre College Oxford, Lecturer (2004-)<br />

Contact: j.d.lippiat@leeds.ac.uk<br />

Electrophysiology and molecular<br />

biology <strong>of</strong> ion channels<br />

Our research explores the molecular<br />

and cellular properties <strong>of</strong> ion<br />

channels, how they can be targeted<br />

by drugs, and why particular genetic<br />

variations cause disease. A number <strong>of</strong><br />

laboratory techniques are employed:<br />

electrophysiology, protein biochemistry,<br />

DNA manipulation, optical imaging<br />

and FRET.<br />

CLC-5 chloride channel<br />

These proteins are found in the kidney<br />

and are involved in the reabsorption <strong>of</strong><br />

protein, minerals and vitamins back into<br />

the body from the glomerular filtrate.<br />

Loss <strong>of</strong> functional CLC-5 channels<br />

by inherited genetic mutation causes<br />

Dent’s disease, resulting in kidney<br />

stones and eventual kidney failure.<br />

Our studies will help understand why<br />

very small changes in particular regions<br />

<strong>of</strong> the chloride channel structure<br />

(Figure 1) can affect protein function<br />

and result in disease.<br />

Intracellular ion-regulated<br />

potassium channels<br />

We are studying a family <strong>of</strong> ion<br />

channels activated by intracellular<br />

ion concentrations, enabling them to<br />

regulate cellular events. Members <strong>of</strong><br />

this family <strong>of</strong> channels are differentially<br />

sensitive to calcium, magnesium,<br />

sodium (Figure 2) and chloride ions.<br />

Pharmacology<br />

Ion channels are interesting targets<br />

for drug action in a wide range <strong>of</strong><br />

diseases. Activation <strong>of</strong> potassium<br />

channels in smooth muscle cells would<br />

have a relaxing effect and may have<br />

applications in the treatment <strong>of</strong> blood<br />

pressure, asthma and incontinence. In<br />

the brain, activation <strong>of</strong> these channels<br />

would reduce neuronal activity and<br />

could help treat epilepsy or stroke.<br />

Our research helps to identify which ion<br />

channels are valid drug targets for the<br />

treatment <strong>of</strong> particular diseases.<br />

Modulation by<br />

protein interaction<br />

Ion channels are rarely isolated<br />

structures in cell membranes,<br />

but associate with other proteins.<br />

Interactions affect how the channels<br />

behave and their drug sensitivity. One<br />

protein can have different physiological<br />

roles in different cell types, and<br />

particular protein assemblies present<br />

more cell-specific drug targets. We are<br />

investigating how different ion-channel<br />

proteins co-assemble and which<br />

other types <strong>of</strong> protein interact with ion<br />

channels to alter their properties.<br />

Funding: Wellcome Trust, BBSRC<br />

Figure 1: Model <strong>of</strong> a CLC Cl − channel structure.<br />

Figure 2: Single Na +- activated K + channels.<br />

Representative Publications<br />

Toye, AA, Lippiat, JD, Proks, P et al. (2005)<br />

A genetic and physiological study <strong>of</strong> impaired<br />

glucose homeostasis control in C57BL/6J<br />

mice. Diabetelogia 48: 675–686.<br />

Proks, P, Antcliff, JF, Lippiat, J, Gloyn,<br />

AL, Hattersley, AT & Ashcr<strong>of</strong>t, FM (2004)<br />

Molecular basis <strong>of</strong> Kir6.2 mutations associated<br />

with neonatal diabetes or neonatal diabetes<br />

plus neurological features. Proceedings <strong>of</strong><br />

the National Academy <strong>of</strong> Sciences USA 50:<br />

17539–17544.<br />

Tsuboi, T, Lippiat, JD, Ashcr<strong>of</strong>t, FM & Rutter,<br />

G (2004) ATP-dependent interaction <strong>of</strong> the<br />

cytosolic domains <strong>of</strong> the inwardly rectifying<br />

K + channel Kir6.2 revealed by fluorescence<br />

resonance energy transfer. Proceedings <strong>of</strong><br />

the National Academy <strong>of</strong> Sciences USA 101:<br />

76–81.


David Marples<br />

BA in Physiological Sciences (Oxford) (1984)<br />

MA, BM. BCh. (Oxford) (1990)<br />

D.Phil in Physiology (Oxford) (1990)<br />

House jobs (surgery in High Wycombe & medicine in Oxford) (1990/91)<br />

Post-Doc with A Taylor (Oxford) (1991-1993)<br />

Assistant Pr<strong>of</strong>essor with Søren Nielsen, Aarhus, Denmark (1993-1996)<br />

University Research Fellow, Leeds (1996-2001)<br />

Lecturer, University <strong>of</strong> Leeds (2001-2006)<br />

Senior Lecturer, University <strong>of</strong> Leeds (2006-)<br />

Contact: d.d.r.marples@leeds.ac.uk<br />

Aquaporin water channels<br />

and the kidney in health and disease<br />

My research group is interested in how<br />

the regulation <strong>of</strong> the expression and<br />

distribution <strong>of</strong> aquaporins in the kidney<br />

helps us to maintain a normal body fluid<br />

composition, and how these processes<br />

are disrupted in disease. Ultimately,<br />

we hope to be able to correct these<br />

pathological disorders, and maybe use<br />

the defects therapeutically, for example<br />

to generate novel diuretics.<br />

Aquaporin 2 (AQP2) water channels<br />

are found in the principal cells <strong>of</strong> the<br />

renal collecting duct. They shuttle to<br />

the cell surface in response to<br />

antidiuretic hormone, allowing the<br />

reabsorption <strong>of</strong> water, and production<br />

<strong>of</strong> a concentrated urine. This acute<br />

response is modulated by changes in<br />

AQP2 expression, for example due to<br />

chronic water deprivation.<br />

Defects in the acute trafficking <strong>of</strong><br />

AQP2 cause nephrogenic diabetes<br />

insipidus (NDI), in which patients<br />

produce huge volumes <strong>of</strong> urine. This<br />

disrupts their lives very badly, and<br />

can lead to dehydration and brain<br />

damage, especially in babies. We are<br />

looking for alternative ways to drive this<br />

trafficking, and so overcome the defect.<br />

Some drugs alter AQP2 expression:<br />

for example, lithium greatly decreases<br />

its expression, leading to NDI. We are<br />

studying the factors which regulate<br />

AQP2 expression. This may help us<br />

treat patients with this type <strong>of</strong> NDI by<br />

increasing AQP2, and may allow us<br />

to decrease AQP2, to increase water<br />

loss in fluid-retaining states such as<br />

congestive heart failure.<br />

Other aquaporins (1, 3, 4, 6, 7, 8, and<br />

11) are also found in the kidney: with<br />

the exception <strong>of</strong> AQP1 their roles are<br />

much less well understood. We plan to<br />

study some <strong>of</strong> these aquaporins, and<br />

see if they can also be useful in treating<br />

human disease.<br />

Aquaporins are also found in many other<br />

places in the body: we are studying this<br />

in collaboration with other groups in the<br />

UK and abroad.<br />

A second topic <strong>of</strong> interest is the<br />

organisation <strong>of</strong> microtubules in<br />

epithelia, and their role in vesicle<br />

trafficking, including that <strong>of</strong> AQP2 in<br />

collecting duct cells.<br />

Figure 1: AQP2 shuttling. In the top, control panel, AQP2<br />

(shown in red) lies within the cells. After ADH (bottom<br />

panel), it is in the plasma membrane, together with the<br />

green membrane marker.<br />

Figure 2: Effect <strong>of</strong> Lithium on AQP2 expression :<br />

AQP2 is decreased by about 95%<br />

Funding: MRC, Kidney Research UK<br />

More information:<br />

www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.<br />

php?staff=DDRM<br />

Representative Publications<br />

Baggaley E, Nielsen S, Marples D (2010)<br />

Dehydration-induced increase in aquaporin-2<br />

protein abundance is blocked by nonsteroidal<br />

anti-inflammatory drugs. American Journal <strong>of</strong><br />

Physiology 298: F1051-F1058<br />

Lutken SC, Kim SW, Jonassen T, Marples D,<br />

Knepper MA, Kwon TH, Frokiaer J, Nielsen S<br />

(2009) Changes <strong>of</strong> renal AQP2, ENaC, and NHE3 in<br />

experimentally induced heart failure: response to<br />

angiotensin II AT(1) receptor blockade. American<br />

Journal <strong>of</strong> Physiology 297: F1678-F1688<br />

Floyd RV, Mason SL, Proudman CJ, German AJ,<br />

Marples D, Mobasheri A (2007) Expression and<br />

nephron segment-specific distribution <strong>of</strong> major<br />

renal aquaporins (AQP1-4) in Equus caballus, the<br />

domestic horse American Journal <strong>of</strong> Physiology,<br />

293: R492-R503<br />

Shaw S & Marples DDR (2005) N-ethylmaleimide<br />

causes aquaporin-2 trafficking in the renal inner<br />

medullary collecting duct by direct activation <strong>of</strong><br />

protein kinase A. American Journal <strong>of</strong> Physiology,<br />

288: 832 – 839<br />

Mobasheri A, Marples DDR (2004) Expression<br />

<strong>of</strong> the AQP-1 water channel in normal human<br />

tissues: a semiquantitative study using tissue<br />

microarray technology. American Journal <strong>of</strong><br />

Physiology 286: C529-C537<br />

Christensen BM, Marples DDR, Kim YH, Wang WD,<br />

Frokiaer J, Nielsen S (2004) Changes in cellular<br />

composition <strong>of</strong> kidney collecting duct cells in rats<br />

with lithium-induced NDI. American Journal <strong>of</strong><br />

Physiology 286: C952-C964<br />

Shaw S & Marples DDR (2002) A rat kidney tubule<br />

suspension for the study <strong>of</strong> vasopressin-induced<br />

shuttling <strong>of</strong> AQP2 water channels. American<br />

Journal <strong>of</strong> Physiology 283: F1160-F1166


Neil Messenger<br />

BSc (Salford)<br />

PhD (Salford)<br />

Postdoctoral research, University <strong>of</strong> Salford<br />

Lecturer in Sport Biomechanics, University <strong>of</strong> Brighton<br />

Lecturer in Sport and Exercise Biomechanics (1995-)<br />

Contact: n.messenger@leeds.ac.uk<br />

Biomechanics <strong>of</strong><br />

lower limb function<br />

The principle research undertaken in this<br />

programme examines the biomechanical<br />

aetiology <strong>of</strong> overuse injury in sport and<br />

exercise and we are also interested in the<br />

mechanical adaptation to locomotion<br />

under various external conditions or<br />

novel environments.<br />

Overuse injuries <strong>of</strong> the lower limb are<br />

common in most sports and can, in<br />

extreme cases, result in the complete<br />

withdrawal <strong>of</strong> an athlete from their<br />

chosen activity. These injuries affect<br />

athletes across the full spectrum <strong>of</strong><br />

performance levels, from elite to<br />

recreational, and have a significant impact<br />

on sport and exercise participation. It has<br />

long been postulated that an individual’s<br />

susceptibility to such injuries is in part<br />

due to their particular musculoskeletal<br />

structure and the coupling <strong>of</strong> limb motion<br />

across a number <strong>of</strong> joints. In particular<br />

these injuries have been associated with<br />

malfunction <strong>of</strong> the subtalar joint and its<br />

supporting structures. This joint has <strong>of</strong>ten<br />

been described as a torque converter<br />

in the transition <strong>of</strong> motion above the<br />

support foot during repetitive activities<br />

such as running. However, recent work<br />

has shown that, although this mechanism<br />

is apparent in both running and walking,<br />

the rigidity <strong>of</strong> the coupling is less than<br />

previously thought and that interaction<br />

between the forefoot and the lower<br />

limb is as important as that between the<br />

rearfoot and the lower limb. Future work<br />

seeks to develop a more complete model<br />

<strong>of</strong> these interactions.<br />

Many gait irregularities are functions <strong>of</strong><br />

poor or improper control <strong>of</strong> locomotion.<br />

The consequences <strong>of</strong> these, as in trips and<br />

falls in the elderly, can be devastating.<br />

Sport provides a useful model for the<br />

analysis <strong>of</strong> the control mechanisms<br />

used to maintain effective and efficient<br />

gait as it provides a number <strong>of</strong> well<br />

defined environments in which to<br />

investigate perturbations to and stresses<br />

on the locomotor apparatus. Current<br />

research focuses on: the effect <strong>of</strong> surface<br />

interaction and footwear: limb segment<br />

coordination in gait: the control <strong>of</strong><br />

balance in activities such as archery.<br />

Figure 1: Still image from computer animation <strong>of</strong><br />

human walking: bonecolour depth indicates<br />

More information:<br />

http://www.leeds.ac.uk/sports_science/<br />

staff/nm.htm<br />

Representative Publications<br />

Pohl MB, Messenger N & Buckley JG (2006)<br />

Changes in foot and lower limb coupling due<br />

to systematic variations in step width. Clinical<br />

Biomechanics 21: 175–183<br />

Bailey M, Maillardet FJ & Messenger N (2003)<br />

Kinematics <strong>of</strong> cycling in relation to anterior knee<br />

pain and patellar tendonitis. Journal <strong>of</strong> Sports<br />

Sciences 21: 649–657<br />

Eubank C & Messenger N (2000) The frequency<br />

and causes <strong>of</strong> injury in squash - conference<br />

communication Journal <strong>of</strong> Sports Sciences 18:<br />

13–14.<br />

Messenger N, Patterson WD & Brook DB (2000)<br />

The Science <strong>of</strong> Climbing and Mountaineering.<br />

Human Kinetics, Champaign IL


Paul Millner<br />

BSc (Leeds)<br />

PhD (Leeds)<br />

Postdoctoral research, Purdcue University, USA and Imperial College, London<br />

Reader in Biotechnology (2006-)<br />

Contact: p.a.millner@leeds.ac.uk<br />

Biosensors, Biocatalysis<br />

and Affinity systems<br />

Biosensor design: Enzyme based<br />

sensors: we are engaged in work aimed<br />

at optimisation and stabilisation <strong>of</strong><br />

acetylcholine esterase (AChe) based<br />

electrochemical biosensor chips<br />

for detection <strong>of</strong> organophosphate<br />

pesticide residues in food and water<br />

(EC Project SAFEGARD). Our aim is<br />

to effect efficient immobilisation <strong>of</strong><br />

AchE and other industrially important<br />

enzymes to carbon and metal<br />

electrode surfaces and to improve<br />

their storage and operational stability.<br />

In addition, we are developing a range<br />

<strong>of</strong> immunosensors. These include<br />

biosensors for GM-material in food<br />

stuffs (EC Project IMAGEMO), based<br />

on affinity immobilisation <strong>of</strong> tagged<br />

antibodies to mixed self assembled<br />

mixed monolayers and sensors for<br />

antibiotics, cancer markers and<br />

other analytes (www.immunosensors.<br />

com) based on antibodies attached<br />

to electropolymerised pyrrole layers.<br />

In particular, we are interested in the<br />

nanoscale structure <strong>of</strong> the sensor<br />

surface and the mechanism underlying<br />

generation <strong>of</strong> the amperometric or<br />

impedance signal.<br />

Biocatalysis: the development <strong>of</strong><br />

improved, cheap and biodegradeable<br />

supports for the attachment <strong>of</strong><br />

enzymes for biocatalysis is <strong>of</strong> increased<br />

importance, since it is simpler to<br />

dispose <strong>of</strong> the support matrix after<br />

synthesis than carry out cleanup.<br />

We have developed matrices based on<br />

carageenan, a natural biodegradeable<br />

polysaccharide isolated from red<br />

seaweeds and current EC projects<br />

SANTS and COMBIO are concerned<br />

with using biosilicates and polymeric<br />

nanoparticles as enzyme supports<br />

for biocatalysis.<br />

Funding: Our work is sponsored<br />

by the EC, under Framework 6,<br />

and the HOSDB.<br />

Figure 2: scanning electrochemical image <strong>of</strong> microarray<br />

electrode design P10 (courtesy <strong>of</strong> Dr G Johnson, Uniscan<br />

Instruments Ltd)<br />

Representative Publications<br />

Vakurov A, Simpson CE, Daly CL, Gibson TD,<br />

Millner PA (2005) Acetylecholinesterase-based<br />

biosensor electrodes for organophosphate<br />

pesticide detection. II. Immobilization<br />

and stabilization <strong>of</strong> acetylecholinesterase<br />

Biosensors and Bioelectronics 20, 2324-<br />

2329.<br />

Weiss S, Millner P,Nelson A (2005).<br />

Monitoring protein binding to phospholipid<br />

monolayers using electrochemical impedance<br />

spectroscopy. Electrochimica Acta - 50, 4248<br />

- 4256.<br />

Hays H,Millner PA, Prodromidis MI<br />

(2006). Development <strong>of</strong> capacitance based<br />

immunosensors on mixed self-assembled<br />

monolayers. Sensors & Actuators: B.<br />

Chemical.114, 1064-1070.<br />

Hleli S, Martelet C, Abdelghani A, Bessueille<br />

F, Errachid A, Samitier J, Hays HCW,Millner<br />

PA, Burais N, Jaffrezic-Renault N (2006).<br />

An immunosensor for haemoglobin based<br />

on impedimetric properties <strong>of</strong> a new mixed<br />

self-assembled monolayer Materials Sci.and<br />

Engineering C.26, 322-327.


Hugh Pearson<br />

BSc Pharmacology, University <strong>of</strong> London<br />

PhD Pharmacology, University <strong>of</strong> London<br />

Postdoctoral Research, University <strong>of</strong> London<br />

Lecturer, University <strong>of</strong> Leeds<br />

Contact: h.a.pearson@leeds.ac.uk<br />

The role <strong>of</strong> ion channels<br />

in Alzheimer’s disease<br />

Alzheimer’s disease is becoming an<br />

increasing large clinical problem as<br />

the average age <strong>of</strong> our population gets<br />

higher. An estimated 5-10% <strong>of</strong> the<br />

population over the age <strong>of</strong> 65 develop<br />

Alzheimer’s disease (AD) and this figure<br />

increases even further in more elderly<br />

age groups. A hallmark <strong>of</strong> AD is the loss<br />

<strong>of</strong> neurones in the cortex and memory<br />

forming areas <strong>of</strong> the brain such as the<br />

hippocampus. We have been studying<br />

the effects <strong>of</strong> an AD-related peptide,<br />

amyloid protein (Aß) on voltage-gated ion<br />

channel currents in cultured neurones<br />

using the whole-cell patch clamp<br />

technique (see publications).<br />

The effects on ion channel currents have<br />

also been correlated with the Aß-induced<br />

cell death. To do this we use a variety <strong>of</strong><br />

neurotoxicity assays, some biochemical<br />

such as the MTT, TUNEL or LDH<br />

release assays, and some morphological,<br />

where we fix and stain cells and look<br />

for changes in membrane and nuclear<br />

structure. We also use fluorescent assays<br />

to monitor cell death/survival following<br />

challenge with Aß. Related to this project<br />

are studies regarding the effect <strong>of</strong> free<br />

radicals on ion channel activity and<br />

cell survival.<br />

A project recently funded by the<br />

Alzheimer’s Research Trust is looking at<br />

potential side effects <strong>of</strong> novel Alzheimer’s<br />

disease treatments. We have shown<br />

that Aß is not just a toxic peptide, but<br />

may also have a physiological role in<br />

the brain. This suggests that drugs that<br />

prevent Aß production may have serious<br />

side effects on brain function. We are<br />

using cell death assays and biochemical<br />

studies to investigate this possibility.<br />

Funding: Medical Research Council,<br />

BBSRC, Alzheimer’s Research Trust,<br />

Wellcome Trust<br />

Figure 2: Amyloid ß protein expression in cultured<br />

neurones from cerebellum).<br />

Representative Publications<br />

Pearson HA, Peers C (2006). Physiological<br />

roles for amyloid peptides. J. Physiol. 575:<br />

5-10<br />

LD Plant, NJ Webster, JP Boyle, M Ramsden,<br />

DB Freir C Peers, HA Pearson. (2006) Amyloid<br />

β peptide as a physiological modulator <strong>of</strong><br />

neuronal ‘A’-type K+ current Neurobiology <strong>of</strong><br />

Aging 27: 1673-1683<br />

Atkinson L, Boyle JP, Pearson HA, Peers C.<br />

Chronic hypoxia inhibits Na+/Ca 2+ exchanger<br />

expression in cortical astrocytes. (2006)<br />

Neuroreport. 17:649-652.<br />

LD Plant, JP. Boyle , IF Smith, C Peers & HA<br />

Pearson (2003). The production <strong>of</strong> amyloid<br />

β peptide is a critical requirement for the<br />

viability <strong>of</strong> central neurones. J. Neurosci.<br />

23:5531-5535 activated current (Ih) in the<br />

medial septum/diagonal band complex in the<br />

mouse. Brain Research 1006: 74–86.<br />

Yeung, SYM, Thompson, D, Wang, Z, Fedida,<br />

D & Robertson, B (2005) Modulation <strong>of</strong> Kv3<br />

subfamily potassium currents by the sea<br />

anemone toxin BDS: Significance for CNS and<br />

biophysical studies. Journal <strong>of</strong> Neuroscience<br />

25: 8735–8745<br />

Figure 1: Fluorescence based Live/Dead assay in a<br />

neuronal cell line


Harry B. Rossiter<br />

BSc (Birmingham)<br />

MSc (King’s College, University <strong>of</strong> London)<br />

PhD (St. George’s Hospital Medical School, University <strong>of</strong> London)<br />

Fellow <strong>of</strong> The American College <strong>of</strong> Sports Medicine<br />

Editorial Board, The European Journal <strong>of</strong> Applied Physiology<br />

Wellcome Trust International Prize Travelling Research Fellow, UC San Diego (2001-2003)<br />

National <strong>Institute</strong>s <strong>of</strong> Health National Research Service Award, UC San Diego (2003-2005)<br />

Lecturer in Exercise Physiology (2005-)<br />

Contact: h.b.rossiter@leeds.ac.uk<br />

Exercise bioenergetics<br />

in health and disease<br />

Research in our labs focuses on the<br />

mechanisms controlling and limiting oxygen<br />

transport and utilisation during exercise.<br />

The ability to sustain muscular exercise is<br />

a key determinant <strong>of</strong> health and mortality<br />

in ageing and chronic disease (Myers et al.<br />

New Eng J Med 46:793-801, 2002). As such,<br />

we investigate the effective integration <strong>of</strong><br />

the cardio-pulmonary and neuromuscular<br />

systems that conflate to allow muscular<br />

exercise to be sustained - with a special<br />

focus on the nonsteady-state.<br />

To achieve this we use a multi-scale systems<br />

biology approach.<br />

Figure 1: Cardiopulmonary Exercise Testing (CPET) with<br />

tissue oxygenation in chronic heart failure.<br />

Patient Studies<br />

In the Exercise Physiology Laboratory we use<br />

state-<strong>of</strong>-the-art CPET by mass spectrometry<br />

and turbinometry for alveolar gas exchange,<br />

12-Lead ECG, and tissue oxygenation<br />

monitoring to investigate exercise responses<br />

in patients with chronic heart failure (CHF)<br />

and chronic obstructive pulmonary disease<br />

(COPD). We collaborate with Dr Klaus Witte<br />

(pictured; Fig 1), Consultant Cardiologist at<br />

Leeds General Infirmary, which is one <strong>of</strong> the<br />

largest teaching hospitals in the UK serving<br />

a population <strong>of</strong> >0.75M. Our focus is on<br />

the mechanisms contributing to systems<br />

limitations in order to better understand<br />

their relationships with mortality, and to<br />

design strategies for the amelioration <strong>of</strong><br />

exercise intolerance.<br />

Human Performance<br />

Exercise intolerance extends throughout<br />

the spectrum <strong>of</strong> human performance. This<br />

strand <strong>of</strong> our research focuses on exercise<br />

responses in health, ageing and elite<br />

athletes (in collaboration with UK Sport)<br />

to provide a better understanding <strong>of</strong> the<br />

dynamic integration <strong>of</strong> physiological<br />

systems for the optimisation <strong>of</strong> human<br />

performance. We use non-invasive<br />

techniques for muscle and whole-body<br />

investigation <strong>of</strong> exercise dynamics, such<br />

as: Magnetic resonance spectroscopy and<br />

imaging (Fig 2); Dopplar ultrasound for<br />

blood flow; Near-infrared spectroscopy <strong>of</strong><br />

tissue oxygenation; and muscle structure<br />

and metabolism from biopsy samples.<br />

In Vivo Models<br />

To probe deeper into tissue and organelle<br />

function we use animal models <strong>of</strong> exercise<br />

and chronic disease: Pulmonary structure<br />

and function in models <strong>of</strong> COPD is<br />

investigated using combined magnetic<br />

resonance imaging and molecular<br />

biological techniques (Fig 3); Muscle<br />

morphology and mitochondrial function<br />

is probed in models <strong>of</strong> ageing and CHF;<br />

Cardiovascular and skeletal muscle<br />

interactions are investigated using selfand<br />

pump-perfused muscle models.<br />

Figure 2: 3T magnetic resonance spectroscopy and<br />

imaging <strong>of</strong> quadriceps mitochondrial function, acid-base<br />

status, and recruitment patterns during exercise.<br />

Figure 3: 3D models <strong>of</strong> murine lung structures from 80μm<br />

isotropic magnetic resonance images at 7 Tesla.<br />

<strong>Systems</strong> <strong>Biology</strong> <strong>of</strong> Exercise<br />

Multi-scale measurements are integrated<br />

in computational models combining,<br />

muscle metabolism, circulatory dynamics<br />

and pulmonary responses during exercise<br />

transients. These quantitative models aim<br />

to elucidate the emergent properties <strong>of</strong><br />

the integrated physiological systems in<br />

limiting exercise tolerance.<br />

Funding: Biotechnology & <strong>Biological</strong><br />

Sciences Research Council UK, Medical<br />

Research Council UK, The Wellcome Trust<br />

UK, National <strong>Institute</strong>s <strong>of</strong> Health USA;<br />

Leeds MCRC.<br />

More information: http://www.<br />

cardiovascular.leeds.ac.uk/staff/<br />

Rossiter_H/index.htm<br />

Representative Publications<br />

Cubbon RM et al. (2010) Human exercise-induced<br />

circulating progenitor cell mobilization is nitric<br />

oxide-dependent and is blunted in South Asian men.<br />

Arteriosclerosis, Thrombosis and Vascular <strong>Biology</strong><br />

30: 878-884<br />

Rossiter HB et al. (2008) Doxycycline treatment<br />

prevents alveolar destruction in VEGF-deficient<br />

mouse lung. Journal <strong>of</strong> Cellular Biochemistry 104:<br />

525-535<br />

Endo MY et al. (2007) Thigh muscle activation<br />

distribution and pulmonary VO2 kinetics during<br />

moderate, heavy and severe intensity cycling<br />

exercise in humans. American Journal <strong>of</strong> Physiology<br />

293: R812-R820<br />

Rossiter HB et al. (2006) A test to establish maximal<br />

O2 uptake despite no plateau in the O2 uptake<br />

response to ramp incremental exercise. Journal <strong>of</strong><br />

Applied Physiology 100: 764-770


Asipu Sivaprasadarao<br />

BSc, MSc (Andhra University, Visakhapatnam, India)<br />

PhD (Leeds; Commonwealth Fellow 1987)<br />

Lecturer/Senior Lecturer in Biomedical Sciences (1993-2006)<br />

Reader in <strong>Membrane</strong> <strong>Biology</strong> (2006-2007)<br />

Pr<strong>of</strong>essor <strong>of</strong> <strong>Membrane</strong> <strong>Biology</strong> (2007-)<br />

Contact: a.sivaprasadarao@leeds.ac.uk<br />

Potassium channels<br />

in health and disease<br />

Research in my laboratory is focused<br />

on ion channels, a family <strong>of</strong> membrane<br />

proteins that control the flow <strong>of</strong> ions<br />

across the cell membrane. Ion channels<br />

play crucial roles in a wide range <strong>of</strong><br />

physiological processes including<br />

transmission <strong>of</strong> nerve impulses,<br />

heartbeat, muscle contraction, hormone<br />

secretion, cell proliferation and<br />

apoptosis. Consequently, defects in the<br />

function <strong>of</strong> ion channels lead to diseases<br />

such as epilepsy, cardiac arrhythmias,<br />

kidney dysfunction and diabetes.<br />

As such, ion channels represent<br />

important drug targets. Our goals are to<br />

understand (i) how ion channels work<br />

at the molecular level and (ii) how they<br />

are made and transported to and out<br />

<strong>of</strong> their site <strong>of</strong> action. To address the<br />

first question, we use techniques in<br />

molecular biology, electrophysiology<br />

and protein chemistry, and monitor<br />

conformational changes associated with<br />

the function <strong>of</strong> channels. This approach<br />

has allowed us to make important<br />

discoveries on how ion channels sense<br />

changes in voltage (for example, see<br />

refs 1,2 and Figure 1). To address the<br />

second question, we combine the above<br />

techniques with approaches in cell<br />

biology. These integrated approaches<br />

have led to several new findings: for<br />

example, we have demonstrated that a<br />

genetic mutation in the ER exit signal <strong>of</strong><br />

the pancreatic K ATP<br />

channel (a potassium<br />

channel that regulates insulin secretion)<br />

underlies congenital hyperinsulinism (ref<br />

3, Figure 2), while an inherited mutation<br />

in the internalization signal contributes<br />

to neonatal diabetes (ref 4). Building<br />

on this expertise, we have begun<br />

expanding our research into other ion<br />

channels including the hERG potassium<br />

channel, sodium channels and TRP<br />

channels in order to understand their<br />

role in health and disease. Once the<br />

molecular and cellular mechanisms are<br />

revealed, we aim to screen chemical<br />

libraries and cell penetrating peptides<br />

(ref 3; Figure 2) to identify novel<br />

compounds with therapeutic potential.<br />

Funding: Wellcome Trust, MRC, BHF,<br />

BBSRC, GlaxoSmithKline, Millipore<br />

Figure 1: Structure <strong>of</strong> the voltage sensing domain <strong>of</strong> KvAP<br />

showing reduced membrane thickness around S3 and S4,<br />

as determined by the site-directed cysteine accessibility<br />

approach.<br />

Figure 2: Top: In healthy individuals K ATP<br />

channels traffic<br />

to the cell surface (red), but in neonatal diabetes they fail<br />

to do so due to a mutation in an ER exit signal. Bottom:<br />

a designer peptide bearing the exit signal prevents cell<br />

surface expression <strong>of</strong> the channel- a property that could<br />

be exploited to stimulate insulin secretion.<br />

Representative Publications<br />

Elliott DJS, Neale EJ, Aziz Q, Dunham JP, Munsey<br />

TS, Hunter M, Sivaprasadarao A (2004) Molecular<br />

mechanism <strong>of</strong> voltage sensor movements in a<br />

potassium channel EMBO J 23: 4717-4726<br />

Neale EJ, Rong H, Cockcr<strong>of</strong>t CJ, Sivaprasadarao A<br />

(2007) Mapping the <strong>Membrane</strong>-aqueous Border<br />

for the Voltage-sensing Domain <strong>of</strong> a Potassium<br />

Channel. Journal <strong>of</strong> <strong>Biological</strong> Chemistry 282:<br />

37597-37604<br />

Taneja TK, Mankouri J, Karnik R, Kannan S,<br />

Smith AJ, Munsey T, Christesen HB, Beech DJ,<br />

Sivaprasadarao A (2009) Sar1-GTPase-dependent<br />

ER exit <strong>of</strong> KATP channels revealed by a mutation<br />

causing congenital hyperinsulinism. Hum Mol<br />

Genet 18: 2400-2413<br />

Mankouri J, Taneja TK, Smith AJ, Ponnambalam<br />

S, Sivaprasadarao A (2006) Kir6.2 mutations<br />

causing neonatal diabetes prevent endocytosis<br />

<strong>of</strong> ATP-sensitive potassium channels. EMBO J 25:<br />

4142-4151


Derek Steele<br />

BSc (Glasgow)<br />

PhD (Glasgow)<br />

Postdoctoral research at Glasgow<br />

Lecturer (1996-2000)<br />

Senior Lecturer (1996-)<br />

Cardiovascular Group Leader, <strong>Institute</strong> <strong>of</strong> <strong>Membrane</strong> & <strong>Systems</strong> <strong>Biology</strong> (2005-)<br />

Contact: d.steele@leeds.ac.uk<br />

Ryanodine receptor function in<br />

skeletal and cardiac muscle disease<br />

Most <strong>of</strong> my work addresses the role <strong>of</strong><br />

the sarcoplasmic reticulum (SR) Ca 2+<br />

channel (ryanodine receptor, RyR) in<br />

cardiac and skeletal muscle. In cardiac<br />

and skeletal muscle, electrical excitation<br />

<strong>of</strong> the surface membrane causes Ca 2+<br />

release from the SR and contraction <strong>of</strong><br />

my<strong>of</strong>ilaments. We study SR Ca 2+ release<br />

in normal cells and aberrant Ca 2+<br />

release in disease (ischaemia, inherited<br />

cardiomyopathies). Our recent work on<br />

cardiac muscle has addressed the role<br />

<strong>of</strong> the t-tubules in excitation–contraction<br />

coupling, changes in SR Ca 2+<br />

regulation associated with ischaemia<br />

or anoxia (e.g. ATP depletion) and<br />

the cardioprotective effects <strong>of</strong> volatile<br />

anaesthetics. Recently we described<br />

long-lasting Ca 2+ -release events that<br />

may be involved in regulation <strong>of</strong><br />

gene transcription or expression. In<br />

a collaborative project (with Noriaki<br />

Ikemoto and Graham Lamb) we are<br />

using peptides to induce and study<br />

abnormal RyR2 gating associated<br />

with inherited arrhythmias. This<br />

‘peptide probe’ technique is based<br />

on the hypothesis that in normal<br />

RyR2 channels interaction between<br />

the N-terminal and central domains<br />

stabilizes the channel (Figure 1).<br />

Mutations in these regions can induce<br />

abnormal Ca 2+ release and consequent<br />

arrhythmias. Our work on skeletal<br />

muscle addresses the development <strong>of</strong><br />

fatigue and malignant hyperthermia,<br />

an inherited condition associated<br />

with potentially fatal increases in<br />

core body temperature, triggered<br />

by exposure to volatile anaesthetics<br />

(Figure 2). The abnormality is known<br />

to be caused by mutations in RyR1,<br />

which result in sustained Ca 2+ release<br />

and contraction during anaesthesia.<br />

Working with clinicians and geneticists<br />

(St James’s Hospital, Leeds) we have<br />

shown that reduced Mg 2+ inhibition <strong>of</strong><br />

RyR1 is a common feature <strong>of</strong> many<br />

channel mutations and may explain<br />

the abnormal response to anaesthetics.<br />

Ongoing work involves characterization<br />

<strong>of</strong> novel RyR1 mutations and<br />

development <strong>of</strong> non-invasive DNAbased<br />

screening.<br />

Figure 1: Proposed interaction between the N-terminal<br />

and central RyR2 domains.<br />

Figure 2: Image sequence showing a propagated<br />

Ca2+ wave induced by halothane in a mechanically<br />

skinned human skeletal muscle fibre from a patient with<br />

malignant hyperthermia<br />

Funding: Wellcome Trust, British Heart<br />

Foundation, Department <strong>of</strong> Health<br />

Overseas collaborators: Noriaki Ikemoto,<br />

Graham Lamb (Australia<br />

More information:<br />

http://www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.<br />

php?staff=DSS<br />

Representative Publications<br />

Yang, Z, Harrison, SM & Steele, DS (2005)<br />

ATP-dependent effects <strong>of</strong> halothane on<br />

SR Ca2+ regulation in permeabilized atrial<br />

myocytes. Cardiovascular Research 65:<br />

167–176.<br />

Yang, Z & Steele, DS (2005) Characteristics<br />

<strong>of</strong> prolonged Ca2+ release events associated<br />

with the nuclei in adult cardiac myocytes.<br />

Circulation Research 96: 82–90.<br />

Duke, M, Hopkins, PM, Halsall, JP & Steele,<br />

DS (2004) Mg 2+ dependence <strong>of</strong> halothaneinduced<br />

Ca 2+ release from the sarcoplasmic<br />

reticulum in skeletal muscle from humans<br />

susceptible to malignant hyperthermia.<br />

Anesthesiology 101: 1339–1346.<br />

Yang, Z, Pascarel C, Steele, DS, Komukai,<br />

K, Brette, F & Orchard, CH (2002) Na+-Ca 2+<br />

exchange activity is localized in the T-tubules<br />

<strong>of</strong> rat ventricular myocytes. Circulation<br />

Research 91: 315–322.


Andrea Utley<br />

BA (Hons) Human Movement Studies (1984)<br />

PGCE Physical Education (1985)<br />

PhD University <strong>of</strong> Leeds (1998)<br />

Lecturer, School <strong>of</strong> Education, University <strong>of</strong> Leeds, (1993-1997)<br />

Senior Lecturer. Motor Control and Learning, Leeds Metropolitan University (1997-1999)<br />

Senior Lecturer, Director Centre for Sport and Exercise Sciences, <strong>Institute</strong> <strong>of</strong> <strong>Systems</strong> and <strong>Membrane</strong>s <strong>Biology</strong>,<br />

University <strong>of</strong> Leeds (1999-)<br />

Contact: A.Utley@leeds.ac.uk<br />

Motor and<br />

Behavioural Science<br />

This group addresses the mechanisms<br />

<strong>of</strong> control and disorders <strong>of</strong> co-ordination<br />

in conditions such as hemiplegic<br />

cerebral palsy and Developmental<br />

Coordination Disorder. A particular<br />

research interest is how children and<br />

adults with a range <strong>of</strong> movement<br />

difficulties are able to control their<br />

movements in a variety <strong>of</strong> contexts.<br />

Detailed analysis <strong>of</strong> reaching/ grasping<br />

and catching in children has revealed<br />

how these children attempt to control<br />

multiple degrees <strong>of</strong> freedom. It also<br />

addresses the assessment <strong>of</strong> movement<br />

and how manipulating the movement<br />

context can be used as a rehabilitative<br />

strategy. Explanations <strong>of</strong> motor<br />

development have taken a step forward<br />

through the application <strong>of</strong> ideas from<br />

proponents <strong>of</strong> dynamic systems, here<br />

movement involves the final product<br />

or whole being the active cooperation<br />

<strong>of</strong> many parts, and contains multiple<br />

subsystems all contributing in a unique<br />

manner (Thelen and Spencer 1998).<br />

The potential for some <strong>of</strong> these ideas<br />

has been initially explored in the context<br />

<strong>of</strong> reaching and grasping in children<br />

with hemiplegic cerebral palsy (Utley<br />

and Sugden 1998) and catching in<br />

children with DCD (Utley and Astill<br />

2006). Such children have to overcome<br />

intrinsic constraints where the neural<br />

properties provide a direct link to the<br />

type <strong>of</strong> movement observed. External<br />

constraints such as task demands<br />

and context also influence the nature<br />

and extent <strong>of</strong> interlimb coupling. We<br />

are especially interested in the nature<br />

and extent <strong>of</strong> interlimb coupling and<br />

have provided evidence on the nature<br />

<strong>of</strong> bimanual co-ordination in children<br />

with cerebral palsy. This work has<br />

indicated that one solution to the<br />

degrees <strong>of</strong> freedom problem during<br />

upper limb movements is to couple the<br />

limbs therefore reducing the number <strong>of</strong><br />

degrees <strong>of</strong> freedom to be controlled.<br />

Collaboration with Leeds University<br />

Motor Impairment Group (LUMIG) and<br />

external collaborators at the University<br />

<strong>of</strong> Nijmegen (The Netherlands)<br />

and University <strong>of</strong> Minnesota (USA).<br />

Techniques employed include<br />

kinematic analysis, electromyography,<br />

eye-tracking and modelling.<br />

More information:<br />

http://www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.<br />

php?tag=utley<br />

Funding: British Council, ESRC,<br />

Nuffield <strong>Institute</strong><br />

Representative Publications<br />

Utley A, Steenbergen B, Astill SL (2007)<br />

Ball catching in children with developmental<br />

coordination disorder: control <strong>of</strong> degrees <strong>of</strong><br />

freedom Developmental Medicine and Child<br />

Neurology 49 (1): 34-38<br />

Utley A, Sugden DA, Lawrence G, and Astill<br />

S. L (2007). The influence <strong>of</strong> perturbing the<br />

working surface during reaching and grasping<br />

in children with hemiplegic cerebral palsy.<br />

Disability and Rehabilitation 29 (1): 79-89<br />

Astill S.L.; Utley, A. (2006) Two-Handed<br />

Catching in Children with Developmental<br />

Coordination Disorder. Motor Control, 10(2),<br />

pp.109-124.<br />

Utley, A. & B. Steenbergen (2006). Discrete<br />

bimanual co-ordination in children and young<br />

adolescents with hemiparetic cerebral palsy:<br />

Recent findings, implications and future<br />

research directions. Pediatric Rehabilitation,<br />

9(2), 127-136.<br />

Steenbergen, B. & Utley A. (2005). Cerebral<br />

palsy: Recent insights into movement<br />

deviations. Motor Control, 9(4), 353-356.<br />

Utley A.; Steenbergen B.; Sugden D.A.<br />

(2004) The influence <strong>of</strong> object size on<br />

discrete bimanual co-ordination in children<br />

with hemiplegic cerebral palsy. Disability and<br />

Rehabilitation, 26(10), pp.603-613 .


Ed White<br />

University <strong>of</strong> Wales, Aberystwyth, B.Sc., Zoology<br />

University <strong>of</strong> Reading: Ph.D., Zoology<br />

University <strong>of</strong> Exeter, Dept. <strong>of</strong> Education: PGCE, <strong>Biology</strong> and Outdoor Education<br />

Post. doc: Oxford University<br />

Lectureship: Universite de Tours, France<br />

Lloyds <strong>of</strong> London and Wellcome Trust Fellowships, Pr<strong>of</strong>essor <strong>of</strong> Cardiac Physiology (2007-): University <strong>of</strong> Leeds.<br />

Contact: e.white@leeds.ac.uk<br />

Mechanical stimulation<br />

<strong>of</strong> the heart<br />

Acute mechanical stimulation (e.g.<br />

stretch) affects the contractility <strong>of</strong><br />

cardiac muscle in situations such as<br />

exercise, when diastolic ventricular<br />

volume is increased. But stretch has<br />

also been implicated in the triggering<br />

<strong>of</strong> cardiac arrhythmias. Chronic stretch<br />

provokes cardiac hypertrophy. Thus<br />

stretch may be involved in the beneficial<br />

effects <strong>of</strong> regular exercise and the<br />

cause <strong>of</strong> heart attacks, depending upon<br />

the circumstances. My research group<br />

is interested in the cellular mechanisms<br />

that cause these varied effects. We<br />

have research collaborators in France<br />

and the UAE and the laboratory has<br />

welcomed researchers from Brazil,<br />

Canada, France, Japan, Poland and<br />

the UAE.<br />

We can study the effects <strong>of</strong> acute<br />

stretch on single cardiac myocytes<br />

by attaching carbon fibres to the cell<br />

(Figure 1A) and recording the change<br />

in force (Figure 1B) or intracellular<br />

calcium (see Calaghan & White,<br />

2004 J.Physiol. 559, 205-214) or<br />

electrophysiology (Belus & White,<br />

2003).<br />

The triggers for hypertrophy in response<br />

e.g. to hypertension or to voluntary<br />

exercise are studied using functional<br />

approaches in conjunction with<br />

molecular biological techniques such<br />

as mRNA arrays and Western blotting.<br />

We are also interested in the role<br />

components <strong>of</strong> the cytoskeleton such<br />

as microtubules (Figure 2, Calaghan<br />

et al, 2001) may play as transducers<br />

<strong>of</strong> mechanical stimuli. A recent<br />

interest is the role that caveloae, small<br />

invaginations on the surface <strong>of</strong> the<br />

myocytes might play in the signalling<br />

<strong>of</strong> stretch (Calaghan & White, 2006).<br />

Overseas collaborators: J-Y LeGuennec<br />

Tours, France. FC Howarth Al Ain, UAE<br />

Funding: Wellcome Trust, British Heart<br />

Foundation; MRC, The British Council<br />

More information:<br />

http://www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.<br />

php?staff=EW<br />

(A)<br />

Figure 2: Microtubule cytoskeleton in a cardiac myocyte<br />

imaged with confocal microscopy.<br />

Representative Publications<br />

Calaghan, S.C. & White, E. (2006) Caveolae<br />

modulate excitation-contraction coupling and<br />

β2-adrenergic signalling in adult rat ventricular<br />

myocytes Cardiovasc. Res. 69, 816-824 .<br />

White, E. (2005) Temporal modulation <strong>of</strong><br />

mechano-electric feedback in cardiac muscle.<br />

In, Cardiac mechano-electric feedback and<br />

arrhythmias: from pipette to patient. Elsevier,<br />

eds KOHL, P., SACHS, F and FRANZ, M.<br />

83-91.<br />

McCrossan, Z.A., Billeter, R. & White, E.<br />

(2004) Transmural changes in size, contractile<br />

and electrical properties <strong>of</strong> SHR left ventricular<br />

myocytes during compensated hypertrophy .<br />

Cardiovasc. Res. 63, 283-292.<br />

Belus, A. & White, E. (2003) Streptomycin and<br />

intracellular calcium modulate the response <strong>of</strong><br />

single guinea pig ventricular myocytes to axial<br />

stretch. J. Physiol. 546, 501-509<br />

Natali, A.J., Wilson, L.A., Peckham, M. Turner,<br />

D. L., Harison, S. M. & White, E. (2002)<br />

Different regional effects <strong>of</strong> voluntary exercise<br />

on mechanical and electrical properties <strong>of</strong><br />

rat ventricular myocytes. J. Physiol. 541,<br />

863-875.<br />

(B)<br />

Figure 1: (A) Cardiac myocyte attached to carbon fibres<br />

(B) stretch <strong>of</strong> the myocte (up arrow) increase contractile<br />

force, release <strong>of</strong> the stretch (down arrow) reverses<br />

this effect.


Stanley White<br />

BSc (Manchester)<br />

PhD (Manchester)<br />

Beit Memorial Fellow, Yale University, CT, USA; MRC Senior Fellow, Leeds<br />

Lecturer and Senior Lecturer, Sheffield<br />

Senior Lecturer (2002-)<br />

Reviewing and Associate Editor, Nephron (2000-)<br />

Contact: s.j.white@leeds.ac.uk<br />

lon-channel and transporter<br />

function in renal epithelia<br />

I investigate mechanisms <strong>of</strong> regulation<br />

and function <strong>of</strong> membrane proteins<br />

in epithelial cells, with emphasis on<br />

the kidney. Epithelia are comprised<br />

<strong>of</strong> polarized cells and to function<br />

properly membrane proteins such as<br />

transporters, receptors and channels<br />

must be targeted to the appropriate<br />

part <strong>of</strong> the cell. This is especially<br />

important in the kidney, because salt<br />

transport across kidney epithelial cells<br />

is an important determinant <strong>of</strong><br />

blood pressure.<br />

Recently we have focused on the<br />

cellular targeting <strong>of</strong> ROMK potassium<br />

channels, which secrete potassium into<br />

the renal tubule. To do this we fuse the<br />

channel protein to green fluorescent<br />

protein. These fluorescent proteins are<br />

then visible in cultured epithelial cells.<br />

Some inherited mutations <strong>of</strong> ROMK<br />

that occur in Bartter’s syndrome lead<br />

to mistargeting <strong>of</strong> the channel. We are<br />

extending these approaches to study<br />

the targeting <strong>of</strong> other proteins, such as<br />

the P2X receptor, in the kidney<br />

(Figure 1).<br />

The function <strong>of</strong> ion channels is<br />

<strong>of</strong>ten regulated by interactions with<br />

other proteins. One protein known<br />

to interact with potassium channels,<br />

the sulphonylurea receptor, was<br />

discovered in my laboratory to be<br />

strongly expressed in kidney. However,<br />

its function is unknown. Kidney-specific<br />

gene-knockout studies may yield clues<br />

to its function.<br />

The zebrafish (Danio rerio) embryo<br />

has great potential as a model for<br />

human diseases because <strong>of</strong> its rapid<br />

development, optical transparency and<br />

the ease <strong>of</strong> genetic manipulation by<br />

antisense approaches. We have isolated<br />

the gene encoding the zebrafish version<br />

<strong>of</strong> ROMK, and have found that it is<br />

expressed in the embryonic renal tubule<br />

and skin (Figure 2). We are developing<br />

preparations <strong>of</strong> these tubules, allowing<br />

us to study the function <strong>of</strong> membrane<br />

proteins in these cells and to determine<br />

the role <strong>of</strong> potential partner proteins in<br />

membrane targeting and function.<br />

Overseas collaborators:<br />

Chou Long-Huang (USA)<br />

Funding: MRC, NKRF, Wellcome Trust<br />

More information:<br />

http://www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.<br />

php?tag=white_s<br />

Figure 1: Cultured kidney cells expressing a P2X receptor<br />

(green). Endogenous markers for apical membranes<br />

(red) and basolateral membranes (blue).<br />

Representative Publications<br />

Stewart, GS, Fenton, RA, Wang, W et al.<br />

(2004) The basolateral expression <strong>of</strong> mUT-A3<br />

in the mouse kidney. American Journal <strong>of</strong><br />

Physiology Renal Physiology 286: F979–F987.<br />

Hill, C, Giesberts, AN & White, SJ (2002)<br />

Expression <strong>of</strong> is<strong>of</strong>orms <strong>of</strong> the Na+/H+<br />

exchanger in M1 collecting duct cells.<br />

American Journal <strong>of</strong> Physiology Renal<br />

Physiology 282: F649–F654.<br />

Zeng, W-Z, Babich, V, Ortega, B et al. (2002)<br />

Evidence for endocytosis <strong>of</strong> ROMK potassium<br />

channels via clathrin-coated vesicles. American<br />

Journal <strong>of</strong> Physiology Renal Physiology 283:<br />

F630–F639.<br />

Kibble, J, Neal, A, White, SJ, Green, R,<br />

Evans, MJ & Taylor, C (2001) Renal effects <strong>of</strong><br />

glibenclamide in cystic fibrosis mice. Journal<br />

<strong>of</strong> the American Society <strong>of</strong> Nephrology 12:<br />

Figure 2: 24-h zebrafish embryo expressing ROMK mRNA<br />

in the pronephric duct and surface epithelial cells.


Andrew Wilson<br />

BSc (Hons) University <strong>of</strong> Otago, NZ<br />

PhD Indiana University, Bloomington IN, USA<br />

Post-Doctoral Research: University <strong>of</strong> Aberdeen, University <strong>of</strong> Warwick<br />

Lecturer in Motor Control (2009- )<br />

Contact: A.D.Wilson@leeds.ac.uk<br />

Perception, Action and Learning<br />

Perceptual information plays a key<br />

role in the coordination and control <strong>of</strong><br />

skilled action. The perception-action<br />

approach to studying human movement<br />

and motor control seeks to identify this<br />

perceptual information and establish<br />

the structure <strong>of</strong> the perception-action<br />

dynamic that leads to skilled behaviour.<br />

The main focus <strong>of</strong> my work is learning<br />

- how do we acquire new skills? What<br />

changes to allow these skills to develop?<br />

How does this change with age? How<br />

can we influence the process <strong>of</strong> learning,<br />

in both typical and clinical populations?<br />

Coordinated Rhythmic Movement<br />

A classic model system for studying<br />

skilled movement and learning is<br />

coordinated rhythmic movement.<br />

Human movement only exhibits two<br />

stable coordinations; in-phase (doing<br />

the same thing at the same time) and<br />

anti-phase (doing the opposite thing<br />

at the same time). Other coordinations<br />

(e.g. a syncopated rhythm) must be<br />

learned. This structure emerges from<br />

a perception-action task dynamic<br />

that includes very specific perceptual<br />

information (the relative direction <strong>of</strong><br />

motion). I have developed a full set <strong>of</strong><br />

experimental tools to assess and train<br />

both the perception and performance<br />

<strong>of</strong> coordinated rhythmic movements,<br />

and research in my lab is interested in<br />

identifying the nature <strong>of</strong> the changes<br />

Figure 1: Perceptual judgment displays<br />

that occur with learning. These tools<br />

include perceptual judgments (Figure 1),<br />

coordinated movements (Figure 2) and<br />

eye-tracking (Figure 3), and recent work<br />

has been investigating how learning<br />

changes with healthy age.<br />

Figure 2: Unimanual action training showing feedback<br />

Action Selection and Execution<br />

Skilled movement entails selecting<br />

a task appropriate movement and<br />

then executing it correctly. There<br />

are numerous factors that influence<br />

which action you select and how you<br />

perform it, including the affordances<br />

<strong>of</strong> the task environment, immediate<br />

movement history, and your ongoing<br />

perceptual monitoring <strong>of</strong> the world.<br />

I have a collaboration to investigate<br />

these processes in a more complex task,<br />

namely targeted and distance throwing.<br />

These are behaviours unique to humans<br />

that are considered to have played<br />

an essential role in our evolutionary<br />

success. Throwing entails perception<br />

<strong>of</strong> object affordances (suitability for<br />

throwing) as well as the coordination<br />

and control <strong>of</strong> the dynamics <strong>of</strong> throwing<br />

and projectile motion to actually<br />

execute the throw.<br />

Overseas Collaborators<br />

Ge<strong>of</strong>frey P. Bingham, Winona Snapp-<br />

Childs – Indiana University<br />

Qin Zhu – University <strong>of</strong> Wyoming<br />

Figure 3: Typical eye kinematics for an observer viewing<br />

two dots moving at 180° mean relative phase.<br />

Developmental Coordination<br />

Disorder (DCD)<br />

As many as 5% <strong>of</strong> young children have<br />

serious difficulties in producing skilled<br />

movements. Using many <strong>of</strong> the standard<br />

techniques mentioned above, I am<br />

interested in understanding the source<br />

<strong>of</strong> their difficulties and in developing<br />

interventions that allow them to learn<br />

skilled movements.<br />

More information: http://www.fbs.leeds.<br />

ac.uk/staff/pr<strong>of</strong>ile.php?tag=Wilson_A<br />

Representative Publications<br />

Wilson AD, Snapp-Childs W & Bingham GP (in<br />

press) Improved perception leads immediately<br />

to improved movement stability. Journal <strong>of</strong><br />

Experimental Psychology: Human Perception and<br />

Performance<br />

Elders V, Sheehan S, Wilson AD, Levesley M,<br />

Bhakta B & Mon-Williams M (2010) Head-torsohand<br />

coordination in children with and without<br />

coordination difficulties. Developmental<br />

Medicine and Child Neurology 52(3): 238-243<br />

van Swieten LM, van Bergen E, Williams JGH,<br />

Wilson AD, Plumb MS, Kent SW & Mon-Williams M<br />

(2010) A test <strong>of</strong> motor (not executive) planning in<br />

developmental coordination disorder and autism.<br />

Journal <strong>of</strong> Experimental Psychology: Human<br />

Perception and Performance 36(2): 493-499<br />

Kent SW, Wilson AD, Plumb MS, Williams JHG &<br />

Mon-Williams M (2009) Immediate movement<br />

history influences reach-to-grasp action selection<br />

in children and adults. Journal <strong>of</strong> Motor Behavior<br />

41(1): 10-15<br />

Wilson AD & Bingham GP (2008) Identifying the<br />

information for the visual perception <strong>of</strong> relative<br />

phase. Perception & Psychophysics 70(3): 465-476


Ian Wood<br />

BSc, Imperial College, University <strong>of</strong> London<br />

PhD, University College London, University <strong>of</strong> London<br />

Postdoc Scripps Research <strong>Institute</strong>, La Jolla, USA<br />

Postdoc, University College London; Research Fellowship University <strong>of</strong> Leeds<br />

Lecturer, University <strong>of</strong> Leeds (2000-2007)<br />

Senior Lecturer University <strong>of</strong> Leeds (2007-)<br />

Contact: i.c.wood@leeds.ac.uk<br />

Uncovering the molecular mechanisms that control<br />

the gene expression in human disease<br />

We are interested in identifying<br />

the molecular mechanisms that<br />

are important in regulating gene<br />

transcription in human disease. Our<br />

work uses many molecular biological<br />

techniques, in vitro and in vivo model<br />

systems as well as clinical samples to<br />

provide a complete understanding <strong>of</strong><br />

disease mechanisms.<br />

Cardiovascular disease: Smooth<br />

muscle cells within blood vessels are<br />

important for controlling blood flow<br />

and pressure. In response to damage<br />

these cells proliferate to produce<br />

new smooth muscle cells which are<br />

important for vascular repair, but<br />

excessive proliferation is a major factor<br />

in cardiovascular diseases such as<br />

atherosclerosis, in-stent restenosis and<br />

a contributing factor to cardiovascular<br />

disease associated with diabetes.<br />

We have recently identified that the<br />

transcription factor REST plays an<br />

important role in normal blood vessels<br />

by repressing genes important for<br />

Figure 1: REST recruits several corepressor complexes to<br />

DNA to regulate gene expression. Adapted from Ooi L and<br />

Wood IC. Nature Reviews Genetics 8, 544-554. Copyright<br />

(2007)<br />

smooth muscle cell proliferation,<br />

including a specific potassium<br />

channel (IKCa) that is important for<br />

vascular smooth muscle proliferation.<br />

We are currently interested in the<br />

other molecular mechanisms that<br />

are responsible for increasing IKCa<br />

expression levels in disease as well<br />

as identifying the contributions <strong>of</strong><br />

environmental factors in promoting<br />

changes in gene expression.<br />

Neuronal disease: Correct functioning<br />

<strong>of</strong> the nervous system requires that<br />

neurones are able to communicate<br />

with each other effectively. Neurones<br />

transmit signals via propagation <strong>of</strong><br />

action potentials, the regulation <strong>of</strong><br />

which is <strong>of</strong> utmost importance. One ion<br />

channel important in determining the<br />

excitability <strong>of</strong> neurones is the M-channel<br />

which is composed <strong>of</strong> subunits <strong>of</strong><br />

the KCNQ potassium channel gene<br />

family. Mutations in KCNQ genes have<br />

been linked to heart disease, epilepsy,<br />

deafness and most recently pain.<br />

Despite their obvious importance, very<br />

little is known about how expression<br />

<strong>of</strong> these potassium channel genes<br />

is regulated. We are interested in<br />

determining how expression <strong>of</strong> these<br />

genes is regulated in normal physiology<br />

and in neuronal disorders such as<br />

epilepsy and chronic pain.<br />

Cancer: The transcription factor REST<br />

is associated with neuronal and nonneuronal<br />

cancers. REST is normally<br />

expressed at very low levels in neurones<br />

though increased REST expression is<br />

associated with a type <strong>of</strong> childhood<br />

brain cancer – medulloblastoma. In<br />

non-neuronal cells REST is normally<br />

expressed at quite high levels and<br />

reduced REST expression has recently<br />

been associated with colon cancer and<br />

may also be important in other cancers<br />

such as breast cancer. We are currently<br />

investigating a potential role for REST<br />

in bladder cancer and determining<br />

the cell specific effects <strong>of</strong> altered<br />

REST expression to understand the<br />

mechanisms by which REST can act as a<br />

tumour suppressor or an oncogene.<br />

Funding: British Heart Foundation<br />

More information:<br />

http://www.fbs.leeds.ac.uk/staff/pr<strong>of</strong>ile.<br />

php?tag=wood_I<br />

Representative Publications<br />

Johnson R, Samuel J, Ng CKL, Jauch R, Stanton<br />

LW, Wood IC (2009) Evolution <strong>of</strong> the Vertebrate<br />

Gene Regulatory Network Controlled by the<br />

Transcriptional Repressor REST. Mol Biol Evol<br />

26(7): 1491-1507<br />

Ooi L, Wood IC (2008) Regulation <strong>of</strong> gene<br />

expression in the nervous system. Biochem J 414:<br />

327-341<br />

Ooi L, Wood IC (2007) Chromatin crosstalk in<br />

development and disease: lessons from REST. Nat<br />

Rev Genet 8(7): 544-54<br />

Bingham AJ, Ooi L, Kozera L, White E, Wood<br />

IC (2007) The repressor element 1-silencing<br />

transcription factor regulates heart-specific gene<br />

expression using multiple chromatinmodifying<br />

complexes. Mol Cell Biol 27(11): 4082-92<br />

Ooi L, Belyaev ND, Miyake K, Wood IC, Buckley<br />

NJ (2006) BRG1 chromatin remodelling activity<br />

is required for efficient chromatin binding by<br />

repressor element 1-silencing transcription factor<br />

(REST) and facilitates REST-mediated repression. J<br />

Biol Chem 281(51): 38974-80<br />

Bruce AW, Krejci A, Ooi L, Deuchars J, Wood IC,<br />

Dolezal V, Buckley NJ (2006) The transcriptional<br />

repressor REST is a critical regulator <strong>of</strong> the<br />

neurosecretory phenotype. J Neurochem 98(6):<br />

1828-40<br />

Johnson R, Gamblin RJ, Ooi L, Bruce AW,<br />

Donaldson IJ, Westhead DR, Wood IC, Jackson<br />

RM, Buckley NJ (2006) Identification <strong>of</strong> the REST<br />

regulon reveals extensive transposable elementmediated<br />

binding site duplication. Nucleic Acids<br />

Res 34(14): 3862-77

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!