11.01.2013 Views

conference booklet - MDC

conference booklet - MDC

conference booklet - MDC

SHOW MORE
SHOW LESS

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

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

International Conference „Development of Somatosensation and Pain 2008“<br />

Welcome Address<br />

Dear Friends and Colleagues,<br />

It's our pleasure to invite you to the International Berlin Spring Meeting 2008<br />

"Development and function of somatosensation and pain" in the Max Delbrück<br />

Communications Center in Berlin-Buch from 14 - 17 May 2008.<br />

The aim of the meeting would be to bring together researchers interested in the<br />

development and function of the somatosensory system. Much progress has been<br />

made in recent years in elucidating the molecular mechanisms responsible for the<br />

phenotypic differentiation of primary afferent neurons as well as spinal dorsal horn<br />

neuronal circuits to which sensory neurons connect. We now also understand a lot<br />

more about the molecules (ion channels and other proteins) that allow sensory<br />

neurons to detect biologically relevant thermal and mechanical stimuli.<br />

The aim of this <strong>conference</strong> would be draw together top researchers working in these<br />

two related fields. This unique constellation of researchers would allow participants to<br />

explore the idea that the development of the somatosensory system may in turn help<br />

us also to understand its functional organization and pathology in human disease.<br />

The Max Delbrück Center (<strong>MDC</strong>) Conferences are international symposia dedicated to<br />

timely topics in Molecular Medicine and Translational Research. The Conference<br />

Series aims at fostering exchange of scientific information in all areas relevant to<br />

modern biomedical sciences including cell and developmental biology; molecular<br />

cardiovascular, neuroscience and oncology research; genetics, genomics and<br />

bioinformatics; as well as molecular pharmacology.<br />

You are invited to participate in what promises to become an exciting scientific<br />

meeting. We look forward to welcoming you in Berlin, an exiting European capital city.<br />

Yours sincerely,<br />

The Organizer<br />

Gary Lewin<br />

1 Welcome Address


Organizers & Scientific Advisory Board<br />

Organizers<br />

• Gary Lewin – Berlin, Germany<br />

• Carmen Birchmeier – Berlin, Germany<br />

• Ines Ibanez Talon – Berlin, Germany<br />

• Michael Schaefer – Berlin, Germany<br />

• Christoph Stein – Berlin, Germany<br />

Scientific Advisory Board<br />

• Carlos Belmonte – Alicante, Spain<br />

• Patrik Ernfors – Stockholm, Sweden<br />

• Martyn Goulding – La Jolla, USA<br />

• Thomas Jessell – New York, USA<br />

• Qiufu Ma – Boston/Cambridge, USA<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Organizers & Scientific Advisory Board 2


International Conference „Development of Somatosensation and Pain 2008“<br />

Organization<br />

Michaela Langer / Jana Droese<br />

Max Delbrück Center for Molecular Medicine (<strong>MDC</strong>) Berlin-Buch<br />

Robert-Rössle-Str. 10<br />

13125 Berlin<br />

phone: +49 30 9406 3720 / 4254<br />

fax: +49 30 9406 2206 / 2170<br />

email: langer@mdc-berlin.de / jana.droese@mdc-berlin.de<br />

Conference Office:<br />

Phone : +49 30 9406 3720 / 4824<br />

Fax: +49 30 9406 2206 / 2170<br />

3 Organization


Conference Information<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Venue:<br />

Max Delbrück Communications Center (<strong>MDC</strong>.C), Robert-Rössle-Str. 10, 13125<br />

Berlin-Buch<br />

Germany<br />

Contact:<br />

Conference secretariat (<strong>MDC</strong>.C), Michaela M. Langer,<br />

phone: +49 30 9406 3720/4824 / fax: +49 30 9406 2206 / e-mail: langer@mdcberlin.de<br />

Date:<br />

From Wednesday, May 14, 2008 afternoon to Saturday, May 17, 2008 3:00 p.m.<br />

Registration fee (late):<br />

Full registration fee 470 €<br />

Trainees 270 €<br />

Daily registration 100 €<br />

Conference Dinner 50 €<br />

The registration fee includes: Access to scientific sessions; Program and abstract<br />

book; free access to the Internet; Coffee breaks; Working lunches; Welcome reception<br />

and Evening Buffet. The Conference Dinner is NOT included in the registration fee.<br />

Posters: Posters will be displayed during the meeting close to the <strong>conference</strong> hall.<br />

The size of a single poster should be no more than 1m (horizontally) x 1.20m<br />

(vertically).<br />

In this abstract volume, you can find a number for your abstract and according to<br />

these numbers your poster can be mounted in the exhibition room.<br />

Mounting material will be available from the registration desk.<br />

Internet café will be located in the 3rd floor, Seminar room Dendrit 1. WLAN is<br />

available.<br />

Social events:<br />

* Welcome reception<br />

Wednesday, May 14, 7.15 p.m. – 9.30 p.m. - Venue: Max Delbrück Communications<br />

Center (<strong>MDC</strong>.C)<br />

* Evening Buffet with Poster Session<br />

Thursday, May 15, 6.00 p.m. – 8.00 p.m. - Venue: Max Delbrück Communications<br />

Center (<strong>MDC</strong>.C)<br />

* Conference Dinner<br />

Friday, May 16, 7.00 p.m. – downtown –<br />

Venue: Restaurant “Guy” Am Gendarmenmarkt, Jägerstrasse 59-60, 10117 Berlin<br />

Information at the Registration desk<br />

Buses<br />

Please notice the information at the Registration desk every day!<br />

Conference Information 4


International Conference „Development of Somatosensation and Pain 2008“<br />

Table of Contents<br />

Conference Information 4<br />

Program 6 - 11<br />

Speakers Abstracts 12 - 42<br />

List of Posters 43 - 46<br />

Poster Abstracts 47 - 74<br />

List of Participants 75 - 82<br />

Author Index 83 - 85<br />

5 Table of Contents


Program<br />

WEDNESDAY, May 14, Plenary Lectures<br />

18:00 – 18:10 Introduction<br />

Gary Lewin<br />

18:10 – 19:10 Using Pain to Block Pain<br />

Clifford Woolf<br />

19:10 Welcome Buffet<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Program 6


International Conference „Development of Somatosensation and Pain 2008“<br />

THURSDAY, May 15<br />

7 Program<br />

Development of Sensory Pathways I<br />

Chair: Carmen Birchmeier<br />

09:00 – 09:30 A new mechanoreception assay reveals intrinsic differences<br />

between sensory subtypes<br />

Patrik Ernfors<br />

09:30 – 10:00 Molecular characterization of low-threshold mechanoreceptor subtypes.<br />

Patrick Carroll<br />

10:00 – 10:15 The homeodomain factor Lbx1 controls the differentiation of<br />

sensory relay neurons in the hindbrain<br />

Robert Storm<br />

10:15 – 10:45 Coffee Break<br />

10:45 – 11:00 A cGMP signalling pathway essential for sensory axon bifurcation<br />

Hannes Schmidt<br />

11:00 – 11:30 NGF signalling and the control of gene expression in developing<br />

somatosensory neurons.<br />

David Ginty<br />

11:30 – 14:00 LUNCH (Buffet) and Poster session<br />

Sensory Transduction Mechanisms I<br />

Chair: Gary Lewin<br />

14:00 – 14:30 Deconstructing the molecular events responsible for touch and<br />

temperature sensation in C. elegans<br />

Miriam Goodman<br />

14:30 – 14:45 Defining a function for the ion channel TRPA1<br />

Sandra Zurborg<br />

14:45 – 15:15 The K2P channels: focus on TREK-1<br />

Eric Honore<br />

15:15 – 15.30 SCF/c-Kit is required for normal noxious heat sensitivity<br />

Alistair Gerratt<br />

15:30 – 16:00 Modulation of thermo-TRP ion channels by phosphorylation<br />

Peter McNaughton<br />

16:00 – 16:30 Coffee Break


Pain Mechanisms I<br />

Chair: Ines Ibanez Tallon<br />

16:30 – 17:00 Trk(ing) on from Pain.<br />

Lorne Mendell<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

17:00 – 17:30 Neuronal circuits and receptors involved in spinal cord pain<br />

processing<br />

Andrew Todd<br />

17:30 – 18:00 New insights into mechanisms of spinal LTP and pain memory<br />

Rohini Kuner<br />

18:00 – 20:00 Poster Session (with CHEESE and WINE)<br />

Program 8


International Conference „Development of Somatosensation and Pain 2008“<br />

FRIDAY, May 16<br />

09:00 – 09:30 TBA<br />

Martyn Goulding<br />

9 Program<br />

Development of Somotosensory pathways II<br />

Chair: Alistair Garratt<br />

09:30 – 10:00 Transcriptional Control in Dorsal Spinal Cord Development: New<br />

Roles for Old Factors<br />

Jane Johnson<br />

10:00 – 10:15 The Novel Estrogen Receptor GPR30 Mediates Estrogen-Induced<br />

And PKCepsilon -Dependent Mechanical Hyperalgesia In Vitro<br />

And In Vivo<br />

Tim Hucho<br />

10:15 – 10:45 Coffee Break<br />

10:45 – 11:00 Adaptations in the C Fiber Nociceptors of Naked Mole-Rats<br />

Render These Animals Insensitive to Specific Air-Borne Irritants<br />

and CO2-Induced Pulmonary Edema<br />

Thomas Park<br />

11:00 – 11:30 Tactile Experience Shapes Behaviour in Etruscan Shrews<br />

Michael Brecht<br />

11:30 – 12:00 Specification and differentiation of dorsal spinal cord interneurons.<br />

Carmen Birchmeier<br />

12:00 – 14:00 LUNCH (Buffet) and Poster Sessions<br />

Pain Mechanisms II<br />

Chair: Christoph Stein<br />

14:00 – 14:30 The menthol receptor TRPM8 is the principal detector of<br />

environmental cold<br />

Jan Siemens<br />

14:30 – 14:45 Pain attenuation in Tethered-Toxin- transgenic mice due to<br />

reduced excitability of sensory neurons<br />

Ines Ibanez-Tallon<br />

14:45 – 15:15 Acid-Sensing Ion Channels (ASICs) and pain in the central and<br />

peripheral<br />

Eric Lingueglia<br />

15:30 – 16:00 Coffee Break and Poster Session<br />

15:15 – 15:30 Leukocyte-derived and exogenous opioids acting at peripheral<br />

opioid receptors control neuropathic pain<br />

Dominika Labuz


May 14 – May 17, 2008 – Berlin, Germany<br />

16:00 – 16:30 Synaptic Dis-Inhibition in Pathological Pain States<br />

Hanns-Ulrich Zeilhofer<br />

19:00 GET TOGETHER and Congress Dinner (downtown)<br />

Program 10


International Conference „Development of Somatosensation and Pain 2008“<br />

SATURDAY, May 17<br />

11 Program<br />

Transduction, Development, and Pain<br />

Chair: Michael Schaefer<br />

09:00 – 09:30 Mechanosensitive ion channels, stomatin-like proteins and<br />

molecular tethers essential for touch.<br />

Gary Lewin<br />

09:30 – 10:00 Sensory Neuron Mechanotransduction<br />

John Wood<br />

10:00 – 10:30 Breathing with Phox2b<br />

Jean-Francois Brunet<br />

10:30 – 11:00 Coffee Break<br />

Transduction, Development, and Pain<br />

Chair: Robert Schmidt<br />

11:00 – 11:30 Roles of Runx1 in controlling nociceptor development and pain<br />

behaviors<br />

Qiufu Ma<br />

11:30 – 12:00 Peripheral mechanisms for cold detection in intact and injured<br />

tissues<br />

Carlos Belmonte<br />

12:00 – 12:30 Genetic Determinants of Pain<br />

Stephen McMahon<br />

12:30 – 14:00 LUNCH (Buffet)


Speaker Abstracts<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Speaker Abstracts 12


International Conference „Development of Somatosensation and Pain 2008“<br />

T1 Using Pain to Bock Pain<br />

Clifford J. Woolf<br />

Department of Anesthesia and Critical Care, Massachusetts General Hospital and<br />

Harvard Medical School, Boston USA<br />

We find that it is possible to selectively block electrical signaling in nociceptors without<br />

affecting signaling by other neurons (Binshtok et al Nature 449: 607-611, 2007). The<br />

method is based on introducing permanently charged sodium channel blockers<br />

through the pore of TRPV1 channels which are present on nociceptors but not motor,<br />

autonomic, or low threshold mechanosensitive neurons. TRPV1 channels are<br />

activated by noxious heat and protons, and also by pungent ligands such as<br />

capsaicin. The pore of the TRPV1 channel is large enough to pass N-methyl-lidocaine<br />

(QX-314), a positively charged hydrophilic lidocaine derivative that is normally<br />

ineffective as a local anesthetic because it cannot permeate through the membrane to<br />

access the lidocaine-binding site on the inner face of voltage-gated sodium channels.<br />

By co-applying capsaicin and N-methyl-lidocaine, it is possible to introduce the Nmethyl-lidocaine<br />

selectively into nociceptors, where it blocksTTX-sensitive and<br />

resistant sodium channels in these neurons to prevent action potential firing. Unlike<br />

conventional local anesthetics, which block electrical activity in all neurons, this<br />

produces a local anesthesia without paralysis or numbness, effectively creating a local<br />

analgesia. Also, the inhibition of pain lasts longer than conventional local anesthetics,<br />

since the charged sodium channel blocker is effectively trapped inside the nociceptor<br />

after the two agents are applied and removed. This approach should lead to the<br />

development of novel combination analgesics comprising agonists of TRP channels to<br />

activate intrinsic drug delivery systems, and impermeant cationic ion channel blockers<br />

whose acces to select cell types is determined by the expression profile of the TRPs.<br />

13 Speaker Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

T2 A new mechanoreception assay reveals intrinsic differences between<br />

sensory subtypes<br />

Patrik Ernfors<br />

Division of Molecular Neurobiology, Department of Medical Biochemistry and<br />

Biophysics Scheelesv 1 Karolinska Institute, S-171 77 Stockholm, Sweden<br />

The somatic sensory system comprises different perceptual modalities. Neurons of the<br />

dorsal root ganglion mediate tactile sensation by mechanoreceptive stimuli, limb<br />

proprioceptive sensation elicited by displacement and the static tension of muscles,<br />

and pain and thermal sensations. .A new functional in vitro assay has been<br />

developed. Groups of neurons have similar response profiles and different neurons<br />

respond to different types and levels of mechanical stimuli. Hence, this in vitro assay<br />

can discriminate between different mechanoreceptors in terms of their stimuli<br />

response profile. This suggests that inherent differences in vivo is preserved in vitro<br />

and could thus be a selectable marker for a neuronal subtype. In this assay, nerve<br />

ending are subjected to pressure in a three dimensional matrix which leads to an<br />

action potential to the cell body where measurements are taken, and is thus very<br />

similar to the in vivo situation where mechanosensation is generated in the periphery<br />

due to local deformation or vibration.<br />

Speaker Abstracts 14


International Conference „Development of Somatosensation and Pain 2008“<br />

T3 Molecular characterization of low-threshold mechanoreceptor sub-types.<br />

Patrick Carroll<br />

INSERM U.583, Physiopathologie et thérapie des déficits sensoriels et moteurs.<br />

Institut des Neurosciences de Montpellier (INM), Hopital St. Eloi, 80 rue Augustin<br />

Fliche, BP 74103, 34091 Montpellier cedex 5<br />

Low-threshold mechanoreceptor neurons of the dorsal root ganglia (DRG) have been<br />

difficult to study because of the dearth of specific molecular markers of these celltypes.<br />

In a screen for genes expressed in specific DRG sub-types we found that a<br />

transcription factor of the large Maf family (Maf-A) is expressed in 8-10% of embryonic<br />

DRG neurons. By co-localization studies with known markers of sensory neuron subtypes<br />

MafA was found to be exclusively co-localized with the tyrosine kinase receptor<br />

c-Ret. Double Maf-A+/Ret+ neurons are observed in the DRG at early developmental<br />

stages (E11 – E13) before the appearance of c-Ret in nociceptors at late embryonic<br />

stages. Analysis of the peripheral and central projections of these neurons at P0<br />

suggests that they are slowly-adapting mechanoreceptors. Preliminary<br />

characterization, using mutant mice, of the roles MafA and c-Ret in these<br />

mechanoreceptors will be presented.<br />

15 Speaker Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

T4 The homeodomain factor Lbx1 controls the differentiation of sensory relay<br />

neurons in the hindbrain<br />

Robert Storm, Thomas Müller, Martin Sieber, Carmen Birchmeier<br />

Max-Delbrück-Centrum für Molekulare Medizin (<strong>MDC</strong>)<br />

The homeodomain factor Lbx1 is expressed in postmitotic neurons in the ventral alar<br />

plate of the developing hindbrain extending from rhombomere 2 into the spinal cord.<br />

Lbx1 expression distinguishes two major classes of postmitotic neurons: class A<br />

(Lbx1-negative) and class B (Lbx1-positive). Here, we provide a classification of the<br />

neuronal subtypes emerging in the alar plate of the hindbrain according to their<br />

transcription factor profiles. Genetic lineage tracing allowed us to follow Lbx1<br />

derivatives throughout embryonic development in control and Lbx1 mutant mice. We<br />

show that most of the Pax2+, GABAergic neurons in the caudal hindbrain and Lmx1b+<br />

somatosensory neurons of the spinal trigeminal nucleus are Lbx1 derivatives. In Lbx1<br />

mutant mice, dorsal Pax2+ neurons are not specified and the spinal trigeminal nucleus<br />

is not formed. Instead, the viscerosensory nucleus of the solitary tract (NTS) as well<br />

as the inferior olive (IO) are enlarged. In normal development the NTS and the IO are<br />

formed by dA3 and dA4 neurons, respectively. We conclude that Lbx1 is a major<br />

determinant in sensory neuron development, which chooses a somatosensory over a<br />

viscerosensory and climbing fiber developmental program in the caudal hindbrain.<br />

Speaker Abstracts 16


International Conference „Development of Somatosensation and Pain 2008“<br />

T5 A cGMP SIGNALING PATHWAY ESSENTIAL FOR SENSORY AXON<br />

BIFURCATION<br />

Hannes Schmidt 1 , Agne Stonkute 1 , René Jüttner 1 , Susanne Schäffer 1 ,<br />

Jens Buttgereit 2 , Robert Feil 3 , Franz Hofmann 3 , Fritz G. Rathjen 1<br />

1 Max Delbrück Center for Molecular Medicine, Developmental Neurobiology Group,<br />

Berlin, Germany, 2 Max Delbrück Center for Molecular Medicine, Molecular Biology of<br />

Peptide Hormones Group, Berlin, Germany, 3 Technical University of Munich,<br />

Department of Pharmacology and Toxicology, Munich, Germany<br />

The complex wiring pattern of the mature nervous system is shaped during<br />

development by the phenomena of axonal guidance and branching. The intracellular<br />

signal transduction mechanisms underlying these processes are only poorly resolved.<br />

We identified a signalling axis in sensory neurons essential for one form of axonal<br />

branching. The central trajectories of dorsal root ganglion axons display at least two<br />

types of ramifications when they enter the spinal cord: (1) bifurcation at the dorsal root<br />

entry zone (DREZ) and (2) interstitial branching from stem axons to generate<br />

collaterals that penetrate the grey matter. We report a cGMP signaling cascade<br />

critically involved in the establishment of the highly stereotyped pattern of T-shaped<br />

axon bifurcation of sensory axons at the DREZ of the spinal cord. Single axon labeling<br />

using DiI revealed that embryonic mice with an inactive receptor guanylyl cyclase<br />

Npr2 or deficient for cGMP-dependent protein kinase I (cGKI) lack the bifurcation of<br />

sensory axons at the DREZ, i.e. the ingrowing axon either turns rostrally or caudally<br />

instead. Cross-breeding experiments of these mutant mice with a mouse line<br />

expressing EGFP in sensory neurons under control of the Thy-1 promoter<br />

demonstrate that the bifurcation error is maintained to mature stages. In contrast,<br />

interstitial branching of collaterals from primary stem axons remains unaffected. At a<br />

functional level, the distorted axonal branching is accompanied by reduced synaptic<br />

input, as revealed by patch clamp recordings of neurons in the superficial layers of the<br />

cord. Hence, our data demonstrate that a cGMP signaling cascade including Npr2 and<br />

cGKI is essential for axonal bifurcation at the DREZ and influences neuronal<br />

connectivity in the dorsal spinal cord.<br />

17 Speaker Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

T6 NGF signaling and the control of gene expression in developing<br />

somatosensory neurons.<br />

S. Rasika Wickramasinghe, Rebecca S. Alvania, Narendrakumar Ramanan,<br />

Kenji Mandai, David D. Ginty<br />

The Solomon H. Snyder Department of Neuroscience, The Howard Hughes Medical<br />

Institute, The Johns Hopkins University School of Medicine, 725 N. Wolfe St, PCTB<br />

1015, Baltimore, MD 21205.<br />

Nerve Growth Factor (NGF) controls survival, maturation and target innervation by<br />

cutaneous sensory neurons. We report that Serum Response Factor (SRF), a<br />

prototypic transcription factor that mediates stimulus-dependent gene expression, is a<br />

critical mediator of NGF signaling, axonal growth, branching, and target innervation by<br />

embryonic DRG sensory neurons. Conditional ablation of the murine SRF gene in<br />

DRGs results in no deficits in sensory neuron viability or differentiation, but causes<br />

dramatic defects in extension and arborization of peripheral axonal projections in the<br />

target field in vivo, a phenotype also observed in mice lacking NGF. Moreover, SRF is<br />

both necessary and sufficient for NGF-dependent axonal outgrowth in vitro, and NGF<br />

regulates SRF-dependent gene expression and axonal outgrowth through activation of<br />

both MEK/ERK and MAL signaling pathways. Finally, we found that NGF is essential<br />

for the expression of several SRF-dependent cytoskeletal genes in embryonic DRG<br />

neurons in vivo. Together, our findings suggest that SRF is a major effector of both<br />

MEK/ERK and MAL signaling by NGF, and that SRF is a key mediator of NGFdependent<br />

target innervation by embryonic sensory neurons.<br />

Speaker Abstracts 18


International Conference „Development of Somatosensation and Pain 2008“<br />

T7 Deconstructing the molecular events responsible for touch and temperature<br />

sensation in C. elegans<br />

Miriam Goodman<br />

Stanford University, Stanford, CA 94305 USA<br />

Sensation guides behavior, providing critical information about the environment. The<br />

senses of touch and vibration are critical for daily activities like standing and walking,<br />

while temperature sensation is essential for efficient thermoregulation. We seek to<br />

understand the molecular events that give rise to touch and temperature sensation<br />

using the nematode Caenorhabditis elegans as a model system. Powerful tools in<br />

classical and molecular genetics and the ability to record electrical responses to<br />

sensory stimuli in living animals make this simple roundworm a nearly perfect animal<br />

for these studies. Our work focuses on the six non-ciliated touch receptor neurons<br />

(TRNs) that detect touch applied to the body wall and a pair of thermosensory neurons<br />

(AFD) that detect changes in ambient temperature. Genetic analyses have shown that<br />

TRN function depends on degenerins encoded by the mec-4 and mec-10 genes, while<br />

AFD function depends on cGMP-gated ion channels encoded by tax-2 and tax-4. We<br />

are using in vivo whole-cell patch-clamp recording in concert with genetic dissection to<br />

investigate the biophysics and molecular basis of touch and temperature transduction.<br />

We have used this approach to show that MEC-4 and MEC-10 form the pore of native<br />

mechanotransduction channels and that such channels rely on the MEC-2 and MEC-6<br />

auxiliary subunits for full functionality in vivo (Nat Neurosci 8: 43). Now, we are<br />

investigating the mechanism by which these channels are activated in vivo. We have<br />

also used in vivo whole-cell patch clamp recording to characterize the biophysical and<br />

molecular basis of temperature-sensitivity in AFD neurons. I will present evidence that<br />

the AFD neurons detect cooling and warming by modulating the activity of the cGMPgated<br />

ion channel encoded by tax-4 and tax-2.<br />

19 Speaker Abstracts


T8 Defining a function for the ion channel TRPA1<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Sandra Zurborg, Brian Yurgionas, Ombretta Caspani, Paul A. Heppenstall<br />

TRPA1 is a non-selective ion channel belonging to the transient receptor potential<br />

(TRP) family. The channel is expressed by a subset of pain sensing neurons in the<br />

peripheral nervous system and can be activated by many pain eliciting compounds<br />

e.g. acrolein and mustard oil. Its endogenous function remains unclear, although it is<br />

generally accepted that TRPA1 is important in the pain pathway. In my PhD. project I<br />

have working on defining a function for TRPA1 at the cellular and whole animal level.<br />

In initial studies we demonstrated that TRPA1 is directly gated by intracellular Ca2+<br />

and can be activated by several stimuli which raise intracellular Ca2+ levels in sensory<br />

neurons. We identified an EF-hand domain in the N-terminus of TRPA1 and<br />

demonstrated that it is responsible for the calcium sensitivity of the channel. More<br />

recently we have been investigating the expression of TRPA1 in mouse models of<br />

inflammatory pain because changes in the expression of TRPA1 might be a critical<br />

mechanism for modifying excitability of painsensing neurons after injury. In order to<br />

model inflammatory pain we injected Complete Freund's Adjuvant (CFA) into the hind<br />

paw of mice. We assessed TRPA1 expression by means of calcium imaging of<br />

dissociated dorsal root ganglia (DRG) neurons and in situ hybridization of DRG<br />

sections after inflammation. We saw no change in the TRPA1 expression when we<br />

compared untreated mice and inflamed animals. However we did observe significantly<br />

larger mustard oil evoked responses in CFA-treated mice compared to control mice.<br />

This suggests that TRPA1 might contribute to enhanced excitability and<br />

hypersensitivity during inflammation.<br />

Speaker Abstracts 20


International Conference „Development of Somatosensation and Pain 2008“<br />

T9 The K2P channels: focus on TREK-1<br />

Eric Honore<br />

Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 6097, Université de<br />

Nice-Sophia Antipolis, 660 route des Lucioles, 06560 Valbonne, France.<br />

Two-pore-domain K+ (K2P) channel subunits are made up of four transmembrane<br />

segments and two pore-forming domains that are arranged in tandem and function as<br />

either homo- or heterodimeric channels. This structural motif is associated with<br />

unusual gating properties including background channel activity and sensitivity to<br />

membrane stretch. Moreover, K2P channels are modulated by a variety of cellular<br />

lipids and pharmacological agents, including polyunsaturated fatty acids and volatile<br />

general anesthetics. Recent in vivo studies have demonstrated that TREK-1, the most<br />

thoroughly studied K2P channel, has a key role in the cellular mechanisms of<br />

neuroprotection, anaesthesia, pain and depression.<br />

21 Speaker Abstracts


T10 SCF/c-Kit is required for normal noxious heat sensitivity<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Nevena Milenkovic 1 , Christina Frahm Frahm 2 , Carola Griffel 2 , Bettina Erdmann 3 ,<br />

Carmen Birchmeier 2 , Gary R. Lewin 1 , Alistair N. Garratt 2<br />

1 Molecular Physiology of Somatic Sensation, 2 Developemental biology/Signal<br />

Transduction, 3 Max-Delbrück Center for Molecular Medicine<br />

Sensory neurons in the dorsal root ganglion (DRG) transduce diverse sensory<br />

sensations like touch, heat and pain. C fibers represent about 60% of DRG cell<br />

population; most of them are polymodal responding both to noxious thermal and<br />

mechanical stimuli. Neurotrophins are known to be involved in determining the final<br />

phenotype of sensory neurons. NGF is a main mediator of inflammatory hyperalgesia<br />

and also critical for the development of normal noxious heat sensitivity. Lower noxious<br />

heat sensitivity and lower expression level of c-Kit after neonatal NGF deprivation<br />

make c-Kit a good candidate as mediator of noxious heat sensitivity. The receptor<br />

tyrosine kinase c-Kit was functionally characterized. In DRG c-Kit is predominately<br />

expressed in small diameter cells expressing TrkA and CGRP. We showed that<br />

SCF/c-Kit signaling system has a key role in setting the thermal threshold for<br />

activation of heat sensing nociceptive neurons. Mice lacking a functional c-Kit receptor<br />

displayed profound thermal hypoalgesia attributable to a marked elevation in the<br />

thermal threshold and reduction in spiking rate of CMH nociceptors. Activation of c-Kit<br />

by SCF resulted in a reduced thermal threshold and profound potentiation of heatactivated<br />

currents in 50% of isolated heat sensitive small diameter neurons. Acute<br />

application of SCF induced thermal hyperalgesia in mice and this action required the<br />

TRP-family cation channel TRPV1. In addition, lack of c-Kit signaling during<br />

development resulted in hypersensitivity of discrete mechanoreceptive neuronal<br />

subtypes to mechanical stimulation. The reduced noxious heat sensitivity was<br />

observed in mice treated with c-Kit blocker, Imatinib. The ability of SCF to potentiate<br />

capsaicin induced Ca2+ intracellular concentration in isolated DRG neurons is<br />

currently used to investigate the SCF/c-Kit signaling pathway. Thus c-Kit, can be be<br />

grouped into a small family of receptor tyrosine kinases, including c- Ret and TrkA,<br />

that control the transduction properties of distinct types of sensory neuron to thermal<br />

and mechanical stimuli.<br />

Speaker Abstracts 22


International Conference „Development of Somatosensation and Pain 2008“<br />

T11 Modulation of thermo-TRP ion channels by phosphorylation<br />

Xuming Zhang, Peter McNaughton<br />

Dept of Pharmacology, University of Cambridge<br />

The ability of vertebrates to detect and avoid damaging extremes of temperature<br />

depends on activation of ion channels belonging to the thermo-TRP family. Injury or<br />

inflammation lowers the threshold for detection of painful levels of heat, a process<br />

known as heat hyperalgesia. A wide range of inflammatory mediators, amongst which<br />

the best-studied are bradykinin, prostaglandin E2 and nerve growth factor (NGF), are<br />

liberated by inflammation or injury and are able to cause heat hyperalgesia.<br />

Inflammatory mediators activate at least three distinct intracellular signalling pathways<br />

which lower the threshold of the heat-activated ion channel TRPV1. Bradykinin<br />

activates PKC-epsilon and phosphorylates two serine residues on TRPV1, while<br />

prostaglandin E2 activates PKA and phosphorylates a partially overlapping set of<br />

residues. Both these mechanisms enhance the open probability of TRPV1 channels<br />

already located in the surface membrane. NGF, on the other hand, activates the<br />

tyrosine kinase Src to phosphorylate a single tyrosine in the N-terminal domain and so<br />

to promote movement of new channels from a vesicular store to the surface<br />

membrane. Some work had suggested that a reducution in membrane PIP2 was a<br />

major mechanism for modulation of TRPV1, but recent work in our lab and elsewhere<br />

has now discounted this possibility.<br />

In recent work we have shown that modulation of the sensitivity of TRPV1 by the<br />

protein kinases PKA and PKC, and by the phosphatase calcineurin, depends on the<br />

formation of a signalling complex between the scaffolding protein AKAP79/150 and<br />

TRPV1. We have identified a critical AKAP79/150 binding region in the TRPV1 Cterminal<br />

domain. If binding is prevented then sensitisation by both bradykinin and<br />

PGE2 is completely abrogated. AKAP79/150 is therefore a final common element,<br />

vital for heat hyperalgesia, on which the effects of multiple pro-inflammatory mediators<br />

converge.<br />

23 Speaker Abstracts


T12 Trk(ing) on from Pain<br />

J. Petruska, V. Boyce, L. M. Mendell<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Department of Neurobiology and Behavior, SUNY- Stony Brook, Stony Brook, NY,<br />

11794, USA<br />

Previous work from this laboratory has demonstrated that the neurotrophins NGF and<br />

BDNF can acutely sensitize peripheral and central components of the nociceptive<br />

pathway (Mendell, Handbook of the Senses, 2008). Further experiments revealed that<br />

another neurotrophin, NT-3, acutely sensitizes the monosynaptic spindle- evoked<br />

EPSP via trkC receptors (Arvanov et al., J. Neurophysiol. 2000). Additional work<br />

demonstrated that long lasting NT-3 administration to axotomized nerves or to the<br />

spinal cord during development strengthens synapses made by muscle spindle<br />

afferent fiber (group Ia) fibers on motoneurons (Mendell et al., J. Neurosci. 1999;<br />

Arvanian et al., J. Neurosci. 2003). Step training in spinal rats transected as neonates<br />

improves stepping performance. It also increases the monosynaptic EPSP in ankle<br />

extensor motoneurons (Petruska et al., J. Neurosci., 2007) which should improve hind<br />

limb weight bearing and facilitate initiation of the swing phase (“stepping off”).<br />

Because neurotrophin levels, including NT-3 are elevated in the spinal cord of step-<br />

trained spinal rats, we have investigated whether direct administration of NT-3 elicits<br />

effects similar to those of step training in neonatally transected rats. In the present<br />

work we made use of AAV-NT-3 viral constructs to administer NT-3. When injected<br />

into peripheral tissues they are transported to the spinal cord and DRG in an AAV<br />

serotype- dependent manner. NT-3 levels are substantially increased in the spinal<br />

cord for months after a single administration of the appropriate viral particles into<br />

hindlimb muscles. We have found electrophysiological changes in treated transected<br />

rats that are qualitatively similar to those observed after step training. Furthermore,<br />

these rats show improved stepping ability compared to untreated rats, in general<br />

agreement with findings in the cat (Boyce et al., J. Neurophysiol., 2007). This work<br />

may have translational potential. We are using vectors and neurotrophins delivered in<br />

a manner that is consistent with clinical application. Here we demonstrate that these<br />

agents elicit useful behavioral effects that may be explained at least in part by the<br />

electrophysiological changes we measure in the spinal cord. Drs. R. Ichiyama and<br />

V.R. Edgerton (UCLA) carried out behavioral aspects of this work. Drs. B. Kaspar and<br />

F. Gage (Salk Institute) designed and prepared the engineered AAV viruses.<br />

Supported by the Christopher and Dana Reeve Foundation, and NIH (2 RO1 NS<br />

16996).<br />

Speaker Abstracts 24


International Conference „Development of Somatosensation and Pain 2008“<br />

T13 Neuronal circuits and receptors involved in spinal cord pain processing<br />

Andrew Todd<br />

Spinal Cord Group, West Medical Building, University of Glasgow, University Avenue,<br />

Glasgow G12 8QQ, U.K.<br />

The spinal dorsal horn receives sensory information from primary afferents, and this is<br />

transmitted to the brain through projection neurons. It also contains numerous<br />

excitatory and inhibitory interneurons, and receives descending modulatory inputs.<br />

Despite its importance in pain mechanisms, we still know little about the neuronal<br />

circuitry within this region. Projection neurons are concentrated in lamina I and<br />

scattered throughout deeper laminae. Lamina I neurons in lumbar cord project to the<br />

lateral parabrachial area, periaqueductal grey matter and caudal medulla. There are<br />

few lamina I spinothalamic neurons in lumbar segments, although these cells are<br />

common in the cervical enlargement. The targets of lamina I spinothalamic neurons<br />

include the posterior triangular nucleus, which projects to second somatosensory and<br />

insular cortices. Most lamina I projection neurons express the neurokinin 1 receptor<br />

(NK1r). These cells, together with projection neurons in laminae III-IV that also<br />

express this receptor, are densely innervated by substance P-containing primary<br />

afferents. This provides a powerful route linking nociceptors with brain regions<br />

involved in pain. We have also identified a population of large lamina I projection cells<br />

that lack the NK1 receptor and have a high density of inhibitory synapses. Glutamate<br />

is the main excitatory transmitter in the dorsal horn and acts on several types of<br />

receptor, including AMPA receptors (AMPArs), which play a major role in perception of<br />

somatosensory stimuli. AMPArs are made up from four subunits (GluR1-4), and<br />

subunit composition determines receptor properties. Synaptic AMPArs are difficult to<br />

detect with immunocytochemistry because protein cross-linking at synapses following<br />

fixation. However, they can be revealed with antigen retrieval, and this can be used to<br />

determine the laminar distribution of different subunits and the subunit composition of<br />

synaptic AMPArs on different types of neuron. We have found that lamina III/IV NK1rexpressing<br />

neurons and the large lamina I cells that lack NK1r both have a high<br />

density of GluR4-containing synapses. Cells in the latter population receive numerous<br />

synapses from axons that contain vesicular glutamate transporter 2 (VGLUT2), and<br />

these probably originate from local excitatory interneurons. This suggests that GluR4containing<br />

AMPArs in the spinal cord may play an important role in pain mechanisms.<br />

25 Speaker Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

T14 New insights into mechanisms of spinal LTP and pain memory<br />

Rohini Kuner<br />

Pharmakologisches Institut, Universität Heidelberg, Im Neuenheimer Feld,<br />

Heidelberg, 69120 Germany<br />

The second messenger cGMP is a key mediator of sensitization at spinal synapses,<br />

which constitutes a cellular basis for chronic pain. Because cGMP can activate several<br />

ion-channels and enzymes pre- as well as post-synaptically, the precise identity of<br />

critical signaling molecules, their mechanisms of action and their locus in the spinal<br />

circuitry have remained unclear. I will present some results which indicate that the<br />

cGMP-dependent protein kinase 1 (PKGI) localized presynaptically in nociceptor<br />

terminals mediates long term potentiation at spinal synapses by enhancing<br />

presynaptic vesicular release. In nociceptors, we observed that PKGI produces<br />

activity-dependent phosphorylation of proteins, which directly regulate intracellular<br />

calcium signaling and vesicular release and set the excitation thresholds of painsensing<br />

nerves. Our results indicate that PKGI localized presynaptically in nociceptors<br />

is the most important spinal target of cGMP and represents a key convergence point<br />

of the NMDA-NO-soluble guanylyl cyclase pathway and the natriuretic peptidemembrane<br />

guanylyl cyclase pathway in mediating nociceptor sensitization and chronic<br />

pain.<br />

Speaker Abstracts 26


International Conference „Development of Somatosensation and Pain 2008“<br />

T16 Transcriptional Control in Dorsal Spinal Cord Development: New Roles for<br />

Old Factors<br />

Jane Johnson<br />

University of Texas Southwestern Medical Center<br />

The dorsal horn of the spinal cord contains the neuronal circuitry that modulates<br />

sensory input from the periphery. The formation of this circuitry, including the balance<br />

of inhibitory and excitatory neuronal subtypes, is initially generated through<br />

specification mechanisms controlled by transcription factors, particularly members of<br />

the homeodomain (HD) and the basic helix-loop-helix (bHLH) families. bHLH<br />

transcription factors such as Ascl1 (previously Mash1), Atoh1 (previously Math1) and<br />

Neurog1/2 (previously Ngn1/2) are in a balance with Notch signaling to regulate the<br />

number of progenitor cells undergoing neuronal differentiation as determined by cell<br />

cycle exit, movement out of ventricular zones, and expression of neuronal specific<br />

markers. In the developing dorsal spinal cord, these same bHLH factors, and related<br />

family members, function in neuronal subtype specification as well. For example, Ptf1a<br />

is essential for the generation of GABAergic inhibitory neurons in multiple regions of<br />

the nervous system. In the absence of Ptf1a, dorsal horn GABAergic neurons are misspecified<br />

to glutamatergic neurons. Probing deeper into the mechanism of Ptf1a<br />

function, the existence of a novel DNA binding complex was uncovered. This complex<br />

contains Ptf1a and its E-protein partner, plus Rbpj—the transducer of the Notch<br />

signaling pathway. We demonstrate the Ptf1a--Rbpj interaction is required in vivo for<br />

specification of the GABAergic neurons, a function that cannot be substituted by the<br />

classical form of the bHLH heterodimer with E-protein or Notch signaling through Rbpj.<br />

Thus, this unique Ptf1a-Rbpj complex controls the balanced formation of inhibitory and<br />

excitatory neurons in the developing spinal cord, and reveals a novel Notch<br />

independent function for Rbpj in nervous system development. The interplay between<br />

bHLH transcription factors and Notch signaling components occurs at multiple levels<br />

and is key in directing both neuronal differentiation and neuronal subtype specification.<br />

27 Speaker Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

T17 The Novel Estrogen Receptor GPR30 Mediates Estrogen-Induced And<br />

PKCepsilon -Dependent Mechanical Hyperalgesia In Vitro And In Vivo<br />

Julia Kuhn 1 , Olayinka Dina 2 , Jon Levine 2 , Tim Hucho 1<br />

1 Max Planck Institut für molekulare Genetik, 2 University of California, San Francisco<br />

The epsilon isoform of protein kinase C (PKCepsilon) is an important second<br />

messenger in models of acute as well as chronic mechanical sensitization. Only few<br />

receptors have been identified to lead to activation of PKCepsilon (beta2-adrenergic<br />

receptor, bradykinin receptor). Recently, we reported estrogen to activate PKCepsilon<br />

in primary sensory neurons as well as induce PKCepsilon-dependent sensitization in<br />

male rats. Which of the known estrogen receptors (estrogen receptor alpha (ERalpha),<br />

estrogen receptor beta (ERbeta), G-protein coupled receptor 30 (GPR30)) mediates<br />

this effect remained unknown. Here we show, that the agonist of the novel estrogen<br />

receptor GPR30, G-1, induces rapid PKCepsilon-translocation in primary nociceptive<br />

neurons. Also, ICI 182 780, which acts as an agonist of GPR30 while blocking the<br />

classical estrogen receptors ERalpha and ERbeta, leads to fast activation of<br />

PKCepsilon. In contrast, the specific agonists of ERalpha, PPT, and ERbeta, DPN, do<br />

not have any effect. RT-PCR studies show, indeed, GPR30 to be expressed in rat<br />

dorsal root ganglia. Also, in behavioural experiments intradermal injection of G-1 into<br />

the dorsum of the paw of male rats induces strong mechanical hyperalgesia in a<br />

concentration dependent manner. These data indicate an involvement of the novel<br />

estrogen receptor GPR30 in the modulation of nociceptive signaling pathways by sex<br />

hormones.<br />

Speaker Abstracts 28


International Conference „Development of Somatosensation and Pain 2008“<br />

T18 Adaptations in the C Fiber Nociceptors of Naked Mole-Rats Render These<br />

Animals Insensitive to Specific Air-Borne Irritants and CO2-Induced<br />

Pulmonary Edema<br />

Thomas Park 1 , Gary Lewin 2<br />

1 Laboratory of Integrative Neuroscience, Department of Biological Sciences,<br />

University of Illinois at Chicago, Chicago, Illinois, USA., 2 Max-Delbrück Center for<br />

Molecular Medicine, Robert-Rössle-Str. 10, Berlin-Buch D-13092 Germany and<br />

Charité-Universitätsmedizin, Berlin.<br />

African Naked mole-rats have a unique ecology in that they are fully subterranean and<br />

they live in very high numbers. Hence, the air in their poorly ventilated living space has<br />

chronically high concentrations of CO2 and ammonia. In the respiratory tract these<br />

chemical irritants activate C fiber nociceptors (also referred to as chemoreceptors or<br />

irritant detectors). We have identified two putative adaptations in the C fiber<br />

nociceptors of naked mole-rats that are consistent with evolving under the challenging<br />

conditions this species faces. First, their C fibers lack the neuropeptides Substance P<br />

(SP) and calcitonin gene related peptide (CGRP), and second, the fibers are<br />

unresponsive to low pH (acidosis). We hypothesized that these adaptations would<br />

render the mole-rats behaviorally insensitive to chemical irritants that act on trigeminal<br />

C fibers (high CO2, ammonia, capsaicin), and physiologically resistant to the effects of<br />

lung acidosis associated with breathing high levels of CO2 (neurogenic inflammation<br />

and pulmonary edema). Here we report that naked mole-rats do not respond to<br />

capsaicin applied to the nasal cavity while the same concentration of capsaicin<br />

induces vigorous nose wiping in mice. In an avoidance test, we found that the naked<br />

mole-rats do not avoid fumes from ammonia or acetic acid. They do, however, avoid<br />

fumes from nicotine which acts on trigeminal Aδ fibers, not C fibers. Laboratory rats<br />

avoided all three irritants. To test the effects of high levels of CO2 on the lungs, we<br />

exposed naked mole-rats and mice to a variety of CO2 concentrations while holding<br />

O2 constant. Mice showed significant pulmonary edema at concentrations of 15%<br />

CO2 and higher. The mole-rats showed no edema even at 50% CO2. These results<br />

support the idea that the anomalies found in naked mole-rat C fibers represent<br />

adaptations for surviving in chronically high levels of CO2 and ammonia. Experiments<br />

that exploit the unique features of the naked mole-rat, like those presented here, are<br />

helping us better understand basic trigeminal and pulmonary physiology as well as<br />

adaptations to challenging environments.<br />

29 Speaker Abstracts


T19 Tactile Experience Shapes Behavior in Etruscan Shrews<br />

Michael Brecht, Farzana Anjum<br />

Bernstein Center for Computational Neuroscience, Berlin<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

A crucial role of tactile experience for the maturation of neural response properties in<br />

the somatosensory system is well established, but little is known about the role of<br />

tactile experience in the development of tactile behaviors. Here we study how tactile<br />

experience affects prey capture behavior in Etruscan shrews, Suncus etruscus. Prey<br />

capture in adult shrews is a high-speed behavior that relies on precise attacks guided<br />

by tactile Gestalt cues. We studied the role of tactile experience by three different<br />

approaches. First, we analyzed the hunting skills of young shrews right after weaning.<br />

We found that prey capture in young animals is most but not all aspects similar to that<br />

of adults. Second we performed whisker trimming for three to four weeks after birth.<br />

Such deprivation resulted in a lasting disruption of prey capture even after whisker regrowth:<br />

attacks lacked precise targeting and had a lower success rate. Third, we<br />

presented adult shrews with an entirely novel prey species, the giant cockroach. The<br />

shape of this roach is very different from the shrew’s normal (cricket) prey and the<br />

thorax – which is the preferred point of attack in crickets – is protected a heavy cuticle.<br />

Initially shrews attacked giant roaches the same way they attack crickets and targeted<br />

the thoracic region. With progressive experience, however, shrews adopted a new<br />

attack strategy targeting legs and underside of the roaches and only rarely other body<br />

parts. Speed and efficiency of attacks improved. These data suggest that tactile<br />

experience shapes prey capture behavior.<br />

Speaker Abstracts 30


International Conference „Development of Somatosensation and Pain 2008“<br />

T20 Specification and differentiation of dorsal spinal cord interneurons.<br />

Thomas Müller 1 , Hendrik Wildner 1 , Domenique Bröhl 1 , Mathias Treier 2 ,<br />

Carmen Birchmeier 1<br />

1 Max-Delbrück-Center for Molecular Medicine, Berlin, Germany, 2 EMBL, Heidelberg,<br />

Germany<br />

The dorsal horn of the spinal cord receives sensory information from the periphery,<br />

processes this information and relays it to higher brain centers and to motor neurons<br />

in the ventral spinal cord. Interneurons of the dorsal horn are organized in laminae,<br />

which receive sensory input of characteristic modalities. For instance, nociceptive<br />

sensory fibers synapse in superficial laminae, whereas proprioceptive sensory fibers<br />

project into the deep dorsal horn. Interneurons in the dorsal horn have diverse<br />

physiological characteristics. Their molecular characteristics are only now beginning to<br />

be understood. We have focused on the analysis of molecular mechanisms that are<br />

responsible for the specification of different neuron types of the dorsal horn, and<br />

analyzed a number of transcription factors that control development of dorsal neurons.<br />

Recent data on this work will be discussed. Hori, K., J. Cholewa-Waclaw, et al. (2008).<br />

"A nonclassical bHLH Rbpj transcription factor complex is required for specification of<br />

GABAergic neurons independent of Notch signaling." Genes Dev 22(2): 166-78.<br />

Zechner, D., T. Muller, et al. (2007). "Bmp and Wnt/beta-catenin signals control<br />

expression of the transcription factor Olig3 and the specification of spinal cord<br />

neurons." Dev Biol 303(1): 181-90. Sieber, M. A., R. Storm, et al. (2007). "Lbx1 acts<br />

as a selector gene in the fate determination of somatosensory and viscerosensory<br />

relay neurons in the hindbrain." J Neurosci 27(18): 4902-9. Wildner, H., T. Muller, et<br />

al. (2006). "dILA neurons in the dorsal spinal cord are the product of terminal and nonterminal<br />

asymmetric progenitor cell divisions, and require Mash1 for their<br />

development." Development 133(11): 2105-13. Muller, T., K. Anlag, et al. (2005). "The<br />

bHLH factor Olig3 coordinates the specification of dorsal neurons in the spinal cord."<br />

Genes Dev 19(6): 733-43. Zechner, D., Y. Fujita, et al. (2003). "beta-Catenin signals<br />

regulate cell growth and the balance between progenitor cell expansion and<br />

differentiation in the nervous system." Dev Biol 258(2): 406-18. Muller, T., H.<br />

Brohmann, et al. (2002). "The homeodomain factor lbx1 distinguishes two major<br />

programs of neuronal differentiation in the dorsal spinal cord." Neuron 34(4): 551-62.<br />

31 Speaker Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

T21 The menthol receptor TRPM8 is the principal detector of environmental cold<br />

Jan Siemens 1 , Diana M. Bautista 1 , Josh Glazer 2 , Pamela R. Tsuruda 1 ,<br />

Allan I. Basbaum 3 , Cheryl L. Stucky 2 , Sven-E. Jordt 4 , David Julius 1<br />

1 Departments of Physiology and Cellular & Molecular Pharmacology, University of<br />

California, San Francisco, CA 94158, USA, 2 Department of Cell Biology, Neurobiology<br />

and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA,<br />

3 Departments of Anatomy and Physiology and W.M. Keck Center for Integrative<br />

Neuroscience, University of California, San Francisco, CA 94158, USA, 4 Department<br />

of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA<br />

Sensory nerve fibers can detect changes in temperature over a remarkably wide<br />

range, a process that has been proposed to involve direct activation of<br />

thermosensitive excitatory TRP ion channels. One such channel, TRPM8 or CMR1, is<br />

activated by chemical cooling agents (such as menthol) or when ambient<br />

temperatures drop below ~26°C, suggesting that it mediates the detection of cold<br />

thermal stimuli by primary afferent sensory neurons. However, some studies have<br />

questioned the contribution of TRPM8 to cold detection or proposed that other<br />

excitatory or inhibitory channels are more critical to this sensory modality in vivo. Here<br />

we show that cultured sensory neurons and intact sensory nerve fibers from TRPM8deficient<br />

mice exhibit profoundly diminished responses to cold. These animals also<br />

show clear behavioral deficits in their ability to discriminate between cold and warm<br />

surfaces, or to respond to evaporative cooling. At the same time, TRPM8 mutant mice<br />

are not completely insensitive to cold as they avoid contact with surfaces ≤10°C, albeit<br />

with reduced efficiency. Thus, our findings demonstrate an essential and predominant<br />

role for TRPM8 in thermosensation over a wide range of cold temperatures, validating<br />

the hypothesis that TRP channels are the principal sensors of thermal stimuli in the<br />

peripheral nervous system.<br />

Speaker Abstracts 32


International Conference „Development of Somatosensation and Pain 2008“<br />

T22 PAIN ATTENUATION IN TETHERED-TOXIN TRANSGENIC MICE DUE TO<br />

REDUCED EXCITABILITY OF SENSORY NEURONS<br />

Annika Stuerzebecher 1 , Jing Hu 2 , Gary R. Lewin 2 , Inés Ibañez-Tallon 1<br />

1 Molecular Neurobiology, Max-Delbrück-Centrum, Germany, 2 Molecular Physiology of<br />

Somatic Sensation, Max-Delbrück-Centrum, Germany<br />

Toxins derived from venomous animals have been widely employed in neuroscience<br />

research because of their ability to bind and modulate specific ion channels. However,<br />

the detailed characterization of the structure and function of receptors in neuronal<br />

circuits is often hampered by the fact that the action of soluble neurotoxins cannot be<br />

targeted to specific cells. Our group has developed a new strategy using peptide<br />

toxins that are tethered to the membrane via a GPI anchor and retain their specific<br />

action on ion channel subtypes while acting in a cell-autonomous manner. Since<br />

alterations in the function of voltage gated sodium channels (VGSCs) lead to<br />

hyperexcitability that causes chronic and neuropathic pain, we wanted to address<br />

whether this strategy could be used to specifically manipulate these channels in vivo.<br />

For this purpose, we generated transgenic mice using the bacterial artificial<br />

chromosome (BAC) of the Nav1.8 VGSC to drive expression of the MrVIA conotoxin.<br />

This toxin is a potent blocker of this tetrodoxin resistant (TTX-R) channel, which plays<br />

a major role in pain transduction. Using whole cell patch clamp recording from isolated<br />

sensory neurons a significant reduction of VGSC currents in nociceptive neurons was<br />

observed in these mice while mechanoreceptors where not affected, demonstrating<br />

the cell subtype specificity of this approach. This block was restricted to TTX-R<br />

currents and not compensated by upregulation of TTX-S currents as was observed in<br />

Nav1.8 knockout mice (Akopian et al). The Nav1.8 gene is predominantly expressed<br />

in non-peptidergic nociceptors (IB4+) and consistent with this we observed an<br />

enhanced block in this cell subpopulation. Behavioral studies revealed a remarkable<br />

lower sensitivity to noxious cold stimuli in the transgenic toxin mice. Thus these<br />

studies provide the first proof of function of the BAC transgenic tethered toxin<br />

approach in mammals and its suitability for developing new therapeutic mouse models<br />

for pain research. (Supported by the DFG SFB 665)<br />

33 Speaker Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

T23 Acid-Sensing Ion Channels (ASICs) and pain in the central and peripheral<br />

nervous system<br />

Eric Lingueglia<br />

Institut de Pharmacologie Moléculaire et Cellulaire (IPMC) CNRS/UNSA UMR 6097,<br />

660 Route des Lucioles, 06560 Valbonne-Sophia Antipolis, France.<br />

Acid-sensing ion channels (ASICs) are neuronal voltage-insensitive cationic channels<br />

activated by extracellular acidification, which can trigger membrane depolarization in<br />

response to local acidosis. They are expressed in the central nervous system and in<br />

peripheral sensory neurons, where they have been proposed to sense painful tissue<br />

acidosis that occurs for instance in ischemic, damaged or inflamed tissue. ASICs have<br />

been associated with a number of different sensory processes including nociception,<br />

visual transduction, sour taste perception, hearing functions, and mechanoperception.<br />

In the recent years, we have been interested in the role of these channels in pain<br />

perception and modulation. We have shown that peripheral ASIC expression and<br />

activity are largely increased during inflammation. We have identified several ASICassociated<br />

proteins and regulatory mechanisms. We have characterized ASICs in the<br />

spinal cord and we have developed and used a specific toxin blocker to demonstrate<br />

the implication of the ASIC1a isoform in pain modulation in the central nervous system<br />

through the opioid system. All these data support a role for ASICs in nociception both<br />

at the central and peripheral level. Supported by CNRS, INSERM, AFM, ANR and<br />

ARC/INCa<br />

Speaker Abstracts 34


International Conference „Development of Somatosensation and Pain 2008“<br />

T24 Leukocyte-derived and exogenous opioids acting at peripheral opioid<br />

receptors control neuropathic pain<br />

Dominika Labuz, Anja Schreiter, Yvonne Schmidt, Alexander Brack, Heike Rittner,<br />

Halina Machelska<br />

Anaesthesiologie, Charité - Campus Benjamin Franklin, Berlin, Germany<br />

Background and aims: Neuropathic pain results from nerve injury that can lead to<br />

inflammation. Challenging the current view that immune cells act predominately as<br />

generators of neuropathic pain here we investigate analgesic effects of opioids<br />

derived from leukocytes in response to application of corticotropin-releasing factor<br />

(CRF) in neuritis. We also assess whether opioid delivery relative to the primary site of<br />

nerve injury is critical for efficient peripheral analgesia in neuropathic pain. Methods:<br />

At 14 days after chronic constriction injury (CCI) we examined the expression of opioid<br />

receptors, CRF receptors and opioid peptides by immunohistochemistry, quantified<br />

opioid-containig leukocytes by flow cytometry, and measured nociceptive thresholds<br />

with von Frey test in wild type, beta-endorphin knockout (END KO) and in mice with<br />

severe combined immunodeficiency (SCID). CRF was injected at the CCI site alone or<br />

together with antibodies against opioid peptides, antagonists of CRF- and opioidreceptors,<br />

or after intraperitoneally injected antibody against intercellular adhesion<br />

molecule-1 (ICAM-1). Mu-, delta- and kappa-opioid receptor selective agonists<br />

(DAMGO, DPDPE and U50,488H, respectively) were injected at the CCI site or into<br />

the paw innervated by the ligated nerve. Results: Opioid receptors and opioidcontaining<br />

leukocytes co-expressing CRF receptors accumulated at the site of nerve<br />

injury. Wild type, END KO and SCID mice developed mechanical allodynia in paws<br />

supplied by the ligated nerve. In wild type mice, but not in END KO mice, CRF injected<br />

at the CCI site produced local antinociception that was reversed by antibodies against<br />

beta-endorphin, met-enkephalin and dynorphin, by CRF receptor antagonist, by<br />

selective mu-, delta- and kappa-opioid receptor antagonists, by a peripheral opioid<br />

receptor antagonist, and by anti-ICAM-1-mediated attenuation of opioid-containing<br />

leukocyte accumulation at the injured nerve. In SCID mice, which lack T lymphocytes,<br />

CRF antinociception was significantly decreased as compared with wild type animals.<br />

Mu-, delta- and kappa-receptor selective agonists injected either at the CCI site or into<br />

the paw innervated by the injured nerve dose-dependently attenuated mechanical<br />

allodynia. However, the antinociceptive potency and efficacy of opioids was greater<br />

when they were injected at the site of nerve injury than into the paw innervated by the<br />

ligated nerve. Conclusions: Activation of opioid peptide-containing leukocytes that<br />

accumulate at the site of nerve injury is critical for alleviation of neuropathic pain.<br />

Unspecific immunosuppression may limit opioid-mediated beneficial effects of<br />

neuroinflammation. Further, peripheral opioid antinociception in neuropathic pain is<br />

more prominent when opioid receptors are activated at the site of inflammatory<br />

reaction associated with nerve injury. Supported by DFG/KFO 100/2.<br />

35 Speaker Abstracts


T25 Synaptic Dis-Inhibition in Pathological Pain States<br />

Hanns Ulrich Zeilhofer<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Institute of Pharmacology and Toxicology, University of Zurich, and Institute of<br />

Pharmaceutical Sciences, ETH Zurich<br />

Inflammatory diseases and neuropathic insults trigger signaling cascades, which<br />

frequently lead to intense and long-lasting pain syndromes in affected patients. Such<br />

pain syndromes are characterized not only by an increased sensitivity to painful stimuli<br />

(hyperalgesia), but also by a qualitative change in the sensory perception of other,<br />

tactile stimuli (allodynia) and the occurrence of spontaneous pain in the absence of<br />

any sensory input. Long-term potentiation (LTP)-like changes in synaptic transmission<br />

between nociceptive C-fibers and spino-periaqueductal grey projection neurons as<br />

well as a loss of inhibitory control by GABAergic and glycinergic spinal dorsal horn<br />

neurons have repeatedly been proposed as underlying principles. While considerable<br />

evidence supports a significant contribution of C-fiber LTP to hyperalgesia, such<br />

monosynaptic plasticity can hardly explain the occurrence of allodynia and<br />

spontaneous pain. Here, I will focus on mechanisms of synaptic dis-inhibition in<br />

inflammatory and neuropathic pain and show that pathologically heightened pain<br />

sensitivity can be reversed by restoring synaptic inhibition with drugs that target<br />

specific spinal GABA-A receptor subtypes.<br />

Speaker Abstracts 36


International Conference „Development of Somatosensation and Pain 2008“<br />

T26 Mechanosensitive ion channels, stomatin-like proteins and molecular<br />

tethers essential for touch.<br />

Gary Lewin<br />

Department of Neuroscience, Max-Delbrück Center for Molecular Medicine<br />

The skin is our largest sensory surface, which we use to contact the outside world<br />

through our sense of touch. The molecular mechanisms of mechanoreception in the<br />

skin are poorly understood. Molecules have been identified in flies and worms that are<br />

necessary for mechanosensation, but similar molecules have yet to be identified in<br />

mammals. We have characterized the function of stomatin-like protein 3 (SLP3) a<br />

protein that is expressed in all dorsal root ganglion neurons. The SLP3 amino acid<br />

sequence is highly homologous to the original member of this family stomatin and to<br />

the C.elegans protein MEC-2. Genetic and electrophysiological evidence has<br />

demonstrated that MEC-2 is essential for the function of mechanotransduction<br />

channels in the touch receptor neurons in the worm. It has also been postulated that<br />

MEC-2 forms an integral and essential part of the worm mechanotransduction channel<br />

complex, which also includes two members of Deg/ENaC family Mec-4, and MEC-10.<br />

Using SLP3 mutant mice, we find that SLP3 is required for the normal function of a<br />

large number of skin mechanoreceptors. Thus in SLP3-/- mice around 35% of the<br />

myelinated sensory afferents in the skin show no mechanosensitivity. We also used<br />

whole cell patch clamp techniques to directly measure mechanosensitive currents that<br />

can be activated by sub-micron displacement of the neurites of sensory neurons<br />

acutely isolated in culture. We found that again in around 30% of the cells recorded<br />

from SLP3-/- mice no mechanosensitive current could be found. By reintroducing the<br />

SLP3 gene into SLP3-/- mutant sensory neurons we could rescue the function of ion<br />

channels that underlie the mechanosensitive current. We also found that when SLP3<br />

is expressed heterologously in cells together with ASIC channels the gating of ASIC<br />

by protons was inhibited. Thus SLP3 can interact with and modulate the gating of<br />

ASIC channels. We have recently found that the mechanosenosesnitive currents that<br />

can be recorded in isolated sensory neurons are highly dependent on the extracellular<br />

matrix. I will provide evidence that a molecular tether exists that links<br />

mechanosensitive channels to the extracellular matrix. The molecular nature of this<br />

tether is at this point still unclear. In summary mammalian somatic sensory neurons<br />

probably possess a range of mechanotransduction complexes of varying composition<br />

and the nature of the proteins involved is at the present time still only poorly<br />

understood. Supported by the DFG.<br />

37 Speaker Abstracts


T27 Sensory Neuron Mechanotransduction<br />

John Wood, Francois Rugiero, Ramin Raouf<br />

UCL London<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Light tough, a sense of muscle position, and the responses to tissue-damaging levels<br />

of pressure all involve mechanosensitive sensory neurons that originate in the dorsal<br />

root or trigeminal ganglia. A variety of mechanisms of mechanotransduction have<br />

been proposed. These range from direct activation of mechanically-activated channels<br />

at the tips of sensory neurons to indirect effects of intracellular mediators, or chemical<br />

signals released from distended tissues or specialized mechanosensory end organs<br />

We have used mammalian sensory neurons in culture to characterise mechanicallygated<br />

currents, in an attempt to identify their molecular structure. We will present data<br />

on potential binding targets of the conopeptide NMB-1 (noxious mechanoblocker-1),<br />

which blocks slowly adapting mechanosensitive currents in DRG neurons, as well as<br />

the perception of pressure-evoked pain.<br />

Speaker Abstracts 38


International Conference „Development of Somatosensation and Pain 2008“<br />

T28 Breathing with Phox2b<br />

Jean-François Brunet<br />

Ecole normale supérieure, Paris, France.<br />

Visceral sensation —as opposed to somatosensation, the main focus of this<br />

meeting— conveys to the brain inputs that are mostly unconscious and directly<br />

relevant to homeostasis: arterial pressure, gaz contents of the blood, chemical<br />

composition of the alimentary bolus or the presence of toxins in the gut or<br />

bloodstream. We have shown that most of the neurons involved, in the central and<br />

peripheral nervous systems, develop under the control of the homeodomain<br />

transcription factor Phox2b. The anatomical and cellular locus for one of the key<br />

visceral sensory modalities —the detection of elevated CO2 in the blood or<br />

hypercapnia— has remained contentious for many years. I will present our<br />

contribution to the recent evidence that this essential component of the “drive to<br />

breathe” is insured by a group of neurons in the hindbrain, also depending on Phox2b.<br />

39 Speaker Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

T29 Roles of Runx1 in controlling nociceptor development and pain behaviors<br />

Qiufu Ma<br />

Department of Neurobiology, Harvard Medical School and Department of Cancer<br />

Biology, Dana-Farber Cancer Institute<br />

In mammals, the perception of pain is initiated by the stimulation of a heterogeneous<br />

population of nociceptive sensory neurons, or nociceptors. However, how individual<br />

subgroups of nociceptors control pain perception is still largely unclear. Here we<br />

present data suggesting that nociceptors undergoing transient Runx1 expression<br />

might play a critical role for thermal pain and neuropathic pain. Runx1 is a runt domain<br />

transcription factor that is initially expressed in most, if not all, embryonic nociceptors,<br />

but its expression is progressively extinguished in about 50% of nociceptors, most of<br />

which are peptidergic neurons. Genetic studies show that Runx1 is required for the<br />

expression of many ion channels/receptors and neurotrophin receptors, including TRP<br />

class thermal receptors, Na+-gated and ATP-gated channels, Mrgpr class G-protein<br />

coupled receptors, and the Ret receptor tyrosine kinase. Behavioral studies<br />

demonstrated that mice lacking Runx1 exhibit severe deficits in thermal and<br />

neuropathic pain. In this presentation, I will show that Runx1-dependent genes are<br />

divided into two categories, A and B. Category A genes are expressed in nociceptors<br />

with persistent Runx1 expression, including TRPM8, TRPA1 (weak Runx1<br />

expression), TRPC3, MrgprD, SNS2, P2X3, Ret and others. Category B genes are<br />

expressed in nociceptors with transient Runx1 expression, including TRPV1 (high<br />

level), MrgprA/B/C, and others. A series of genetic manipulations then suggest that<br />

category B, but not category A, Runx1 targets may be required for<br />

thermal/neuropathic pain, implying that nociceptors with transient Runx1 expression<br />

might play a prominent role for these types of pain.<br />

Speaker Abstracts 40


International Conference „Development of Somatosensation and Pain 2008“<br />

T30 Peripheral mechanisms for cold detection in intact and injured tissues<br />

Carlos Belmonte<br />

Instituto de Neurociencias de Alicante Universidad Miguel Hernández-Consejo<br />

Superior de Investigaciones Científicas, Campus de San Juan, Apdo. correos 18,<br />

03550 Sant Joan d'Alacant, Alicante (Spain)<br />

Cold-sensitive nerve terminals of primary sensory neurons detect changes in external<br />

temperature values within an ample range. Moderate temperature reductions evoke<br />

cooling sensations whereas higher low temperature values elicit sensations of<br />

unpleasant cold or pain. Cold-sensitive neurons exhibit specific active and passive<br />

membrane properties and are equipped with different ionic conductances. TRPM8 and<br />

background K+ currents, TRPA1, IKD and Ih are variably expressed in the soma and<br />

peripheral endings of primary sensory neurons and determine the final sensitivity to<br />

cold and the firing characteristics of specific and non-specific cold-sensitive sensory<br />

endings. Peripheral axotomy of cold receptor fibers innervating the cornea of the eye<br />

alters their sensitivity and firing response to cold, suggesting that damage to<br />

peripheral cold-sensitive nerve terminals modifies the peripheral sensory message<br />

evoked by cold, thus contributing to cold dysesthesias.<br />

41 Speaker Abstracts


T31 Genetic Determinants of Pain<br />

Stephen McMahon<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Wolfson Centre for Age-Related Diseases, King’s College London, UK<br />

Pain sensitivity is known to vary considerably between people. This is true both for<br />

experimental pain, for instance where a standard force is applied to normal healthy<br />

tissue, but also for disease-related such as osteoarthritis, where it has long been<br />

recognised up to 50% of individuals with radiographic indications of disease do not<br />

report pain, and there is a poor correlation between pain and degree of radiographic<br />

change. The influence of heritable (genetic) factors on pain sensitivity will be reviewed<br />

in this talk. Mutations in a few individual genes have recently been demonstrated to<br />

have a dramatic effect on pain appreciation. The best known example is mutations in<br />

the trkA gene, a number of which lead to the development of congenital insensitivity to<br />

pain with anhydrosis (also know as hereditary sensory neuropathy type IV). These<br />

mutations lead to a failure of small diameter nociceptive neurones to develop. More<br />

recently mutations in a sodium channel gene normally expressed selectively in<br />

peripheral pain sensitive neurones (so-called Nav 1.7) have also been found to lead to<br />

both loss of function (congenital analgesia) and gain of function (erythromyalgia)<br />

phenotypes. We have recently undertaken a classical twin study to evaluate the<br />

relative contributions of genetic and environmental factors on responses to painful<br />

stimuli in human volunteers to a wide variety of pain traits. Statistically significant<br />

genetic components (varying between 22-55%) were seen for the responses to the<br />

majority of painful stimuli including sensitivity to heating the skin, areas of secondary<br />

hyperalgesia brush evoked allodynia following a burn injury and iontophoresis of acid<br />

solutions. Our study demonstrates the importance of genetic factors in determining<br />

human experimental pain sensitivity, and opens the way for its use as a phenotype in<br />

gene discovery. Since experimental pain sensitivity is known to be a predictor for<br />

pathological pain, our data imply that genetic factors have an important aetiological<br />

contribution towards clinical pain states.<br />

Speaker Abstracts 42


International Conference „Development of Somatosensation and Pain 2008“<br />

43 List of Posters<br />

List of Posters


May 14 – May 17, 2008 – Berlin, Germany<br />

P1 DEVELOPMENT OF GENETIC TOOLS FOR SELECTIVE AND REVERSIBLE<br />

SILENCING OF ION CHANNELS USING NATURAL TOXINS<br />

Sebastian Auer, Rene Jüttner, Ines Ibanez-Tallon<br />

P2 Consequences of mPGES-1 deletion on spinal cytokine synthesis and COX-<br />

2 expression<br />

Christian Brenneis, Ovidiu Coste, Carlo Angioni, Gerd Geisslinger, Klaus<br />

Scholich<br />

P3 Neurod genes are essential for neuropeptide expression and diversification<br />

of GABAergic neurons in the dorsal spinal cord<br />

Dominique Bröhl, Michael Strehle, Hagen Wende, Kei Hori, Ingo Bormuth,<br />

Klaus-Armin Nave, Thomas Müller, Carmen Birchmeier<br />

P4 Auxiliary subunits and the biophysics of single C. elegans force<br />

transduction channels<br />

Austin Brown, Zhiwen Liao, Miriam Goodman<br />

P5 THE CONTRIBUTION OF TRPA1 AND TRPM8 TO COLD ALLODYNIA AND<br />

NEUROPATHIC PAIN<br />

Ombretta Caspani, Sandra Zurborg , Paul Heppenstall<br />

P6 Relevance of kinin receptors in painful processes: study of B1/B2 knockout<br />

mice<br />

Cecile Cayla, Michael Bader, Dominika Labuz, Michael Schäfer, Christoph Stein<br />

P7 Role of Extracellular Matrix in Sensory Mechanotransduction<br />

Li-Yang Chiang, Jing Hu, Bettina Erdmann, Manuel Koch, Gary R Lewin<br />

P8 The transcription factor Rbp-J is required for specification of GABAergic<br />

neurons in dorsal spinal cord.<br />

Justyna Cholewa-Waclaw, Hendrik Wildner, Benedetta Martarelli, Carmen<br />

Birchmeier<br />

P9 ANTAGONISTIC FUNCTIONAL INTERACTION BETWEEN TRPM8 AND Kv1<br />

POTASSIUM CHANNELS DETERMINES NOXIOUS COLD DETECTION<br />

Elvira de la Peña, Rodolfo Madrid, Donovan-Rodriguez Tansy, Carlos Belmonte,<br />

Felix Viana<br />

P10 The role of the Tshz1 gene in development of GABA-ergic circuits in the<br />

central nervous system<br />

Alistair Garratt<br />

P11 TRPV4 Biochemically And Functionally Interacts With The Cytoskeleton<br />

Chandan Goswami, Tim Hucho<br />

P12 Stimulation of formyl peptide receptor on PMN mediates peripheral opioid<br />

analgesia<br />

Dagmar Hackel, Dominika Labuz, Alexander Brack, Heike Rittner<br />

List of Posters 44


International Conference „Development of Somatosensation and Pain 2008“<br />

P13 Experimental skin inflammation is impaired by axotomy but promotes the<br />

regeneration of skin nerves<br />

Sven Hendrix, Björn Picker, Frank Siebenhaar, Marcus Maurer, Eva M. Peters<br />

P14 A tether link required for touch<br />

Jing Hu, Li-Yang Chiang, Bettina Erdmann, Gary Lewin<br />

P15 Molecular interactions between stomatin-like proteins and acid-sensing ion<br />

channels<br />

Julia Jira, Paul Heppenstall<br />

P16 The zinc finger transcription factor Bcl11a/Ctip1 is essential for neuronal<br />

differentiation and sensory circuit formation in dorsal spinal cord<br />

development<br />

Anita John, Heike Brylka, Pentao Liu, René Jüttner, E. Bryan Crenshaw III,<br />

Nancy A. Jenkins, Neal G. Copeland, Carmen Birchmeier, Stefan Britsch<br />

P17 Sequential maturation of sensory neuron mechanotransduction during<br />

embryonic development<br />

Stefan G. Lechner, Rui Wang, Henning Frenzel, Gary R. Lewin<br />

P18 Characterization of the peripheral Osmoreceptor<br />

Soeren Markworth, Silke Frahm, Ines Ibanez-Tallon, Jens Jordan, Gary R. Lewin<br />

P19 Analysis of Opioid receptor/K+ channel coupling in sensory neurons<br />

D. Nockemann, C. Stein P. A. Heppenstall<br />

P20 Direct inhibition of nociceptors by mu-opioid receptor agonist in<br />

neuropathic pain<br />

Yvonne Schmidt, Paul A. Heppenstall, Shaaban A. Mousa, Halina Machelska<br />

P21 Prevention of opioid peptide degradation for pain control in peripheral<br />

inflamed tissue<br />

Anja Schreiter, Carmen Gore, Shaaban A. Mousa, Bernard P. Roques,<br />

Christoph Stein, Halina Machelska<br />

P22 TRPC5 channel is activated by osmotically induced membrane stretch<br />

Sergio Soriano, Carlos Belmonte, Félix Viana, Ana Gomis<br />

P23 Opioid withdrawal increases TRPV1 activity in a PKA dependent manner<br />

Viola Spahn, Christian Zoellner<br />

P24 A molecular dissection of TRPV1 sensitisation using the naked mole-rat<br />

Ewan St. John Smith, Gireesh Anirudhan, Gary Lewin<br />

P25 Analysis of phosphorylation targets of cGMP-dependent kinase I involved<br />

in sensory axon bifurcation<br />

Agne Stonkute, Hannes Schmidt, Fritz G. Rathjen<br />

45 List of Posters


May 14 – May 17, 2008 – Berlin, Germany<br />

P26 New In Vitro mechano-assay reveals distinct intrinsic mechanosensitive<br />

subtypes in cultured sensory neurons<br />

Dmitry Usoskin, Per Uhlen, Patrik Ernfors<br />

P27 Function of Cav3.2 in Sensory neuron mechanosensitivity<br />

Rui Wang, Gary R. Lewin<br />

List of Posters 46


International Conference „Development of Somatosensation and Pain 2008“<br />

47 Poster Abstracts<br />

Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P1 DEVELOPMENT OF GENETIC TOOLS FOR SELECTIVE AND REVERSIBLE<br />

SILENCING OF ION CHANNELS USING NATURAL TOXINS<br />

Sebastian Auer, Rene Jüttner, Ines Ibanez-Tallon<br />

Max-Delbrueck-Center for Molecular Medicine, Cellular Neuroscience, Berlin,<br />

Germany,<br />

Toxins derived from venomous animals have been widely employed in neuroscience<br />

research because of their ability to manipulate specific ion channels. However, the<br />

detailed characterisation of the structure and function of receptors in neuronal circuits<br />

is often hampered by the fact that the action of soluble neurotoxins cannot be targeted<br />

to specific cells. Thus we constructed recombinant toxins for the cell-autonomous<br />

manipulation of neuronal receptors by tethering them to the cell membrane via the GPI<br />

anchor of the endogenous prototoxin lynx1. In former studies we showed that<br />

recombinant conotoxins as well as bungarotoxins were not dispersed in solution and<br />

retained their high specificity for target ion channels, indicating that this approach can<br />

be used to restrict the site of neurotoxin action to genetically targeted cells. Here, we<br />

extended our studies to various other toxins derived from snails, spiders, scorpions<br />

and sea anemones by characterising their action on voltage- and ligand-gated ion<br />

channels. Furthermore we optimized the cell membrane attachment of the toxins by<br />

exchanging the GPI anchor with a transmembrane domain fused to the green<br />

fluorescent marker gene GFP, thus allowing the easy monitoring of the recombinant<br />

molecules. In a next step we set up both a constitutive as well as an inducible lentiviral<br />

vector expression system for the efficient in vitro and in vivo transduction of neuronal<br />

cells with the optimized constructs, allowing the detailed structure-function analysis of<br />

the ion channels of interest. To sum up, these novel tethered toxins are now optimized<br />

for in vivo use, thus offering new possibilities for investigations regarding the<br />

physiology of neuronal circuits.<br />

Poster Abstracts 48


International Conference „Development of Somatosensation and Pain 2008“<br />

P2 Consequences of mPGES-1 deletion on spinal cytokine synthesis and COX-2<br />

expression<br />

Christian Brenneis, Ovidiu Coste, Carlo Angioni, Gerd Geisslinger, Klaus Scholich<br />

Institute of Clinical Pharmacology, pharmazentrum frankfurt, ZAFES, Klinikum der<br />

Johann Wolfgang Goethe-Universität, Theodor-Stern-Kai 7, Frankfurt, Germany<br />

Cyclooxygenase-2 (COX-2)-dependent prostaglandin (PG) E2 synthesis in the spinal<br />

cord plays a major role in the development of inflammatory hyperalgesia and<br />

allodynia. In contrast to its pro-nociceptive effects, PGE2 also limits its own synthesis<br />

by inhibiting the release of pro-inflammatory cytokines from microglia. Our previous<br />

work showed that microsomal PGE2 synthase-1 (mPGES-1) -deletion shifts spinal<br />

COX-2-mediated prostaglandin synthesis from PGE2 to other prostaglandins. Using<br />

primary spinal cord cultures we demonstrate here that only PGE2 and not PGD2,<br />

PGF2alpha or the stable prostacyclin-receptor-agonist cicaprost inhibits LPS induced<br />

tumour necrosis factor alpha (TNFalpha) synthesis, COX-2 and mPGES-1 expression<br />

and COX-2 activity. This PGE2 effect can be mimicked by EP-2- and EP-4-receptor<br />

agonists. A multi epitope ligand cartographie (MELC) showed strong EP-2-receptorexpression<br />

on spinal microglia. Next we determined in an in vivo model whether a<br />

deletion of mPGES-1 can increase LPS stimulated spinal cytokine synthesis and<br />

COX-2 expression. The intrathecal injection of LPS resulted in a time-dependent<br />

increase of TNFalpha concentrations as well as COX-2 expression and activity in<br />

lumbal spinal cords. In this in vivo model mPGES-1-/- mice exhibited a 80% reduced<br />

spinal PGE2 synthesis. However, most surprisingly, mPGES-1-/- mice had no<br />

differences in spinal TNFalpha concentration as well as COX-2-expression and -<br />

activity as compared to their littermates. The results suggest that in spinal cells<br />

exogenously applied PGE2 is a potent inhibitor of cytokine synthesis, while in vivo<br />

mPGES-1 derived PGE2 does not have this effect.<br />

49 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P3 Neurod genes are essential for neuropeptide expression and diversification<br />

of GABAergic neurons in the dorsal spinal cord<br />

Dominique Bröhl 1 , Michael Strehle 1 , Hagen Wende 1 , Kei Hori 2 , Ingo Bormuth 3 ,<br />

Klaus-Armin Nave 3 , Thomas Müller 1 , Carmen Birchmeier 1<br />

1 Max-Delbrück-Centrum for Molecular Medicine, 2 University of Texas Southwestern<br />

Medical Center, 3 Max Planck Institute of Experimental Medicine<br />

Neurons in the dorsal horn of the spinal cord receive somatosensory information from<br />

the periphery, integrate the information and transmit it to higher brain centers. These<br />

neurons express many different neuropeptides that modulate sensory perception like<br />

the sensation of pain. Inhibitory neurons of the dorsal horn derive from postmitotic<br />

neurons that express Pax2, Lbx1 and Lhx1/5, and diversify during maturation. In<br />

particular, fractions of maturing inhibitory neurons express various neuropeptides. We<br />

show here that a co-ordinate molecular mechanism determines inhibitory and<br />

peptidergic fate in the developing dorsal horn. A bHLH factor complex that contains<br />

Ptf1a acts as upstream regulator and initiates the expression of several downstream<br />

transcription factors in the future inhibitory neurons, of which Pax2 is known to<br />

determine the neurotransmitter phenotype. We demonstrate that dynorphin, galanin,<br />

NPY, nociceptin and enkephalin expression depends of Ptf1a, indicating that these<br />

neuropeptides are expressed in inhibitory neurons. Furthermore, we show that<br />

Neurod1/2/6 and Lhx1/5, which act downstream of Ptf1a, control distinct aspects of<br />

petidergic differentiation. In particular, the Neurod1/2/6 factors are essential for<br />

dynorphin and galanin expression, whereas the Lhx1/5 factors are essential for NPY<br />

expression. We conclude that a transcriptional network operates in maturing dorsal<br />

horn neurons that co-ordinately determines transmitter and peptidergic fate.<br />

Poster Abstracts 50


International Conference „Development of Somatosensation and Pain 2008“<br />

P4 Auxiliary subunits and the biophysics of single C. elegans force transduction<br />

channels<br />

Austin Brown, Zhiwen Liao, Miriam Goodman<br />

Dept. of Molecular and Cellular Physiology, Stanford University<br />

The stomatin-related protein MEC-2 and the paraoxonase-related protein MEC-6 are<br />

members of the C. elegans touch receptor neuron MEC 4/MEC 10<br />

mechanotransduction ion channel complex. Coexpression of these auxiliary subunits<br />

with MEC-4 and MEC-10 in Xenopus oocytes leads to a dramatic and synergistic<br />

increase in whole-cell current. Using single-channel recordings in outside-out patches,<br />

we demonstrate that this increase cannot be explained by an increase in open<br />

probability of active channels or single-channel conductance. We also show that<br />

coexpression of MEC-2 or MEC-6 does not increase in the amount of MEC-4 protein<br />

in the plasma membrane. We conclude that in the absence of MEC-2 and MEC-6 the<br />

vast majority of channels occupy a non-conducting state despite correct expression at<br />

the plasma membrane. We also are investigating the role of cholesterol in the ion<br />

channel complex, since MEC 2 exhibits cholesterol-binding activity required for<br />

behavioral response to touch in vivo.<br />

51 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P5 THE CONTRIBUTION OF TRPA1 AND TRPM8 TO COLD ALLODYNIA AND<br />

NEUROPATHIC PAIN<br />

Ombretta Caspani, Sandra Zurborg, Paul Heppenstall<br />

Clinic for Anesthesiology and Operative Intensive Care, Charté Universitätsmedizin<br />

Berlin, Germany<br />

Cold allodynia is a common symptom of neuropathic pain. However the underlying<br />

mechanisms of this enhanced sensitivity to cold are not known. TRPM8 and TRPA1<br />

are non-selective cation channels expressed by sensory neurons that have been<br />

proposed as candidates for cold transducers. We have investigated the role of these<br />

ion channels in cold allodynia by examining their expression and function following<br />

nerve injury. We used a chronic constriction injury of the sciatic nerve to model<br />

neuropathic pain in mice. We dissected lumbar dorsal root ganglia (DRG) at 7 and 14<br />

days post surgery and used quantitative RT-PCR, in situ hybridization,<br />

immunohistochemistry and calcium microfluorimetry to examine the expression and<br />

function of TRPM8 and TPRA1 after nerve injury. With the expression analysis we<br />

detected decrease in the percentages of the both subpopulations of neurons, TRPA1<br />

positive and TRPM8 positive, after nerve injury. We then examined functional<br />

properties of cold transduction using calcium imaging. Our experiments revealed a<br />

down-regulation of TRPA1 in the DRG after nerve injury, although no change in the<br />

number of cold responsive neurons.<br />

Poster Abstracts 52


International Conference „Development of Somatosensation and Pain 2008“<br />

P6 Relevance of kinin receptors in painful processes: study of B1/B2 knockout<br />

mice<br />

Cecile Cayla 1 , Michael Bader 2 , Dominika Labuz 1 , Michael Schäfer 1 , Christoph Stein 1<br />

1 Anaesthesiologie, Charite, Campus Benjanin Franklin, Berlin, 2 Max-Delbrück-Center,<br />

Berlin, Germany<br />

Kinins are peptides rapidly produced during inflammation that play a critical role in the<br />

initiation of pain and the development of hypersensitivity. Kinins act through two<br />

distinct G-protein-coupled receptors, constitutively expressed B2 and injury-induced<br />

B1. The B2 receptor has been preferentially associated with acute and the B1 receptor<br />

with chronic phases of inflammation and pain. To progress in our understanding of the<br />

function of the kinin system, we aimed to generate mice lacking both B1 and B2<br />

receptors (B1B2-/-) and to study their nociceptive behaviour. Because of the close<br />

chromosomal proximity of the B1 and B2 receptor genes, this model could not be<br />

obtained by simple breeding of single-defective lines. By homologous recombination<br />

of the B1 receptor gene directly in embryonic stem cells isolated from B2-deficient<br />

animals we generated the mouse model. B1B2-/- mice are viable, morphologically<br />

normal and fertile. The deletion of the two genes was verified by Southern blot and the<br />

absence of receptor mRNAs evidenced in all organs tested. In contrast to wild-type<br />

(WT) mice, an intraplantar injection of bradykinin did not induce nociceptive responses<br />

in B1B2-/- mice. We next evaluated the response to intraperitoneal administration of<br />

0.6% acetic acid, which elicited a significant number of abdominal constrictions in WT,<br />

but a reduced response in the double knockout line. Mechanical and thermal<br />

sensitivities in healthy B1B2-/- animals were also tested as well as the development of<br />

hyperalgesia induced by inflammation or nerve injury.<br />

53 Poster Abstracts


P7 Role of Extracellular Matrix in Sensory Mechanotransduction<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Li-Yang Chiang 1 , Jing Hu 1 , Bettina Erdmann 1 , Manuel Koch 2 , Gary R Lewin 1<br />

1 Molecular Physiology of Somatic Sensation Group, Max Delbrück Center for<br />

Molecular Medicine, Berlin, 2 Center for Biochemistry and Department of Dermatology,<br />

Medical Faculty, University of Cologne<br />

Genetic screens carried out in both Caenorhabditis elegans (C. elegans) and<br />

Drosophila Melanogaster have suggested that extracellular proteins might be essential<br />

for the transducton of body touch and fly bristle movement respectively (Chung et al.<br />

2001. Neuron 29: 415-28; Du et al. 1996. Neuron 16: 183-94; Ernstrom & Chalfie.<br />

2002. Annu Rev Genet 36: 411-53). There is no direct evidence that extracellular<br />

factors are crucial for the gating of somatic mechanotransduction channels in<br />

vertebrates. In order to investigate this, we have established co-cultures of sensory<br />

neurons and skin-derived keratinocytes or fibroblasts. We have asked whether<br />

different extracellular environments modify the transduction of mechanical stimuli by<br />

DRG neurons. In this project, we have found the molecular nature of the extracellular<br />

environment can profoundly modulate mechanosensitive channels in DRG neurons.<br />

Matrix derived from keratinocytes has profoundly inhibitory effect on mechanosensitive<br />

RA currents. It has been shown Laminin5 is exclusively expressed in Keratinocytes.<br />

We found Laminin5 containing matrix can reproduce the inhibitory properties of<br />

keratinocytes matrix on DRG mechanosensitivity. Future experiments with laminin5<br />

deficient matrix will allow us to determine if Lamnin5 itself is sufficient for the inhibitory<br />

effects of keratinocyte matrix.<br />

Poster Abstracts 54


International Conference „Development of Somatosensation and Pain 2008“<br />

P8 The transcription factor Rbp-J is required for specification of GABAergic<br />

neurons in dorsal spinal cord.<br />

Justyna Cholewa-Waclaw, Hendrik Wildner, Benedetta Martarelli, Carmen Birchmeier<br />

Max-Delbruck-Centrum for Molecular Medicine, 13125 Berlin-Buch, Germany<br />

dILA and dILB neurons, the major neuronal subtypes of the dorsal spinal cord, arise in<br />

a salt-and-pepper pattern from a broad progenitor domain that expresses Mash1 and<br />

p48/Ptf1a transcription factors. dILA and dILB differentiate into inhibitory and<br />

excitatory neurons. Specification of dILA neurons depends on the bHLH transcription<br />

factor p48/Ptf1a. Furthermore, dILA neurons are produced by asymmetric progenitor<br />

cell division. To address the role of Notch signaling in the specification of dILA<br />

neurons, we employed two genetic approaches in mice: (i) expression of a dominantnegative<br />

variant of Mastermind-like1 (DNMAML) in an inducible manner; Mastermindlike1<br />

is a transcriptional co-activator that is thought to be essential to activate the<br />

transcription of Notch-dependent target genes of Rbp-J; (ii) conditional mutagenesis of<br />

Rbp-J, the major transcriptional mediator of Notch signals. Induction of expression of<br />

the dominant-negative Mastermind-like1 variant and the introduction of the Rbp-J<br />

mutation were regulated via cre-recombinase (Pax3Cr or Pax7CreERT2 alleles).<br />

Compared to control mice, we observed in Pax3Cre/DNMAML mutants a partial<br />

depletion of the progenitor domain in the dorsal spinal cord at E12.5, consistent with a<br />

reduction of Notch signaling. However, there was no significant alteration in the<br />

proportions of dILA and dILB neurons generated. In contrast, we found a significant<br />

change in the proportions of dILA and dILB neurons in Pax7CreERT2 Rbp-Jflox/flox<br />

mice. In the pancreas, Rbp-L is known to form a transcription factor complex together<br />

with p48/Ptf1a. Our results indicate that Rbp-J can function independently of Notch as<br />

a component of Ptf1a complex in determining the identity of GABAergic neurons.<br />

Supporting results were obtained from analysis of a mutant of p48/Ptf1a<br />

(Ptf1aW298A) that encodes a variant with a single change in the amino acid<br />

sequence. This mutation precludes an interaction between Ptf1aW298A and Rbp-L or<br />

Rbp-J. Ptf1aW298A is a phenocopy of the Ptf1anull mutant, and inhibitory dILa<br />

neurons in the dorsal spinal cord are not formed in either of the two mutant mouse<br />

strains.<br />

55 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P9 ANTAGONISTIC FUNCTIONAL INTERACTION BETWEEN TRPM8 AND Kv1<br />

POTASSIUM CHANNELS DETERMINES NOXIOUS COLD DETECTION<br />

Elvira de la Peña, Rodolfo Madrid, Donovan-Rodriguez Tansy, Carlos Belmonte,<br />

Felix Viana<br />

Instituto de Neurociencias. Universidad Miguel Hernández-CSIC. Campus de San<br />

Juan. Alicante-España.<br />

Detection of temperature decreases in the innocuous range is carried out by a specific<br />

functional class of peripheral sensory nerve endings named cold thermoreceptors.<br />

The somas of cold-sensitive primary sensory neurons cultured in vitro present very<br />

different temperature thresholds (range 34─20ºC) and can be separated into low<br />

threshold (LT) and high threshold (HT) types. TRPM8, a non-selective, calciumpermeable,<br />

cation channel of the transient receptor potential superfamily, that is<br />

activated by cooling and menthol is the principal transduction channel for cold<br />

temperatures in primary sensory neurons. However, additional experimental evidence<br />

has shown that various potassium conductances also exert a strong influence on cold<br />

sensing. We investigated the influence of TRPM8, a cold- and menthol-sensitive TRP<br />

channel, and IKD, a slowly-inactivating, 4-AP-sensitive transient potassium current on<br />

temperature-evoked responses in cold thermoreceptors. We performed calcium<br />

imaging and patch-clamp recordings from neonatal mice cultured trigeminal neurons.<br />

Temperature response threshold was measured on the cold-evoked intracellular<br />

calcium elevation. Expression of TRPM8 was quantified by the amplitude of the<br />

menthol-evoked current (Imenthol). We found that Imenthol was significantly larger in<br />

LT CS neurons while the opposite was true for IKD. Using pharmacological tools we<br />

demonstrate that these two conductances exert opposite influences on temperaturedependent<br />

excitability in trigeminal thermoreceptors. One current, IKD, acts as a brake<br />

while the other, TRPM8, promotes cold-induced activity. Alterations in the balance<br />

between ITRPM8 and IKD may be an important factor in the development of cold<br />

allodynia symptoms, characteristic of many neuropathic conditions. Supported by<br />

funds from the Spanish Ministry of Education and Science, projects SAF2004-01011<br />

to F.V , BFU2005 08741 to C.B and Consolider 2007 CSD2007-0023. Fundación<br />

Marcelino Botín.<br />

Poster Abstracts 56


International Conference „Development of Somatosensation and Pain 2008“<br />

P10 The role of the Tshz1 gene in development of GABA-ergic circuits in the<br />

central nervous system<br />

Alistair Garratt<br />

Department of Neurosciences, Max-Delbrück-Center for Molecular Medicine, Berlin<br />

We first characterized the three Tshz genes as being highly expressed in the<br />

substantia gelatinosa, the first relay station for pain sensation. We chose to analyse<br />

the function of the Tsh1 gene, which we had shown also to be expressed in cells of<br />

the outer granule cell layer of the embryonic olfactory bulb. The early development of<br />

the olfactory bulb is still poorly understood. Our aim was to determine the function of<br />

Tsh1, a potential integrator of signaling downstream of cell surface receptors (e.g.<br />

Wnt/beta-catenin signaling) and a partner for Hox-family transcription factors in<br />

olfactory bulb development. The methods employed were classical mutagenesis in the<br />

mouse, together with immunohistochemistry, in situ hybridisation and microarray<br />

analysis. Tsh1 was expressed in an early emigrating population of GABA-ergic<br />

interneurons, generated in the rostral telencephalon around E11-E12. In Tsh1<br />

homozygous null embryos, mutant cells clumped together and failed to distribute<br />

radially within the olfactory bulb, a phenotype associated with changes in Semaphorin<br />

signaling. In addition, the Tsh1 mutant cells failed to activate expression of GABAergic<br />

markers such as GAD67 and GABA. Conclusions: Tsh1 is essential for the<br />

correct radial migration and differentiation of a pioneer population of GABA-ergic<br />

granule cell neurons within the olfactory bulb. Tsh1 mutant cells failed to express<br />

Semaphorin 3c and a signaling mediator cypin, which may account for the aberrant<br />

clumping of mutant cells within the center of the olfactory bulb.<br />

57 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P11 TRPV4 Biochemically And Functionally Interacts With The Cytoskeleton<br />

Chandan Goswami, Tim Hucho<br />

Max Planck Institute of molecular Genetics<br />

TRPV4 and the cytoskeleton have been reported to influence the development of<br />

mechanical hyperalgesia. If and how these molecules interact is unknown. We now<br />

describe TRPV4 to form a Ca2+-sensitive complex with components of the tubulin-,<br />

actin- and neurofilament-cytoskeleton and and CamKII. The C-terminus of the<br />

nociceptive signaling molecules PKCε TRPV4 is sufficient for complex formation. The<br />

interaction with soluble and filamentous tubulin and actin is direct. Actin and tubulin<br />

bind with high affinity and compete for binding. The presence of TRPV4 strongly<br />

increases the amount of microtubules formed even in presence of microtubule<br />

depolymerising agents. Accordingly, expression of TRPV4 induces striking<br />

morphological changes such as formation of lamellipodia, filopodia, as well as neuritelike<br />

structures. These changes are not restricted to neuron-derived cells but also occur<br />

in transfected HaCat, ChoKI, and HeLa cells. TRPV4 co-localizes with actin and<br />

tubulin both in fixed as well as live cells and stabilizes microtubules at membranous<br />

regions. Activation of TRPV4 induces rapid depolymerization of microtubules. This is<br />

accompanied by elongation of filopodial structures and by a transition of lammelipodial<br />

to filopodial structures. Therefore, cell boundaries and growth cone structures rapidly<br />

retract. Accordingly, rat primary sensory neurons do not extend neurites if cultured in<br />

the presence of low doses of TRPV4 agonists. This phenotype is restricted to a subset<br />

of nociceptive neurons, which binds to isolectin B4.<br />

Poster Abstracts 58


International Conference „Development of Somatosensation and Pain 2008“<br />

P12 Stimulation of formyl peptide receptor on PMN mediates peripheral opioid<br />

analgesia<br />

Dagmar Hackel, Dominika Labuz, Alexander Brack, Heike Rittner<br />

Department of Anesthesiology and Intensive Care, Berlin, Germany<br />

Hyperalgesia can be elicited by tissue destruction e.g. in inflammation. Both proalgesic<br />

(e.g. cytokines, protaglandins, protons) and analgesic (e.g. opioid peptides)<br />

mediators are secreted during the inflammatory process. Opioid peptides bind to<br />

opioid receptors on peripheral nerve endings and thereby elicit potent analgesia.<br />

Polymorphonuclear cells (PMN) are the major source of opioid peptides and are<br />

important for endogenous analgesia especially during early inflammation. PMNs<br />

express the formyl peptide receptor recognizing formylated peptides from bacteria or<br />

mitochondria. The presented work shows that formylated peptides and their receptor<br />

influence release of opioid peptides in vitro and in vivo. Intraplantar injection of fMLP<br />

(N-formyl-MET-LEU-PHE), an agonist of FPR, cause an analgesic effect in rats with<br />

two hours complete Freund's adjuvant (CFA) induced inflammation in the hind paw.<br />

This analgesia abolishes after depletion of PMNs or after additional intraplantar<br />

injection of either the opioid receptor antagonist naloxone or antibodies against opioid<br />

peptides. Moreover PMNs release opioid peptide after fMLP treatment in vitro. The in<br />

vivo and in vitro effects of fMLP are blockable with t-BocPLPLP (tert-butoxycarbonyl-<br />

Phe-Leu-Phe-Leu-Phe) or Cyclosporin H which are competitive antagonists of FPR.<br />

These results reveal that the FPR on PMNs is important for the release of opioid<br />

peptides and endogenous analgesia during the inflammatory processes. This work<br />

was supported by the DFG (German Research Foundation).<br />

59 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P13 Experimental skin inflammation is impaired by axotomy but promotes the<br />

regeneration of skin nerves<br />

Sven Hendrix 1 , Björn Picker 1 , Frank Siebenhaar 2 , Marcus Maurer 2 , Eva M. Peters 3<br />

1 Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité –<br />

Universitätsmedizin Berlin, Germany , 2 Dept. of Dermatology, Charité –<br />

Universitätsmedizin Berlin, Germany, 3 Neuroscience Research Center, Charité –<br />

Universitätsmedizin Berlin, Germany<br />

Purpose: Previously, we have demonstrated that neuronal plasticity in skin is<br />

substantially influenced by inflammatory mediators. Here, we tested the hypothesis<br />

whether experimental inflammation may be a useful tool to promote the regeneration<br />

of skin nerves after lesion. Methods: Skin was denervated by axotomy of all skin<br />

nerves, which innervate the dorsal back skin of mice. Sham-operated mice without<br />

axotomy were used as controls. Experimental dermatitis was induced either by<br />

capsaicin or by sensitizing mice with ovalbumin and complete Freund’s adjuvant to<br />

provoke a contact dermatitis-like skin inflammation or with ovalbumin and aluminium<br />

hydroxide to provoke an atopic dermatitis-like skin inflammation. Passive cutaneous<br />

anaphylaxis (PCA) was induced by specific IgE anti-albumin-dinitrophenol followed by<br />

dinitrophenol-HAS. Mast cell activation was determined by measuring ear swelling and<br />

Evans blue extravasation. The numbers of immune cells and nerve fibers were<br />

determined by standard immunohistochemistry: Mast cells (FITC-avidin, GIEMSA),<br />

macrophages (F4/80), T cells (CD3), nerve fibers (PGP9.5, GAP-43). Results: Skin reinnervation<br />

in all compartments was detectable at day 28 after axotomy. Capsaicininduced<br />

inflammation and PCA was significantly reduced after axotomy. Injection of<br />

inhibitors of the neuropeptides Substance P and Calcitonin gene-related peptide also<br />

substantially reduced mast cell-dependent inflammation. Axotomy also significantly<br />

reduced T cell and macrophage infiltration in atopic and contact dermatitis skin.<br />

However, in contact dermatitis the remaining inflammation was sufficient to increase<br />

significantly the number of regenerating nerve fibers in the epidermis, dermis and<br />

subcutis as well as in selected hair follicle compartments. Conclusions: These data<br />

suggest that mast cell-dependent dermatitis is suppressed by axotomy, while mast<br />

cell-independent skin inflammation is sufficient to promote the regeneration of skin<br />

innervation after lesion.<br />

Poster Abstracts 60


International Conference „Development of Somatosensation and Pain 2008“<br />

P14 A tether link required for touch<br />

Jing Hu, Li-Yang Chiang, Bettina Erdmann, Gary Lewin<br />

<strong>MDC</strong><br />

Genetic analysis of touch sensation in C. elegans and research on mechanosensitive<br />

hair cell in inner cochlea have suggested the existence of an extracellular link which<br />

tethers the mechanosensitive ion channels to matrix and transduces mechanical force<br />

to the channels (Ernstrom & Chalfie. 2002. Annu Rev Genet 36: 411-53; Syntichaki &<br />

Tavernarakis. 2004. Physiol Rev 84(4) 1097-153). So far there is no evidence<br />

demonstrating that an extracellular link is essential for mechanotransduction (Lewin &<br />

Moshourab. 2004. Neurobiol 61(1): 30-44). Recent evidence suggests that<br />

mechanotransduction components and mechanisms may be conserved between C.<br />

elegans and mammals (Wetzel, Hu et al. 2007. Nature 445(7124): 206-9). To address<br />

the question whether an extracellular link is required in mouse mechanosensation, we<br />

have established co-cultures of sensory neurons with mouse 3T3 cells (mouse<br />

embryonic fibrbroblast cell line). We found that stimulating the fibroblast cell adjacent<br />

to the neurite almost always (94%) evoked fast mechanically gated currents with<br />

latencies and activation kinetics indistinguishable from those found by stimulating the<br />

neurite directly. This indicates that there is likely a physical attachment between ion<br />

channels in the neurite and the fibroblast cell that is required for normal gating of the<br />

mechanically sensitive current. We have also found that one type of mechanically<br />

activated current-the rapidly adapting current (RA) is exclusively abolished by a serine<br />

endopeptidase-Subtilisin. This ablation of mechanosensitivity could be totally<br />

recovered after further incubating the cells for 30 hrs in culture medium. Using<br />

transmission electron microscopy (TEM) to visualize the ultrastructure of the<br />

attachment between cultured neurons and the underlying substrate, we identified a<br />

novel link protein (length 104.1 ± 3.1nm) tethering sensory neuronal neurites to<br />

extracellular matrix. After Subtilisin treatment we have found this tether link was<br />

missing. And it reappeared after 30 hours incubation coincident with the reappearance<br />

of RA mechanosensitive currents. This is the first evidence that an extracellular link<br />

required for vertebrate mechanotransducer function exists. We conclude that a long<br />

extracellular tether link plays an essential role in mouse sensory<br />

mechanotransduction.<br />

61 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P15 Molecular interactions between stomatin-like proteins and acid-sensing ion<br />

channels<br />

Julia Jira, Paul Heppenstall<br />

Klinik fuer Anaesthesiologie, Charite CBF<br />

Stomatin-like proteins (SLPs) are widely expressed across species and tissues.<br />

However, the function of this family of membrane-associated proteins remains to be<br />

elucidated. Some SLPs bind cholesterol as well as actin filaments and are therefore<br />

considered to have a scaffolding function, assembling protein complexes at the<br />

plasma membrane. Moreover, there is evidence of SLPs directly interacting with ion<br />

channels. In the nematode worm C.elegans, the stomatin homolog MEC-2 plays an<br />

essential role in mechanosensation by anchoring the transduction channel MEC-4 to<br />

the cytoskeleton. We are interested in defining the nature of the interactions between<br />

mammalian counterparts of the C.elegans proteins, which have been implicated in<br />

mechanotransduction as well. To this end, we have performed co-immunoprecipitation<br />

and FRET experiments to examine interactions between mouse stomatin-like protein 3<br />

(mSLP3) and members of the acid-sensing ion channel (ASIC) family. These are<br />

sodium channels that are homologous to the C. elegans MEC-4 channel and comprise<br />

short intracellular N- and C-termini, two transmembrane domains and a large<br />

extracellular loop. All ASIC subunits (ASIC1a, 1b, 2a, 2b, 3 and 4) and mSLP3 were<br />

epitope tagged and transiently transfected into two cell lines (COS-7 and CHO) that<br />

are thought to contain no endogenous ASICs. We found that mSLP3 did<br />

immunoprecipitate with all ASICs. Interestingly, a splice variant of ASIC3, which<br />

misses the C-terminus and the second transmembrane domain, also interacted with<br />

mSLP3. This suggests that the interaction site is located more towards the N-terminus<br />

of the ASIC channels.<br />

Poster Abstracts 62


International Conference „Development of Somatosensation and Pain 2008“<br />

P16 The zinc finger transcription factor Bcl11a/Ctip1 is essential for neuronal<br />

differentiation and sensory circuit formation in dorsal spinal cord<br />

development<br />

Anita John 1 , Heike Brylka 1 , Pentao Liu 2 , René Jüttner 3 , E. Bryan Crenshaw III 4 ,<br />

Nancy A. Jenkins 5 , Neal G. Copeland 5 , Carmen Birchmeier 3 , Stefan Britsch 1<br />

1 Institute for Molecular and Cellular Anatomy, Ulm University, 89081 Ulm, Germany,<br />

2 The Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, 3 Max Delbrück Center<br />

for Molecular Medicine (<strong>MDC</strong>), 13125 Berlin-Buch, Germany, 4 Mammalian<br />

Neurogenetics Group, Center for Childhood Communication, The Children's Hospital<br />

of Philadelphia, PA 19104, USA, 5 Mouse Cancer Genetics Program, Center for<br />

Cancer Research, National Cancer Institute, Frederick, Maryland 21702, USA<br />

There is emerging evidence that common regulatory mechanisms are utilized in the<br />

control of neurogenesis and the development of the lympho-hematopoetic system.<br />

The zinc finger transcription factor Bcl11a (Evi9, CTIP1) is expressed in<br />

lymphohematopoetic tissues and in the nervous system. Bcl11a is essential for normal<br />

lymphoid development, and Bcl11a null mice lack mature B-lymphocytes. Here we<br />

demonstrate that Bcl11a is essential for neuronal development as well. Conditional<br />

ablation reveals that dorsal spinal neurons require Bcl11a for terminal differentiation<br />

and morphogenesis. Moreover, TrkA positive sensory afferents depend in their ability<br />

to grow into the dorsal horn and to provide synaptic input on the expression of Bcl11a<br />

in postsynaptic spinal target neurons. A genome-wide screen for transcriptional<br />

targets down-regulated in Bcl11a mutant spinal neurons identified genes linked to the<br />

regulation of cytoskeletal dynamics, as well as secreted factors with established<br />

functions in differentiation and guidance of neurons. Together, the genetic analysis of<br />

Bcl11a reveals, for the first time, essential functions of this factor in central nervous<br />

system development. Our data suggest that Bcl11a is required to orchestrate key<br />

developmental events, which ultimately lead to the establishment of functional<br />

neuronal circuits.<br />

63 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P17 Sequential maturation of sensory neuron mechanotransduction during<br />

embryonic development<br />

Stefan G. Lechner, Rui Wang, Henning Frenzel, Gary R. Lewin<br />

Max Delbrück Centrum f. Molekulare Medizin<br />

We have reported the presence of three types of mechanically activated currents in<br />

adult mouse dorsal root ganglion (DRG) neurons. These currents can readily be<br />

distinguished by their inactivation kinetics and are thus classified into rapidly adapting<br />

(RA-type), intermediately adapting (IA-type) and slowly adapting (SA-type) currents. In<br />

the present study we asked when during embryonic development do sensory neurons<br />

acquire these mechanosensitive currents. Our data shows that mechanosensitive<br />

currents emerge in three major waves that coincide with already well-described<br />

epochs in the development of the sensory ganglia. First, at E13.5, mechanoreceptors<br />

and proprioceptors acquire RA-type currents, followed by nociceptors which start to<br />

acquire RA-currents at E15.5. The third wave of mechanosensitivity occurs just after<br />

birth when a large proportion of nociceptors acquire an SA-type current. In addition we<br />

investigated the electrical properties of developing sensory neurons. We found that<br />

both, mechanoreceptors and nociceptors, become electrically excitable immediately<br />

after they are born, at E11.5 and at E12.5-E13.5, respectively. Thus the acquisition of<br />

electrical excitability precedes that of mechanosensitivity by approximately two days.<br />

Moreover, we found that the emergence of mechanically gated currents in nociceptors<br />

correlates with significant changes in the electrical excitability of these neurons. Since<br />

the first two waves coincide with the innervation of peripheral targets by<br />

mechanoreceptors (E13.5) and nociceptors (E15.5), we next asked whether target<br />

derived factors are required for the acquisition of mechanosensitivity. Therefore DRG<br />

neurons from non-mechanosensitive stages were differentiated in-vitro in the<br />

presence of different neurotrophins. These experiments strongly suggest that the<br />

acquisition of mechanosensitive currents by nociceptors is induced by target derived<br />

NGF, whereas the acquisition of RA-currents by mechanoreceptors may be a timedependent<br />

process which is independent of the presence of neurotrophic factors.<br />

Supported by SFB 665<br />

Poster Abstracts 64


International Conference „Development of Somatosensation and Pain 2008“<br />

P18 Characterization of the peripheral Osmoreceptor<br />

Soeren Markworth 1 , Silke Frahm 1 , Ines Ibanez-Tallon 1 , Jens Jordan 2 , Gary R. Lewin 1<br />

1 Molecular Physiology of Somatic Sensation, Department of Neuroscience,<br />

Max-Delbrück Center for Molecular Medicine, Robert-Rössle Str. 10, Berlin-Buch,<br />

2 Franz-Volhard Centrum für Klinische Forschung, Haus 129, Charité Campus Buch,<br />

Wiltbergstr. 50, Berlin-Buch<br />

Osmosensitive fibers are believed to be located in the liver and/or the hepatic branch<br />

of the portal vein and project to the CNS via hepatic nerves. Peripheral osmosensitive<br />

fibers have not yet been visualized or functionally characterized, indeed the location<br />

and sensory origin of such fibers is not known. Here we have studied the origin as well<br />

as the activation mechanism of hepatic osmosensitive nerve fibers. We show that in<br />

rats and mice drinking water leads to a decrease of blood osmolality in the portal vein<br />

by 25mOsm. We visualized activation of hepatic sensory fibers by removing the tissue<br />

shortly after the stimulus and immunostaining for phosphorylated ERK (extra-cellularsignal-related-kinase).<br />

The osmotic stimulus led to a robust induction of pERK in a<br />

sub-population of Isolectin B4-negative sensory nerve fibers. We made acute cultures<br />

of dorsal root ganglion neurons from the thoracic ganglia (T7-T13) a proportion of<br />

which innervate visceral tissues including the liver. Of these cells 31% show a robust<br />

increase in intracellular calcium following stimulation with hypotonic solutions (Fura-2<br />

based calcium imaging). Sensory neurons taken from ganglia that do not innervate<br />

viscera had a significantly smaller number of osmosensitive cells. The Transient<br />

Receptor Potential Vanniloid 4 (Trpv4) is believed to play a role in osmosensation.<br />

Here we show using Ca2+-Imaging and patch clamp techniques a significant decrease<br />

in osmosensitive cells of Trpv4 -/- animals compared to WT animals only in the<br />

thoracic, but not in the cervical and lumbar region. We found that only small to<br />

medium sized cells of Trpv4 -/- animals have lost their ability to respond to osmotic<br />

challenges. We also examined a transgenic mouse in which the alpha3 subunit of the<br />

nicotinic acetylcholinreceptor is fused to GFP (green fluorescent protein). We show,<br />

that ERK becomes activated in this subpopulation of neurons after water intake. The<br />

percentage of GFP+ cells showing osmosensitivity in ca-imaging experiments was<br />

much higher than GFP- cells in thoracic ganglia. This animal model to study the<br />

activation of peripheral osmoreceptors may prove a valuable tool to dissect the<br />

physiological relevance of this sensory pathway.<br />

65 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P19 Analysis of Opioid receptor/K+ channel coupling in sensory neurons<br />

D. Nockemann, C. Stein, P. A. Heppenstall<br />

Opioids are the most effective and widely used drugs in the treatment of severe pain.<br />

They act through G-protein-coupled receptors located on central and peripheral<br />

sensory neurons. Three classes of opioid receptors (mu, delta, kappa) have been<br />

identified. Several mechanisms are involved in opioid analgesia including G-proteingated<br />

inwardly rectifying K+ (GIRK) channel activation and inhibition of voltage-gated<br />

Ca2+ channels. Interestingly, in peripheral sensory neurons Ca2+ channel inhibition<br />

has been the most studied mechanism and GIRK channels have not been considered<br />

in any detail. The GIRK channels form homo- or heterotetrameric complexes, and are<br />

known to support the inhibitory effects of opioids in the central nervous system. The<br />

subunits GIRK1 and 2 have been found to be expressed within the spinal cord dorsal<br />

horn, the cerebellum, the hippocampus and the cortex. We are interested in whether<br />

GIRK channels are present and whether opioids are coupled to GIRKs on peripheral<br />

sensory fibers. Furthermore, we will investigate the role of GIRK/opioid receptor<br />

coupling in a mouse model of inflammatory and neuropathic pain. To approach these<br />

questions, we are currently performing quantitative RT-PCR, western blot and patch<br />

clamping experiments. We found no significant expression of all 4 GIRK subunits in<br />

sensory neurons using RT-PCR and western blot. No change in the expression level<br />

could be observed after inflammation of the mouse hindpaw. We also investigate<br />

functional coupling of GIRK channels and opioid receptors in peripheral sensory<br />

neurons using Patch clamping. We found no evidence for inward rectifying currents in<br />

mouse DRG neurons, either before or after treatment with GIRK agonist DAMGO. Our<br />

data indicate that GIRK channels are not present in sensory neurons and do not<br />

contribute to peripheral analgesia under normal and inflamed conditions.<br />

Poster Abstracts 66


International Conference „Development of Somatosensation and Pain 2008“<br />

P20 Direct inhibition of nociceptors by mu-opioid receptor agonist in<br />

neuropathic pain<br />

Yvonne Schmidt, Paul A. Heppenstall, Shaaban A. Mousa, Halina Machelska<br />

Anaesthesiologie, Charité - Campus Benjamin Franklin, Berlin, Germany<br />

After nerve injury (e.g. compression, limb amputation) normally innocous tactile<br />

stimulation (e.g. skin to clothes contact) can result in painful sensation. Currently used<br />

centrally acting opioids are often associated with CNS-mediated side effects. These<br />

can be avoided by selective activation of peripheral opioid receptors without affecting<br />

their analgesic properties. Here we investigate the expression pattern of peripheral<br />

mu-opioid receptors and the effects of their activation on nociceptor thresholds to<br />

mechanical stimuli in a mouse model of neuropathic pain. In a chronic constriction<br />

injury (CCI) of the sciatic nerve we examined the expression of mu-opioid receptor in<br />

lumbar (L4-L5) dorsal root ganglia (DRG) with immunohistochemistry and a coexpression<br />

of mu-opioid receptors with different sensory neuronal markers in DRGs<br />

and in the ligated nerve by double immunofluorescence. Effects of mu-receptor<br />

agonist DAMGO on von Frey-induced mechanical stimulation were tested in an in vitro<br />

skin-saphenous nerve preparation. All experiments were performed 2 weeks after<br />

CCI, sham-surgery or in animals without operation. We found that the number of DRG<br />

cells expressing mu-opioid receptors did not significantly change after CCI. In DRG<br />

and in the ligated nerves, mu-opioid receptors were co-expressed with calcitonin<br />

gene-related peptide and to a lesser degree with isolectin B4 or neurofilament 200,<br />

markers of peptidergic C and A fibers, non-peptidergic C fibers, and A fibers,<br />

respectively. The conduction velocity and the threshold to mechanical stimuli in<br />

sensory afferent fibers between ligated and unligated nerves did not differ significantly.<br />

In the injured nerve about 50% of Aä and C fibers, but not Aâ fibers, were DAMGOsensitive,<br />

resulting in an increased mechanical threshold after DAMGO application to<br />

their receptive field. This effect was reversed by CTOP, suggesting a selective muopioid<br />

receptor-mediated action. Our results suggest that peripheral mu-opioid<br />

receptors can be directly activated in cutaneous C and Aä fibers to attenuate their<br />

excitability after nerve injury. This might constitute a basis for the peripheral opioidmediated<br />

blockade of mechanical allodynia in neuropathic pain.<br />

67 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P21 Prevention of opioid peptide degradation for pain control in peripheral<br />

inflamed tissue<br />

Anja Schreiter 1 , Carmen Gore 1 , Shaaban A. Mousa 1 , Bernard P. Roques 2 ,<br />

Christoph Stein 1 , Halina Machelska 1<br />

1 Anaesthesiologie, Charité - Campus Benjamin Franklin, Berlin, Germany.,<br />

2 CNRS UMR, Paris, France.<br />

Background and aims. Degradation of opioids by peptidases such as aminopeptidase<br />

N (APN) and neutral endopeptidase (NEP) has been extensively examined in the<br />

central nervous system. In peripheral inflamed tissue immune cells liberate opioid<br />

peptides, which bind to opioid receptors on sensory nerve terminals to inhibit pain. Our<br />

aim was to investigate the prevention of opioid peptide degradation by blockade of<br />

APN and NEP in leukocytes to enhance analgesic activity of opioids in inflammatory<br />

pain. We used classical inhibitors of NEP (thiorphan) and of APN (bestatin) as well as<br />

RB3008, a novel dual inhibitor of NEP and APN. Methods. Experiments were<br />

performed at 4 days after injection of Freund’s adjuvant into one hind paw in rats.<br />

Analgesic effects of peptidase inhibitors were assessed with the paw pressure test.<br />

Expression of APN, NEP and opioid peptides in inflamed tissue was analyzed by<br />

immunohistochemistry. Enzymatic activity of APN and NEP was assessed using<br />

membranes prepared from immune cells isolated from inflamed paws, and was<br />

measured spectrometricaly or with radioimmunoassay. Results. Co-injection of<br />

thiorphan and bestatin or application of RB3008 into inflamed paws produced dosedependent<br />

analgesia. APN, NEP, ß-endorphin and Met-enkephalin were expressed in<br />

leukocytes in inflamed tissue. In in vitro immune cell membrane assay APN and NEP<br />

cleaved their specific exogenously added substrates (Ala-ß-naphthylamin and Suc-<br />

Ala-Ala-Phe-p-nitroanilin, respectively) that could be fully blocked with single and dual<br />

peptidase inhibitors. Importantly, degradation of exogenously applied Met-enkephalin<br />

and Leu-enkephalin, but not of ß-endorphin and dynorphin, was prevented by<br />

thiorphan + bestatin or by RB3008. Conclusions. We show for the first time that<br />

degradation of opioids can be prevented by blockade of APN and NEP expressed on<br />

immune cells in painful inflammation. Enkephalins appear as preferred substrates for<br />

NEP and APN. Inhibiting the enzymatic degradation of opioids in peripheral inflamed<br />

tissue offers a promising strategy for the pain control without adverse side effects.<br />

Finally, the use of dual inhibitors has the additional advantage of avoiding different<br />

pharmacokinetics and bioavailability when two “single” inhibitors are employed.<br />

Poster Abstracts 68


International Conference „Development of Somatosensation and Pain 2008“<br />

P22 TRPC5 channel is activated by osmotically induced membrane stretch<br />

Sergio Soriano, Carlos Belmonte, Félix Viana, Ana Gomis<br />

Instituto de Neurociencias de Alicante. UMH-CSIC. Av. Ramón y Cajal s/n. 03550<br />

Sant Joan d'Alacant. Alicante. Spain<br />

Mammalian canonical transient receptor potential (TRPC) genes encode a family of<br />

nonselective cation channels that are activated following stimulation of G-proteincoupled<br />

membrane receptors linked to phospholipase C. TRPCs are molecular<br />

candidates for Ca2+ entry channels in brain since they are highly expressed in the<br />

nervous system, form Ca2+-permeable channels in recombinant expression systems<br />

and are activated by agonist that induce intracellular Ca2+ release. However, their<br />

mechanisms of activation and endogenous regulators remain poorly understood. We<br />

have been focused our studies on the TRPC5 channel. We used a combination of<br />

cellular Ca2+ imaging and patch-clamp techniques. We show that hypoosmotic stimuli<br />

(210mOsm), that cause membrane stretch, produce an increase in the intracellular<br />

calcium concentration in HEK293 cells transfected with TRPC5. During whole-cell<br />

recordings, the application of hypotonic stimulus activates the characteristic doubly<br />

rectifying currents mediated by TRPC5. The activation of TRPC5 is promoted with the<br />

co-expression of the histamine receptor type I (H1). Activation of inositol<br />

polyphosphate 5-phosphatase IV (5ptase IV), which reduce membrane<br />

phosphatidylinositol 4,5-bisphosphate (PIP2) levels, abolished hipotonically-evoked<br />

activation of TRPC5. This response was recovered by increasing intracellular PIP2<br />

levels through the patch pipette. We also show the expression of TRPC5 in sensory<br />

neurons suggesting that the activation of TRPC5 by osmotically induced stretch may<br />

play a role in sensory transduction.<br />

69 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P23 Opioid withdrawal increases TRPV1 activity in a PKA dependent manner<br />

Viola Spahn, Christian Zoellner<br />

Charite, Klinik fuer Anaesthesiologie und operative Intensivmedizin<br />

Vanilloid receptor type 1 (TRPV1) is a ligand-gated ion channel expressed on sensory<br />

nerves that responds to noxious heat, protons, and chemical stimuli such as<br />

capsaicin. TRPV1 plays a critical role in the development of tissue injury, inflammation<br />

or nerve lesions. Opioids such as morphine have been used widely for the treatment<br />

of many types of acute and chronic pain. Application of morphine leads to a<br />

dissociation of G-proteins and causes a reduced activity of adenylylcyclases (AC),<br />

resulting in a lower amount of cAMP. However, opioid withdrawal following chronic<br />

activation of the ƒÊ opioid receptor induces AC superactivation and subsequently an<br />

increase in cAMP and Protein Kinase A (PKA) activity. In the current project we<br />

investigated wether an increase in cAMP during opioid withdrawal increases the<br />

activity of TRPV1. In whole cell patch clamp and calcium imaging experiments opioids<br />

significantly increase capsaicin induced TRPV1 activity in a nalaxone and pertussis<br />

toxin sensitive manner. The role of different PKA phosphorylation sites at TRPV1 was<br />

investigated using site-directed mutagenesis. In summary, our results demonstrate<br />

that opioid withdrawal can increase the activity of TRPV1. These observations show a<br />

new mechanism underlying hyperalgesia during opioid withdrawal.<br />

Poster Abstracts 70


International Conference „Development of Somatosensation and Pain 2008“<br />

P24 A molecular dissection of TRPV1 sensitisation using the naked mole-rat<br />

Ewan St. John Smith, Gireesh Anirudhan, Gary Lewin<br />

Max-Delbrueck Centrum, Berlin-Buch, Germany<br />

Tissue inflammation leads to pain and behavioural sensitisation to thermal and<br />

mechanical stimuli called hyperalgesia. However, the African naked mole-rat (NMR,<br />

Heterocephalus glaber) has total behavioural insensitivity to capsaicin and acid.<br />

Additionally, nerve growth factor (NGF) fails to induce thermal hyperalgesia, a<br />

phenomenon dependent upon TRPV1. We aimed to discover why NGF fails to induce<br />

thermal hyperalgesia in NMRs. To investigate if NGF sensitises NMR TRPV1, wholecell<br />

patch clamp recordings were made from isolated DRG neurones. In mouse the<br />

isolectin B4 (IB4) labels DRG neurones that lack the NGF receptor TrkA and<br />

immunocytochemistry demonstrated the same pattern in NMR DRG cells. NGF<br />

sensitised the capsaicin-gated current in mouse DRG neurones that were IB4-ve (n =<br />

11), but failed to do so in either IB4+ve or IB-ve NMR DRG neurons (n = 14). NMR<br />

TRPV1 was subsequently cloned and found to possess a tyrosine at position 200 that<br />

in other species is fundamental for NGF-induced sensitisation in TRPV1. Furthermore,<br />

when expressed in CHO cells cloned NMR TRPV1 is activated by heat, capsaicin, pH<br />

and voltage in a similar fashion to TRPV1s from other species. We introduced NMR<br />

TRPV1 into DRG neurones from TRPV1-/- mice and in this context NGF caused<br />

potentiation of capsaicin gated currents in IB4-ve neurones (n = 8), but not in IB4+ve<br />

neurones (n = 3). This data suggests that there is something about NMR neurones or<br />

NMR TrkA that is responsible for the lack of sensitisation observed in NMR DRG<br />

neurones. To determine if the cellular environment is critical for NGF induced<br />

sensitisation, a new fibroblast cell line was established from NMR kidney tissue. Four<br />

clones were isolated and have reached population doublings in excess of 50, a formal<br />

criterion that indicates that these cell clones have been immortalized. We are using<br />

these cells to test the hypothesis of whether the cellular environment of NMR cells is<br />

responsible for the lack of NGF induced sensitisation. In summary we find that NGF<br />

fails to sensitise the NMR TRPV1 ion channel. This finding may account for the lack of<br />

NGF-induced thermal hyperalgesia in NMRs. Evidence suggests that there is a<br />

difference in NMR TrkA or downstream signalling that underlies the lack of<br />

sensitisation and these mechanisms are under investigation.<br />

71 Poster Abstracts


May 14 – May 17, 2008 – Berlin, Germany<br />

P25 Analysis of phosphorylation targets of cGMP-dependent kinase I involved<br />

in sensory axon bifurcation<br />

Agne Stonkute, Hannes Schmidt, Fritz G. Rathjen<br />

Max Delbrück Center for Molecular Medicine, Developmental Neurobiology Group,<br />

Berlin, Germany<br />

Our lab identified a cGMP signaling pathway via Npr2 (natriuretic peptide receptor 2)<br />

and cGMP-dependent protein kinase I (cGKI) that is important for axonal pathfinding<br />

and connectivity of sensory neurons. Upon arrival at the dorsal root entry zone<br />

(DREZ) of the spinal cord sensory axons bifurcate and grow further in both rostral and<br />

caudal directions. However, sensory axons of mice lacking cGKI do not bifurcate, they<br />

turn instead and grow in either caudal or rostral direction. This axonal pathfinding error<br />

has physiological implications. Electrophysiological studies showed that the absence<br />

of bifurcation leads to a loss of functional connectivity between sensory neurons and<br />

second order neurons within the dorsal horn. These initial studies raised the question<br />

which phosphorylation targets of cGKI mediate the process of axon bifurcation. In our<br />

search for downstream components of the cGMP signaling cascade in sensory<br />

neurons we analysed mice deficient for established phosphorylation substrates of<br />

cGKI. As a result we can exclude the Ena/Vasp family members Mena (mammalian<br />

Ena) and VASP (vasodilator-stimulated phosphoprotein) as candidate-proteins that<br />

could play a role in axon bifurcation. Furthermore, we screened for novel<br />

phosphorylation targets of cGKI using an antibody recognizing a phosphorylation<br />

consensus motif of cGKI. Western blots with the phospho-sequence specific antibody<br />

revealed several proteins that become phosphorylated upon stimulation of cGKI in<br />

dorsal root ganglia (DRG) or in the DRG-derived cell line F11. Currently we focus on<br />

the enrichment of these components to enable mass spectrometry analysis.<br />

Poster Abstracts 72


International Conference „Development of Somatosensation and Pain 2008“<br />

P26 New In Vitro mechano-assay reveals distinct intrinsic mechanosensitive<br />

subtypes in cultured sensory neurons<br />

Dmitry Usoskin, Per Uhlen, Patrik Ernfors<br />

Mol.Neurobiology /MBB, Karolinska Institute, Stockholm, Sweden<br />

We developed a novel in vitro assay for studying intrinsic mechanosensitive properties<br />

of sensory neurons. Dissociated sensory neurons are plated in specially developed<br />

compartmentized chamber. After several days in culture axons grow to another<br />

compartment where they are massively mechanically stimulated in a controlled way.<br />

Cell response is monitored using alive Ca2+ imaging of somas. Several functional<br />

subtypes could be reproducibly distinguished based on response profile in this assay.<br />

Some of subtypes have correlation with expressed neuronal marker, soma diameter<br />

and responsiveness to capsaicin.<br />

73 Poster Abstracts


P27 Function of Cav3.2 in Sensory neuron mechanosensitivity<br />

Rui Wang, Gary R. Lewin<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Molecular physiology and somatic sensation Group, Department of Neuroscience,<br />

Max-Delbrück-Center for Molecular Medicine Berlin-Buch, Germany<br />

Aim of investigation: Voltage-gated Ca2+channels mediate calcium influx in response<br />

to membrane depolarization and regulate intracellular processes such as contraction,<br />

secretion, neurotransmission, and gene expression. T-type Ca2+currents are<br />

activated by weak depolarization and are transient. They were first described in<br />

sensory neurons and also expressed in a wide variety of other cell types, where they<br />

were involved in shaping the action potential and controlling repetitive firing behavior.<br />

Previous findings from our lab demonstrated that the cDNA encoding the T-type<br />

Cav3.2 calcium channel was almost exclusively expressed by D-hair receptors in the<br />

DRG (Shin et al., 2003). D-hair receptors are very sensitive mechanoreceptors and we<br />

provided pharmacological evidence that the T-type current is required for normal<br />

excitability of this mechanoreceptor. In contrast several other groups have proposed<br />

that Cav3.2 is expressed by nociceptive neurons and that it may play a role in acute<br />

and chronic pain. In the present study we would like to determine whether Cav3.2<br />

channels contribute to the normal function of identified mechanoreceptors and<br />

nociceptors. Methods: In the current study we used Cav3.2 null mutant mice and<br />

tested the physiological properties of cutaneous sensory neurons with the in vitro skin<br />

nerve preparation. Results: Our results showed that mechanical sensitivity and<br />

thresholds of nociceptors (AM and C-fibers) were not changed in Cav3.2 null mutant<br />

mice compared to wild type. Among the mechanoreceptors the physiological<br />

properties of slowly adapting receptors (SA) were also not dramatically changed in<br />

Cav3.2 mutant mice. We noted a small increase in the mechanical latency of rapidly<br />

adapting mechanoreceptors (RA), mechanical latency is a measure of the threshold<br />

for activation. D-hair receptors have less mechanical sensitivity and higher mechanical<br />

thresholds for activation in Cav3.2 mutant mice. Conclusion: Our results demonstrate<br />

that Cav3.2 channels are directly or indirectly involved in setting the<br />

mechanosensitivity of mechanoreceptors but do not play an appreciable role in<br />

nociceptive sensory neurons. Acknowledgement: Supported by the DFG<br />

Poster Abstracts 74


International Conference „Development of Somatosensation and Pain 2008“<br />

List of Participants<br />

Sebastian Auer<br />

Robert-Rössle-Str. 10<br />

13125 Berlin<br />

Germany<br />

Fon: 030-9406-3415<br />

auer.sebastian@mdc-berlin.de<br />

Carlos Belmonte<br />

Av. Ramón y Cajal, s/n<br />

03550 Sant Joan d´Alacant, Alicante<br />

Spain<br />

Fon: +34 965919530<br />

Fax: +34 965919549<br />

carlos.belmonte@umh.es and<br />

in.angelines@umh.es<br />

Carmen Birchmeier<br />

Robert Roessle Str. 10<br />

13125 Berlin<br />

Germany<br />

Fon: 030 9406 2403<br />

cbirch@mdc-berlin.de<br />

Steeve Bourane<br />

80 rue Augustin Fliche -BP74103<br />

34091 Montpellier<br />

France<br />

Fon: (33) (0)4 99 63 60 50<br />

Fax: (33) (0)4 99 63 60 20<br />

s.bourane@wanadoo.fr<br />

Michael Brecht<br />

Philippstr. 13 Haus 6<br />

10115 Berlin<br />

Germany<br />

Fon: 030 20936772<br />

Fax: 030 20936771<br />

michael.brecht@bccn-berlin.de<br />

Dominique Bröhl<br />

Robert-Rössle-Strasse 10<br />

13125 Berlin<br />

Germany<br />

Fon: +49-30-9406 3359<br />

d.broehl@mdc-berlin.de<br />

75 List of Participants<br />

Isabelle Bachy<br />

Sheelesvag 1<br />

17177 Stockholm<br />

Sweden<br />

Fon: +46 8 524 87866<br />

isabelle.bachy@ki.se<br />

Yinth Andrea Bernal Sierra<br />

Robert- Rössle Straße 10<br />

13125 Berlin<br />

Germany<br />

Fon: +493094063789<br />

yinth-andrea.bernal-sierra@mdcberlin.de<br />

Walter Birchmeier<br />

Robert-Rössle-Str. 10<br />

13125 Berlin<br />

Germany<br />

Fon: 030 9406 3278<br />

Fax: 030 949 7008<br />

wiss.vorstand@mdc-berlin.de<br />

Janko Brand<br />

Torstr.104<br />

10119 Berlin<br />

Germany<br />

Fon: +49 30 9406 3275<br />

Janko.brand@mdc-berlin.de<br />

Christian Brenneis<br />

Theodor Stern Kai 7<br />

60590 Frankfurt<br />

Germany<br />

Fon: 0049069630183103<br />

Fax: 0049069630183378<br />

C.Brenneis@med.uni-frankfurt.de<br />

Austin Brown<br />

Beckman 115, 279 Campus Dr.<br />

94305 Stanford, Ca<br />

USA<br />

Fon: 650-723-8580<br />

austin.brown@stanford.edu


Jean-François Brunet<br />

46 rue d'Ulm<br />

75005 Paris<br />

France<br />

Fon: 01 44 32 23 21<br />

Fax: 01 44 32 23 23<br />

jfbrunet@biologie.ens.fr<br />

Ombretta Caspani<br />

Hindenburgdamm 30<br />

D-12200 Berlin<br />

Germany<br />

Fon: 030 84452131<br />

ombretta.caspani@charite.de<br />

Marta Ceko<br />

Robert-Rössle Str 10<br />

D 13122 Berlin-Buch<br />

Germany<br />

Fon: 0049 1577 198 9404<br />

marta.ceko@gmail.com<br />

Justyna Cholewa-Waclaw<br />

Robert-Rössle-str. 10<br />

13092 Berlin<br />

Germany<br />

Fon: +49 30 9406 3772<br />

jcholewa@mdc-berlin.de<br />

Thomas Delanne<br />

1 avenue Eugene Schueller<br />

93600 Aulnay sous Bois<br />

France<br />

Fon: 33 1 58 31 71 38<br />

Fax: 33 1 47 56 42 58<br />

tdelanne@rd.loreal.com<br />

Usoskin Dmitry<br />

Scheeles väg 1, A1<br />

171 77 Stockholm<br />

Sweden<br />

Fon: +46-8-52482269<br />

Fax: +46-8-34 19 60<br />

dmitry.usoskin@ki.se<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Patrick Carroll<br />

80, rue Augustin Fliche,<br />

34091 Montpellier cedex 5<br />

France<br />

Fon: (33) 4 99636041<br />

Fax: (33) 4 99636020<br />

carroll@univ-montp2.fr<br />

Cecile Cayla<br />

Hindenburgdamm 30<br />

12200 Berlin<br />

Germany<br />

Fon: 01636927046<br />

cecile.cayla@charite.de<br />

Li-Yang Chiang<br />

Robert-Rössle-str.<br />

13125 Berlin<br />

Germany<br />

Fon: +49-30-94063783<br />

Fax: +49-30-94062793<br />

li-yang.chiang@mdc-berlin.de<br />

Elvira De la Peña García<br />

Campus de San Juan<br />

03550 San Juan de Alicante. Alicante<br />

Spain<br />

Fon: +34965919212<br />

Fax: +34965919561<br />

elvirap@umh.es<br />

Bristol Denlinger<br />

Avda. Ramón y Cajal s/n<br />

03550 San Juan de Alicante<br />

Spain<br />

Fon: +34965919344<br />

Fax: +34965919561<br />

blayne@umh.es<br />

Patrik Ernfors<br />

Scheelesv 1<br />

S-17177 Stockholm<br />

Sweden<br />

Fon: +46 70 3297659<br />

Fax: +46 8 341960<br />

patrik.ernfors@ki.se<br />

List of Participants 76


International Conference „Development of Somatosensation and Pain 2008“<br />

Axel Fischer<br />

Augustenburger Platz 1<br />

13353 Berlin<br />

Germany<br />

Fon: +49-30-450 559851<br />

Fax: +49-30-450 559851<br />

axel.fischer@charite.de<br />

Alistair Garratt<br />

Rpbert-Rössle-Strasse 10<br />

13125 Berlin<br />

Germany<br />

Fon: +493094063785<br />

agarratt@mdc-berlin.de<br />

Ana Gomis<br />

Av. Santiago Ramón y Cajal<br />

03550 Sant Joan d'Alacant. Alicante<br />

Spain<br />

Fon: +34 965919597<br />

Fax: +34 9659549<br />

agomis@umh.es<br />

Martyn Goulding<br />

10010 North Torrey Pines Rd<br />

CA 92037 La Jolla<br />

USA<br />

Fon: +1858 4534100 x1558<br />

goulding@salk.edu<br />

Sven Hendrix<br />

Charitepl. 1<br />

10117 Berlin<br />

Deutschland<br />

Fon: 0176 411 93 272<br />

sven.hendrix@charite.de<br />

Jing Hu<br />

Robert-Rössle-str.<br />

13125 Berlin<br />

Germany<br />

Fon: +49-30-94063783<br />

Fax: +49-30-94062793<br />

jinghu@mdc-berlin.de<br />

77 List of Participants<br />

Henning Frenzel<br />

Weichselstrasse 24a<br />

10247 Berlin<br />

Germany<br />

Fon: +491793936630<br />

henning.frenzel@mdc-berlin.de<br />

David Ginty<br />

725 N. Wolfe Street, PCTB 1015<br />

21205 Baltimore, Maryland<br />

USA<br />

Fon: 410-614-9494<br />

Fax: 410-614-8423<br />

dginty@jhmi.edu<br />

Miriam Goodman<br />

279 Campus Dr<br />

94305 Stanford, CA<br />

USA<br />

Fon: 650-721-5976<br />

Fax: 650-725-8021<br />

mbgoodman@stanford.edu<br />

Dagmar Hackel<br />

Krahmerstr 6-10 /FEM<br />

12207 Berlin<br />

Germany<br />

Fon: 030-84453867<br />

dagmar.hackel@charite.de<br />

Eric Honore<br />

660, route des Lucioles<br />

06560 Valbonne<br />

France<br />

Fon: 33 4 93 95 77 45<br />

Fax: 33 4 93 95 77 04<br />

honore@ipmc.cnrs.fr<br />

Tim Hucho<br />

ihnestrasse<br />

14195 Berlin<br />

Germany<br />

Fon: 030-8413 1243<br />

Fax: 030-8413 1383<br />

hucho@molgen.mpg.de


Ines Ibanez-Tallon<br />

Robert Rossle 10<br />

10125 Berlin<br />

Germany<br />

Fon: +49-30-9406-3411<br />

Fax: +49-30-9406-2430<br />

ibanezi@mdc-berlin.de<br />

Anita John<br />

Albert-Einstein-Allee 11<br />

89081 Ulm<br />

Germany<br />

Fon: 0551-397007<br />

ajohn@uni-goettingen.de<br />

Alexey Kozlenkov<br />

Robert-Roessle str. 10<br />

13125 Berlin<br />

Germany<br />

Fon: +49 30 9406 3212<br />

alexey.kozlenkov@mdc-berlin.de<br />

Dominika Labuz<br />

Hindenburgdamm 30<br />

12200 Berlin<br />

Germany<br />

Fon: +49 30 84453867<br />

dominika.labuz@charite.de<br />

Michaela Langer<br />

Robert Rössle Str. 10<br />

13125 Berlin<br />

Germany<br />

Fon: +49 30 9406 3720<br />

Fax: +49 30 9406 2206<br />

langer@mdc-berlin.de<br />

Stefan Lechner<br />

Robert Rössle Str. 10<br />

13125 Berlin<br />

Germany<br />

Fon: 030-94062853<br />

stefan.lechner@mdc-berlin.de<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Julia Jira<br />

Hindenburgdamm 30<br />

12200 Berlin<br />

Germany<br />

Fon: 03084452131<br />

julia.jira@charite.de<br />

Jane Johnson<br />

5323 Harry Hines Blvd<br />

75390-9111 Dallas<br />

USA<br />

Fon: 214-648-1870<br />

Fax: 214-648-1801<br />

jane.johnson@utsouthwestern.edu<br />

Rohini Kuner<br />

Im Neuenheimer Feld 366<br />

69120 Heidelberg<br />

Germany<br />

Fon: 06221-548289<br />

Fax: 06221-548549<br />

rohini.kuner@pharma.uni-heidelberg.de<br />

Francois Lallemend<br />

Scheeles väg 1-A1-plan2<br />

171 77 Stockholm<br />

Sweden<br />

Fon: +46 8524 876 20<br />

francois.lallemend@ki.se<br />

Liudmila Lapatsina<br />

Robert-Roessle str 10<br />

13125 Berlin<br />

Germany<br />

Fon: +49-30-9406-3783<br />

l.lapatsina@mdc-berlin.de<br />

Gary Lewin<br />

Robert-Rössle str 10<br />

13192 Berlin<br />

Gemany<br />

Fon: 030 94062430<br />

glewin@mdc-berlin.de<br />

List of Participants 78


International Conference „Development of Somatosensation and Pain 2008“<br />

Eric Lingueglia<br />

660 Route des Lucioles<br />

06560 Valbonne - Sophia Antipolis<br />

France<br />

Fon: 33 (0)4 93 95 77 20<br />

Fax: 33 (0)4 93 95 77 04<br />

lingueglia@ipmc.cnrs.fr<br />

Halina Machelska<br />

Krahmerstr. 6<br />

12207 Berlin<br />

Germany<br />

Fon: +49-30-8445 3851<br />

halina.machelska@charite.de<br />

Stephen McMahon<br />

Wolfson Wing, Hodgkin Building, Guy's<br />

Campus<br />

SE1 1UL London<br />

UK<br />

Fon: +44 207 8486270<br />

Fax: +44 207 848 6165<br />

stephen.mcmahon@kcl.ac.uk<br />

Lorne Mendell<br />

Life Science Building<br />

NY 11794 Stony Brook<br />

USA<br />

Fon: 631 632 8632<br />

lorne.mendell@sunysb.edu<br />

Goodman Miriam<br />

B-111 Beckman Center, 279 Campus Dr<br />

94305 Stanford, CA<br />

USA<br />

Fon: 650-723-3100<br />

Fax: 650-725-8021<br />

mbgoodman@stanford.edu<br />

Thomas Mueller<br />

Robert-Roessle-Str. 10<br />

13125 Berlin<br />

Germany<br />

Fon: 49 30 9406 2842/3359<br />

Fax: 49 30 9406 3449<br />

thomu@mdc-berlin.de<br />

79 List of Participants<br />

Qiufu Ma<br />

1 Jimmy Fund Way<br />

MA 02115 Boston<br />

the United States<br />

Fon: 617 632-4594<br />

Qiufu_Ma@dfci.harvard.edu<br />

Sören Markworth<br />

Robert-Rössle-Str. 10<br />

13125 Berlin<br />

Germany<br />

Fon: +49-(0)30-9406-2853<br />

soeren.markworth@mdc-berlin.de<br />

Peter McNaughton<br />

Tennis Court Rd<br />

CB1 2ET Cambridge<br />

UK<br />

Fon: +44 1223 334012<br />

pam42@cam.ac.uk<br />

Nevena Milenkovic<br />

Robert Rossle Str. 10<br />

13092 Berlin<br />

Germany<br />

Fon: +493064063783<br />

milenkovic@mdc-berlin.de<br />

Rabih Moshourab<br />

hindenburgdamm 30<br />

12200 berlin<br />

germany<br />

Fon: 84452733<br />

rabih.moshourab@charite.de<br />

Robert Naumann<br />

Schlegelstr. 5<br />

10115 Berlin<br />

Germany<br />

Fon: 030 21230778<br />

robert.naumann@bccn-berlin.de


Dinah Nockemann<br />

Krahmerstr. 6-10<br />

12207 Berlin<br />

Germany<br />

Fon: 030-84452131<br />

dinah.nockemann@charite.de<br />

Thomas Park<br />

840 West Taylor St.<br />

60607 Chicago<br />

USA<br />

Fon: 312 413 3020<br />

Fax: 312 996 2805<br />

TPark@uic.edu<br />

Kate Poole<br />

Robert-Rössle Str 10<br />

13125 Berlin-Buch<br />

Germany<br />

Fon: (030)94063789<br />

kate.poole@mdc-berlin.de<br />

Michael Schäfer<br />

Hindenburgdamm 30<br />

12200 Berlin<br />

Germany<br />

Fon: 030 8445 2731<br />

micha.schaefer@charite.de<br />

Robert F. Schmidt<br />

Röntgenring 9<br />

97070 Würzburg<br />

Germany<br />

Fon: 0931 31 2639<br />

Fax: 0931 592 10<br />

rfs@mail.uni-wuerzburg.de<br />

Mikael Schnizler<br />

Pauwelsstraße 30<br />

52074 Aachen<br />

Germany<br />

Fon: 0241-80888-06<br />

mschnizler@physiology.rwth-aachen.de<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Otilia Obreja<br />

Theodor-Kutzer-Ufer 1-3<br />

68167 Mannheim<br />

Germany<br />

Fon: +49 621 3835619<br />

Fax: +49 621 3831463<br />

otilia.obreja@anaes.ma.uniheidelberg.de<br />

Andrés Parra<br />

ctra N-332 km 37<br />

03550 Alicante<br />

Spain<br />

Fon: +34 965919344<br />

aparra@umh.es<br />

Fritz G. Rathjen<br />

Robert-Rössle-Str.10<br />

13092 Berlin<br />

Germany<br />

Fon: 49 30 9406 3709<br />

Rathjen@mdc-berlin.de<br />

Hannes Schmidt<br />

Robert-Rössle-Str. 10<br />

13125 Berlin<br />

FR Germany<br />

Fon: +49-30-94063583<br />

hannes.schmidt@mdc-berlin.de<br />

Yvonne Schmidt<br />

Hindenburgdamm 30<br />

12203 Berlin<br />

Germany<br />

Fon: 030 8445 2131<br />

yvonne.schmidt@charite.de<br />

Anja Schreiter<br />

Hindenburgdamm 30<br />

12203 Berlin<br />

Germany<br />

Fon: 0049 30 84453867<br />

anja.schreiter@charite.de<br />

List of Participants 80


International Conference „Development of Somatosensation and Pain 2008“<br />

Jan Siemens<br />

600 16th Street<br />

94158-2517 San Francisco<br />

USA<br />

Fon: +415-476-0432<br />

Fax: +415-502-8644<br />

jan@cmp.ucsf.edu<br />

Viola Spahn<br />

Krahmerstr. 6-10<br />

12207 Berlin<br />

Germany<br />

Fon: 030-8445 2131<br />

Fax: 030-8339 389<br />

viola.spahn@charite.de<br />

Agne Stonkute<br />

Robert-Roesslestr. 10<br />

13092 Berlin<br />

Germany<br />

Fon: 030 94063316<br />

stonkute@mdc.berlin.de<br />

Annika Stuerzebecher<br />

Robert Rössle Straße 10<br />

13125 Berlin<br />

Germany<br />

Fon: 030/94063415<br />

annika.stuerzebecher@mdc-berlin.de<br />

Jean Valmier<br />

80 augustin Fliche<br />

34090 Montpellier<br />

France<br />

Fon: 0033499636007<br />

Fax: 0033499636020<br />

jean.valmier@univ-montp2.fr<br />

Hong Wang<br />

Hindenburgdamm 30<br />

12200 Berlin<br />

Germany<br />

Fon: 00493084452131<br />

hong.wang@charite.de<br />

Dominik Wiemuth<br />

Pauwelsstraße 30<br />

52074 Aachen<br />

Germany<br />

Fon: 0241-8088814<br />

dominik.wiemuth@uni-wuerzbug.de<br />

81 List of Participants<br />

Anantha Krishna Sivasubramaniam<br />

coppistr. 14<br />

10365 Berlin<br />

Germany<br />

Fon: 015775769488<br />

anand987@yahoo.com<br />

Ewan St. John Smith<br />

Robert Rössle Strasse 10<br />

13125 Berlin<br />

Deutschland<br />

Fon: 00493094063783<br />

ewan.smith@mdc-berlin.de<br />

Robert Storm<br />

Robert-Rössle-Str. 10<br />

13125 Berlin<br />

Germany<br />

Fon: +49 - 30/9406 2842<br />

rstorm@mdc-berlin.de<br />

Andrew Todd<br />

University Avenue<br />

G12 8QQ Glasgow<br />

UK<br />

Fon: 0141 330 5868<br />

A.Todd@bio.gla.ac.uk<br />

Daniel Vardeh<br />

Ladenburgerstr. 19<br />

60120 Heidelberg<br />

Germany<br />

Fon: 06221 9143676<br />

danv@gmx.de<br />

Rui Wang<br />

Robert-Rössle-Str 10<br />

13092 Berlin<br />

Germany<br />

Fon: 03094062853<br />

ruiwang@mdc-berlin.de<br />

John Wood<br />

GowertStreet<br />

wc1e 6bt London<br />

UK<br />

Fon: 02076797800<br />

j.wood@ucl.ac.uk


Clifford Woolf<br />

149 13th Street<br />

02129 Charlestown<br />

USA<br />

Fon: 617 724 3622<br />

cwoolf@partners.org<br />

Sandra Zurborg<br />

Krahmerstraße 6-10<br />

12207 Berlin<br />

Germany<br />

Fon: 03084452131<br />

sandra.zurborg@charite.de<br />

May 14 – May 17, 2008 – Berlin, Germany<br />

Hanns Ulrich Zeilhofer<br />

Winterthurerstrasse 190<br />

CH-8057 Zurich<br />

Switzerland<br />

Fon: -41-44-6355912<br />

Fax: -41-44-6355988<br />

zeilhofer@pharma.uzh.ch<br />

List of Participants 82


International Conference „Development of Somatosensation and Pain 2008“<br />

Author Index<br />

A D<br />

Alvania, Rebecca S. T6 de la Peña, Elvira P9<br />

Angioni, Carlo P2 Dina, Olayinka T17<br />

Anirudhan, Gireesh P24<br />

Anjum, Farzana T19 E<br />

Auer, Sebastian P1<br />

Erdmann, Bettina T10<br />

B P7<br />

P14<br />

Bader, Michael P6 Ernfors, Patrik T2<br />

Basbaum, Allan I. T21 P26<br />

Bautista, Diana M. T21<br />

Belmonte, Carlos T30 F<br />

P9<br />

P22 Feil, Robert T5<br />

Birchmeier, Carmen T4 Frahm, Christina T10<br />

T10 Frahm, Silke P18<br />

T20 Frenzel, Henning P17<br />

P3<br />

P8 G<br />

P16<br />

Bormuth, Ingo P3 Garratt, Alistair T10<br />

Boyce, V. T12 P10<br />

Brack, Alexander T24 Geisslinger, Gerd P2<br />

P12 Ginty, David D. T6<br />

Brecht, Michael T19 Glazer, Josh T21<br />

Brenneis, Christian P2 Gomis, Ana P22<br />

Britsch, Stefan P16 Goodman, Miriam T7<br />

Bröhl, Domenique T20 P4<br />

P3 Gore, Carmen P21<br />

Brown, Austin P4 Goswami, Chandan P11<br />

Brunet, Jean-François T28 Griffel, Carola T10<br />

Brylka, Heike P16<br />

Buttgereit, Jens T5 H<br />

C Hackel, Dagmar P12<br />

Hendrix, Sven P13<br />

Carroll, Patrick T3<br />

Caspani, Ombretta T8 Heppenstall, Paul T8<br />

P5 P5<br />

Cayla, Cecile P6 P15<br />

Chiang, Li-Yang P7 P20<br />

P14 Hofmann, Franz T5<br />

Cholewa-Waclaw, Justyna P8 Honore, Eric T9<br />

Copeland, Neal G. P16 Hori, Kei P3<br />

Coste, Ovidiu P2 Hu, Jing T22<br />

Crenshaw III, E. Bryan P16 P7<br />

P14<br />

83 Author Index


May 14 – May 17, 2008 – Berlin, Germany<br />

Hucho, Tim T17<br />

P11<br />

M<br />

Ma, Qiufu T29<br />

I Machelska, Halina T24<br />

P20<br />

Ibanez-Tallon, Ines T22 P21<br />

P1 Madrid, Rodolfo P9<br />

P18 Mandai, Kenji T6<br />

Markworth, Soeren P18<br />

J Martarelli, Benedetta P8<br />

Maurer, Marcus P13<br />

Jenkins, Nancy A. P16 McMahon, Stephen T31<br />

Jira, Julia P15 McNaughton, Peter T11<br />

John, Anita P16 Mendell, L. M. T12<br />

Johnson, Jane T16 Milenkovic, Nevena T10<br />

Jordan, Jens P18 Mousa, Shaaban A. P20<br />

Jordt, Sven-E. T21 P21<br />

Julius, David T21 Müller, Thomas T4<br />

Jüttner, Rene T5 T20<br />

K<br />

P1<br />

P16<br />

N<br />

P3<br />

Nave, Klaus-Armin P3<br />

Koch, Manuel P7<br />

Kuhn, Julia T17 P<br />

Kuner, Rohini T14<br />

P. A. Heppenstall, D. P19<br />

Nockemann, C. Stein<br />

L Park, Thomas T18<br />

Peters, Eva M. P13<br />

Labuz, Dominika T24 Petruska, J. T12<br />

P6 Picker, Björn P13<br />

P12<br />

Lechner, Stefan G. P17 R<br />

Levine, Jon T17<br />

Lewin, Gary T10 Ramanan, Narendrakumar T6<br />

T18 Raouf, Ramin T27<br />

T22 Rathjen, Fritz G. T5<br />

T26 P25<br />

P7 Rittner, Heike T24<br />

P14 P12<br />

P17 Roques, Bernard P. P21<br />

P18 Rugiero, Francois T27<br />

P24<br />

P27 S<br />

Liao, Zhiwen P4<br />

Lingueglia, Eric T23 Schäfer, Michael P6<br />

Liu, Pentao P16 Schäffer, Susanne T5<br />

Schmidt, Hannes T5<br />

P25<br />

Author Index 84


International Conference „Development of Somatosensation and Pain 2008“<br />

Schmidt, Yvonne T24 Y<br />

P20<br />

Scholich, Klaus P2 Yurgionas, Brian T8<br />

Schreiter, Anja T24<br />

P21 Z<br />

Siebenhaar, Frank P13<br />

Sieber, Martin T4 Zeilhofer, Hanns Ulrich T25<br />

Siemens, Jan T21 Zhang, Xuming T11<br />

Soriano, Sergio P22 Zoellner, Christian P23<br />

Spahn, Viola P23 Zurborg, Sandra T8<br />

St. John Smith, Ewan P24 P5<br />

Stein, Christoph P6<br />

P21<br />

Stonkute, Agne T5<br />

P25<br />

Storm, Robert T4<br />

Strehle, Michael P3<br />

Stucky, Cheryl L. T21<br />

Stuerzebecher, Annika T22<br />

T<br />

Tansy , Donovan-Rodriguez P9<br />

Todd, Andrew T13<br />

Treier, Mathias T20<br />

Tsuruda, Pamela R. T21<br />

U<br />

Uhlen, Per P26<br />

Usoskin, Dmitry P26<br />

V<br />

Viana, Felix P9<br />

P22<br />

W<br />

Wang, Rui P17<br />

P27<br />

Wende, Hagen P3<br />

Wickramasinghe, S. Rasika T6<br />

Wildner, Hendrik T20<br />

P8<br />

Wood, John T27<br />

Woolf, Clifford J. T1<br />

85 Author Index

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!