cytoskeleton - Institut d'études scientifiques de Cargèse (IESC)

cytoskeleton - Institut d'études scientifiques de Cargèse (IESC) cytoskeleton - Institut d'études scientifiques de Cargèse (IESC)

iesc.univ.corse.fr
from iesc.univ.corse.fr More from this publisher
22.01.2014 Views

2010 22 Février 26 Février CYTOSKELETON: CONTRACTILITY AND MOTILITY Laurent BLANCHOIN institut de Recherches en Technologie et Sciences pour le Vivant - iRTSV CEA Grenoble 17, rue des Martyrs 38054 Grenoble cedex 9 33(0)438784978 laurent.blanchoin@cea.fr Direction scientifique : Giovanna Chimini Contact : Dominique Donzella tél : 04 95 26 80 40 www.iesc.univ-corse.fr

2010<br />

22 Février<br />

26 Février<br />

CYTOSKELETON:<br />

CONTRACTILITY<br />

AND MOTILITY<br />

Laurent BLANCHOIN<br />

institut <strong>de</strong> Recherches en<br />

Technologie et Sciences pour le Vivant -<br />

iRTSV<br />

CEA Grenoble<br />

17, rue <strong>de</strong>s Martyrs 38054 Grenoble<br />

ce<strong>de</strong>x 9<br />

33(0)438784978<br />

laurent.blanchoin@cea.fr<br />

Direction scientifique :<br />

Giovanna Chimini<br />

Contact :<br />

Dominique Donzella<br />

tél : 04 95 26 80 40<br />

www.iesc.univ-corse.fr


FEBS Advanced Lecture Course on<br />

« Cytoskeleton, Contractility and<br />

Motility »<br />

Pierre Gilles <strong>de</strong> Gennes Winter School 2010<br />

22 - 26 February 2010<br />

Monday 22th February<br />

<strong>Institut</strong> d'Etu<strong>de</strong>s Scientifiques <strong>de</strong> <strong>Cargèse</strong><br />

Corsica, France<br />

08h30 09h00<br />

09h00 10h00<br />

10h00 11h00<br />

11h00 11h30<br />

11h30 12h30<br />

12h30 14h00<br />

14h00 16h00<br />

16h00 16h30<br />

16h30 17h30<br />

17h30 18h30<br />

19h00<br />

09h00 10h00<br />

10h00 11h00<br />

11h00 11h30<br />

11h30 12h30<br />

12h30 14h00<br />

14h00 16h00<br />

16h00 16h30<br />

16h30 17h30<br />

17h30 18h30<br />

19h00<br />

Participants welcome<br />

Phong Tran University of Pennsylvania, USA, and <strong>Institut</strong> Curie, Paris, France<br />

Cytoskeleton , Ce Polarity and Ce Shap<br />

Michael Sixt Max Planck <strong>Institut</strong>e, Martinsried, Germany<br />

Force generation and force transduction in crawling ce s<br />

Co ee break<br />

David Kovar The University of Chicago, USA<br />

Biochemistry of actin and actin binding proteins<br />

Lunch<br />

Oral presentations participants<br />

Co e break<br />

Laurent Blanchoin <strong>Institut</strong>e of life sciences research and technologies iRTSV , Grenoble, France<br />

Actin Dynamics<br />

Jonathan Howard Max Planck <strong>Institut</strong>e, Dres<strong>de</strong>n, Germany<br />

Motor proteins as nanomachines<br />

Dinner<br />

Tuesday 23th February<br />

Enrique M. <strong>de</strong> la Cruz Molecular Biophysics & Biochemistry Department, Yale University, USA<br />

Methods of equilibrium and transient kinetic analysis<br />

Matthias Rief Technische Universität, München, Germany<br />

How forces can be used to influence and investigate biomolecular kinetics in a complex energy landscape part 1<br />

Co ee break<br />

Gijsje Koen<strong>de</strong>rink AMOLF, the Netherlands<br />

Mechanical properties of active cytoskeletal networks part 1<br />

Lunch<br />

Poster presentations + Practical <strong>de</strong>monstrations<br />

Co ee break<br />

Pekka Lappailainen <strong>Institut</strong>e of Biotechnology, Helsinki, Finland<br />

Cytoskeleton membrane interactions<br />

Marko Kaksonen EMBL, Hei<strong>de</strong>lberg, Germany<br />

Actin <strong>cytoskeleton</strong> in membrane trafic<br />

Dinner


Oral Présentations<br />

Names in alphabetical or<strong>de</strong>r<br />

5


Timo BETZ : Timo.Betz@curie.fr<br />

Laboratoire P.C.C. UMR 168, Paris, France<br />

ATP-<strong>de</strong>pen<strong>de</strong>nt mechanics of red blood cells<br />

Msham BRITTO : mishanv@gmail.com<br />

Dublin City University, Ireland<br />

Moesin is a pivotal cytoskeletal modulator in vascular cells during<br />

mechanotransduction, globally regulated by microRNAs<br />

Sebastian FUERTHAUER : bastian@pks.mpg.<strong>de</strong><br />

Max Planck <strong>Institut</strong> für Physik komplexer Systeme, Dres<strong>de</strong>n, Germany<br />

Antisymmetric stress and the role of angular momentum conservation in complex<br />

fluids<br />

Florian HUBER : florian.huber@uni-leipzig.<strong>de</strong><br />

University of Leipzig, Germany<br />

Linker Induced Actin Network Formation un<strong>de</strong>r Cell-Sized Confinement<br />

Svenja-Marei KALISCH : s.kalisch@amolf.nl<br />

FOM-<strong>Institut</strong>e AMOLF Amsterdam, Netherlands<br />

Against the wall: Microtubule dynamics is regulated by force and end-binding<br />

proteins<br />

Jimmy Le DIGABEL : j<br />

jimmy.ledigabel@univ-paris-di<strong>de</strong>rot.fr<br />

Matière et Systèmes Complexes, Benoit Ladoux, Paris, France<br />

Active substrates to study mechanotransduction<br />

Chiu Fan LEE : cflee@pks.mpg.<strong>de</strong><br />

Max-Planck-<strong>Institut</strong>e, Dres<strong>de</strong>n, Germany<br />

Protein amyloid self-assembly<br />

Olga MARKOVA : markova@ibdm.univ-mrs.fr<br />

IBDML – UMR 6216, <strong>Institut</strong>e of Developmental Biology Marseilles<br />

France<br />

Hydrodynamic <strong>de</strong>scription of tissue elongation during Drosophila embryo<br />

morphogenesis: the role of active anisotropic cytoskeletal stresses<br />

Helen MATTHEWS : h.matthews@ucl.ac.uk<br />

MRC Laboratory for Molecular Cell Biology, London; UK<br />

Amsha PROAG : amsha.proag@univ-paris-di<strong>de</strong>rot.fr<br />

Laboratoire Matière et systèmes complexes UMR 7057, Paris, France<br />

Microstructured chambers to study the sensitivity of cells to the geometry and<br />

stiffness of their environment<br />

6


Michal REICHMAN-FRIED : mreichm@uni-muenster.<strong>de</strong><br />

<strong>Institut</strong>e of Cell Biology, ZMBE, Munster, Germany<br />

Migration of primordial germ cell in zebrafish embryo<br />

Nessy TANIA : ntania@math.ubc.ca<br />

Department of Mathematics The University of British Columbia<br />

Vancouver, Canada<br />

Stanilas VINOPAL : vinopal@img.cas.cz<br />

<strong>Institut</strong>e of Molecular Genetics ASCR, Prague, Czech Republic<br />

Localization of gamma-tubulin in nucleolus and its dynamics<br />

7


Posters<br />

Names in alphabetical or<strong>de</strong>r<br />

9


Julia ARENS : julia.arens@mpi-dortmund.mpg.<strong>de</strong><br />

Max-Planck <strong>Institut</strong>e of Molecular Physiology, Dortmund, Germany<br />

A HIGH-CONTENT SCREEN TO IDENTIFY MICROTUBULE<br />

REGULATORSINVOLVEDIN NEURITE INITIATION<br />

During neurite initiation, microtubules are reorganized from a disor<strong>de</strong>red array to form<br />

thick bundles, which fill the neurite shaft. Microtubule associated proteins (MAPs) are<br />

important regulators of this transition. Neuronal cells express MAPs, which modulate<br />

various aspects of microtubule behavior, such as their dynamic stability, their physical<br />

rigidity and force-generating interactions with each other as well as other cellular<br />

compartments.<br />

To evaluate their role in neurite initiation, we knocked-down all known MAPs in a stem<br />

cell-like mo<strong>de</strong>l system via mixtures of siRNAs, and quantified resulting changes in<br />

neuronal morphology via automated fluorescence microscopy and image analysis. In<br />

this screen, we i<strong>de</strong>ntified 40 candidate MAPs, which show significant morphological<br />

changes compared to control cells. Such changes inclu<strong>de</strong> increases and <strong>de</strong>creases in<br />

neurite length as well as enlarged neurite diameter. Of these primary candidates, 24<br />

were confirmed in an in<strong>de</strong>pen<strong>de</strong>nt screen using individual siRNAs. The confirmed<br />

candidates inclu<strong>de</strong> well known MAPs which were expected to play important roles in<br />

neurite initiation, and new candidate regulators, for which no clear role in neurite<br />

initiation has been proposed until now. We are currently performing follow-up studies,<br />

which employ time-lapse microscopic imaging to characterize the dynamics of neurite<br />

initiation in control and knockdown conditions.<br />

10


Richard ARNOLDI : Richard.Arnoldi@unige.ch<br />

Dpt of Pathology and Immunology, Faculty of Medicine, CMU<br />

Geneva, Switzerland<br />

SMOOTH MUSCLE ACTINS: TWO ISOFORMS FOR TWO DISTINCT FUNCTIONS?<br />

Richard Arnoldi and Christine Chaponnier<br />

Dpt of Pathology and Immunology, Faculty of Medicine, University of Geneva, CMU,<br />

Geneva, Switzerland<br />

a-SMA and g-SMA, the two smooth muscle actins which predominate in vascular and<br />

enteric smooth muscle, respectively, are highly conserved across species, differing in<br />

their sequence by only three amino acids. a-SMA has been extensively studied and<br />

recognized for its “contractile” activity in several cell types including smooth muscle,<br />

myofibroblasts, and myoepithelial cells. Anti-a-SMA (clone 1A4) has proven to be a<br />

precious tool in pathology and basic research. Whether g-SMA is its equivalent in the<br />

enteric system is not known mainly because of the lack of tools for an appropriate<br />

investigation. We <strong>de</strong>veloped a new specific monoclonal antibody against g-SMA and<br />

confirmed that g-SMA predominates in the enteric system and is minor in the vascular<br />

system, although more expressed in highly compliant veins than in stiff arteries.<br />

Contrary to a-SMA, g-SMA is completely absent from myofibroblasts within various<br />

fibrosis and stromal reactions, as well as in vitro. g-SMA is present in myoepithelial<br />

cells, but heterogeneously distributed. When g-SMA and a-SMA are coexpressed, our<br />

experiments suggest that their subcellular locations are partly distinct, with a-SMA<br />

contractile fibres extending throughout the entire cell and g-SMA fibres being restricted<br />

to its central part. We raised the hypothesis that, whereas a-SMA is responsible for the<br />

“contractile” activity (active tension), g-SMA would be involved in the “controlled<br />

dilation” (passive tension) in SM and SM-like cells. Several mo<strong>de</strong>ls seem to support<br />

this hypothesis, namely artery vs. vein, and the physiological modifications occurring in<br />

the uterus and mammary glands during pregnancy and lactation. Our results suggest<br />

that, in addition to enteric smooth muscles, g-SMA is expressed in all the tissues<br />

submitted to an important dilation including veins, gravid uterus, and lactating<br />

mammary glands.<br />

11


Katrin BENAKOVITSCH :<br />

Katrin.benakovitsch@unibas.ch<br />

Center for Biomedicine; University Basel, Switzerland<br />

NEW TOOLS FOR SYSTEMS BIOLOGY ANALYSIS OF SPATIO-TEMPORAL<br />

SIGNALING DURING POLARIZED CELL MIGRATION<br />

Polarized cell migration is critical to <strong>de</strong>velopment, the immune system but its<br />

regulation is compromised in pathologies such as metastatic cancer or vascular<br />

diseases. Cell migration processes are highly spatio-temporal regulated and involve<br />

cellular dynamics such as changes in the <strong>cytoskeleton</strong>, generation of polarity or<br />

adhesion dynamics.<br />

In or<strong>de</strong>r to elucidate the highly spatial organized signaling components and networks,<br />

we applied to novel technologies to enable the study of prototypical mo<strong>de</strong>s of<br />

polarized cell migration.<br />

First, we were able to separate pseudopods from their cell bodies and performed<br />

biochemical analysis as well as large scale shotgun proteomics. These informations<br />

about proteomic dynamics in pseudopods and cell bodies will be used in a second<br />

approach performing live cell imaging in a microfluidic system.<br />

This system enables to apply chemokine gradients in a very robust way to stimulate<br />

cells growing on integrated line patterns. One specific application of this tool will be the<br />

tracking of live cells after gene silencing in a highly standardized fashion.<br />

Together, these two techniques will help to resolve spatio-temporal signaling dynamics<br />

during polarized cell migration.<br />

Timo BETZ : Timo.Betz@curie.fr<br />

Laboratoire P.C.C. UMR 168, Paris, France<br />

ATP-DEPENDENT MECHANICS OF RED BLOOD CELLS<br />

Red blood cells are amazingly <strong>de</strong>formable structures able to recover their initial<br />

shape even after large <strong>de</strong>formations as when passing through tight blood capillaries.<br />

The reason for this exceptional property is found in the composition of the membrane<br />

and the membrane-<strong>cytoskeleton</strong> interaction. We investigate the mechanics and the<br />

dynamics of RBCs by a unique noninvasive technique, using weak optical tweezers<br />

to measure membrane fluctuation amplitu<strong>de</strong>s with µs temporal and sub nm spatial<br />

resolution. This enhanced edge <strong>de</strong>tection method allows to span over >4 or<strong>de</strong>rs of<br />

magnitu<strong>de</strong> in frequency. Hence, we can simultaneously measure red blood cell<br />

membrane mechanical properties such as bending modulus, tension and an effective<br />

viscosity that suggests unknown dissipative processes. We furthermore show that<br />

cell mechanics highly <strong>de</strong>pends on the membrane-spectrin interaction mediated by<br />

the phosphorylation of the interconnection protein 4.1R. Inhibition and activation of<br />

this phosphorylation significantly affects tension and effective viscosity. Our results<br />

show that on short time scales (slower than 100 ms) the membrane fluctuates as in<br />

thermodynamic equilibrium. At time scales longer than 100 ms, the equilibrium<br />

<strong>de</strong>scription breaks down and fluctuation amplitu<strong>de</strong>s are higher by 40% than predicted<br />

by the membrane equilibrium theory. Possible explanations for this discrepancy are<br />

influences of the spectrin that is not inclu<strong>de</strong>d in the membrane theory or<br />

nonequilibrium fluctuations that can be accounted for by <strong>de</strong>fining a nonthermal<br />

effective energy that corresponds to an actively increased effective temperature.<br />

12


Thomas BORNSCHOGL: Thomas.Bornschloegl@curie.fr<br />

PhysicoChimie Curie :UMR CNRS 168, Paris, France<br />

Filopodia are small cell protrusions playing important mechanical and sensory roles for<br />

a variety of different cell processes occurring for example during pathogenesis, cell<br />

migration, wound healing or during the dorsal closure in the embryonic <strong>de</strong>velopment.<br />

Filopodia have also been observed pulling invasive bacteria towards the host cell<br />

before infection occurred. However the exact mechanics and the functionality behind<br />

the filopodial retraction mechanism are far from being un<strong>de</strong>rstood. We want to shed<br />

light onto the molecular principles behind the filopodial retraction mechanism when it is<br />

triggered by pathogens such as Shigella. To measure the mechanics we will use an<br />

optical trap combined with confocal microscopy. We will quantify the time resolved<br />

force that a single filopodium exerts onto beads that are coated with different bacterial<br />

ligands or other effectors inducing retraction. By simultaneously observing the<br />

movement of fiduciary marks placed on the filopodial actin core and by locally<br />

perfusing drugs that interfere with cytoskeletal components insi<strong>de</strong> the filopodium we<br />

intend to <strong>de</strong>cipher the un<strong>de</strong>rlying molecular principles. The local perfusion of drugs<br />

that affect the early stage of filopodial adhesion to beads or to the Shigella bacterium<br />

may also help to <strong>de</strong>fine the signaling pathway leading to retraction.<br />

Msham BRITTO : mishanv@gmail.com<br />

Dublin City University, School of health and human performance<br />

Dublin, Ireland<br />

MOESIN IS A PIVOTAL CYTOSKELETAL MODULATOR IN VASCULAR CELLS<br />

DURING MECHANOTRANSDUCTION, GLOBALLY REGULATED BY MICRORNAS<br />

The <strong>cytoskeleton</strong> recruits adapter, linker and scaffolding proteins that modulate cell<br />

plasticity with haemodynamic forces. The FERM domain containing ERM (Ezrin,<br />

Radixin, Moesin) family are key actin binding proteins. Cardiovascular diseases are a<br />

major cause of morbidity and mortality, which results in remo<strong>de</strong>ling of the vasculature.<br />

This remo<strong>de</strong>ling is effected by cell fate <strong>de</strong>cisions such as migration, proliferation etc,<br />

effected by aberrant blood flow.<br />

Our studies show moesin in contrast to ezrin is artheroprotective and regulated by<br />

mechanotransduction in vasculature. Moesin unlike ezrin is found sensitive to<br />

thrombosis and haemostatic stimuli. Moesin through cyclic strain is also <strong>de</strong>monstrated<br />

to mediate αVβ3/α5β1 integrins, G-coupled protein FPRL1 (Formyl pepti<strong>de</strong> receptor<br />

1), urokinase-uPAR interaction. These interactions were elucidated through the<br />

investigation of cell motility. Proliferation, angiogenesis, permeability and<br />

microparticles in vascular cells showed the same paradigm. Moreover, microRNA is<br />

<strong>de</strong>monstrated to globally regulate these major functions in endothelial cells, including<br />

endothelial cell and actin realignment with with blood flow.<br />

For cell motility/mechanics/mechanotransduction among other functions, we find a<br />

requirement for moesin <strong>de</strong>phosphorylation, regulated by microRNA and RhoA. These<br />

interactions, their effect on functions and global regulation by microRNAs provi<strong>de</strong> a<br />

platform of novel targets during cardiovascular diseases.<br />

13


Matthieu CARUEL : caruel@lms.polytechnique.fr<br />

Laboratoire <strong>de</strong> Mécanique <strong>de</strong>s Soli<strong>de</strong>s, Ecole Polytechnique, Orsay,<br />

France<br />

FAST FORCE RECOVERY IN SKELETAL MUSCLE<br />

Muscle contraction is an ATP driven process that causes relative sliding of actin and<br />

myosin filament. Sliding is at least partially due to the power stroke: a conformational<br />

change in the heads of myosin II , that are attached to actin. This conformational<br />

change, however, is the main player in the response of a myofibrlil submitted to a fast<br />

length change.<br />

In 1971, A.F Huxley and R.M Simmons [Huxley, A. and Simmons, R. (1971). Proposed<br />

mechanism of force generation in striated muscle. Nature , 233:533–538. ] proposed a mo<strong>de</strong>l that has<br />

been since that time the main reference regarding fast force recovery. They assumed<br />

that the head can be viewed as a particle in a 2 state energy landscape coupled with a<br />

linear elastic element. They also assumed that the rate constants of the power stroke<br />

<strong>de</strong>pend on the mechanical potential energy of the system and thus on the strain<br />

applied to the myofibrils.<br />

In this now classical framework, the tension response to a shortening step has two<br />

main components : 1) an instant elastic drop to an extreme value (T1) and 2) a quick<br />

recovery due to power-stroke in the attached heads that leads to a plateau in the<br />

force-time response (T2). The time scale of fast recovery is such that no attachement<strong>de</strong>tachement<br />

process can occur. The rate constant of the fast recovery was shown by<br />

Huxley and Simmons to have an exponential <strong>de</strong>pen<strong>de</strong>nce on the size of the step.<br />

Recently the mo<strong>de</strong>l of Huxley and Simmons has been reconsi<strong>de</strong>red in the paper of<br />

Marcucci and Truskinovsky (2009, submitted). They proposed a new mechanical<br />

mo<strong>de</strong>l of fast recovery which assumes a continuous energy landscape and which<br />

links the tension response to length step with solving a system of stochastic<br />

Langevin's equations. In this poster presentation, we discuss the new step in the<br />

<strong>de</strong>velopment of this mo<strong>de</strong>l. The <strong>de</strong>velopment consists in a systematic account of the<br />

passive elasticity of the filaments. We show that such augmentation of the mo<strong>de</strong>l leads<br />

unambiguously to two unusual conclusions:<br />

1) The rate of fast force recovery cannot <strong>de</strong>pend on the step size exponentially as<br />

it is wi<strong>de</strong>ly assumed<br />

2) The fast recovery is highly non symmetric with respect to shortening and<br />

stretching which suggests that the response in stretching cannot be explained<br />

as a reverse of the power-stroke as proposed, for instance, by G. Piazzesi et al<br />

[ Piazzesi, G., Linari, M., Reconditi, M., Vanzi, F., and Lombardi, V. (1997). Cross-bridge<br />

<strong>de</strong>tachment and attachment following a step stretch imposed on active single frog muscle fibres.<br />

JOURNAL OF PHYSIOLOGY-LONDON , 498(1):3–15. ].<br />

14


Leif DEHMELT : leif.<strong>de</strong>hmelt@mpi-dortmund.mpg.<strong>de</strong><br />

Max-Planck-<strong>Institut</strong>e for molecular Physiology and TU Dortmund,<br />

Germany<br />

MECHANISMS IN NEURITE INITIATION: DIRECT OBSERVATION OF<br />

MICROTUBULE PUSHING BY CORTICAL DYNEIN/DYNACTIN KOMPLEXES.<br />

In neuronal and non-neuronal cells, MAP2c <strong>de</strong>corated microtubule bundles are un<strong>de</strong>r<br />

the influence of a dynein-<strong>de</strong>pen<strong>de</strong>nt pushing force. If such microtubule bundles are<br />

free to move, they are rapidly transported through the cell. At the cell periphery, such<br />

bundles can exert an outward force that is opposed to an actin mediated inward<br />

contractile force. Changes in the balance of these forces can lead to the induction of<br />

new neurite-like cell protrusions. Here, we directly observed cortical dynein/dynactin<br />

complexes and motile microtubule bundles via wi<strong>de</strong>-field and TIRF microscopy.<br />

Cortical dynein/dynactin complexes are preferentially associated with microtubule<br />

bundles that come in close proximity with the plasma membrane in the TIRF field. We<br />

find that at least two different dynein/dynactin populations exist, that are either<br />

stationary or associated with the minus ends of motile microtubule bundles. Our<br />

results support a mo<strong>de</strong>l in which free dynein complexes are transported towards the<br />

microtubule bundle minus-end, while stationary, cortex associated complexes can<br />

push microtubules directionally with leading plus-ends, thereby focusing a dyneinmediated<br />

force to locally induce neurite-like cell protrusions.<br />

Olivier DESTAING : <strong>de</strong>staino@ujf-grenoble.fr<br />

CRI Inserm,La Tronche, France<br />

INTERACTIONS OF BETA1 AND BETA3 INTEGRINS IN THE CONTROL OF THE<br />

DYNAMICS OF INVADOPODIA.<br />

Invadopodia or podosomes are adhesion structures able to <strong>de</strong>gra<strong>de</strong> the extracellular<br />

matrix and are implicated in tissue invasion by mobile cells such as osteoclast or<br />

metastatic cells. Thus, these structures appear as an interface between actin<br />

polymerization, adhesion and membrane trafficking implicated in invasion. In or<strong>de</strong>r to<br />

explore the interaction between invadopodia and extracellular matrix, we investigate<br />

the functions of the integrin family, the class of adhesion receptors composed by<br />

heterodimers of noncovalently associated alpha and beta subunits. Previous works<br />

strongly suggested that beta3 integrins were the main adhesion receptors regulating<br />

these structures. Experiments on micropatterned substrates and TIRF microscopy<br />

revealed an original association between beta1 and beta3 integrins in invadopodia. To<br />

explore the specific functions of each of these class of integrins in invadopodia, we<br />

induced them in beta3 -/- cells or in beta1 conditionnal knock-out cells. Surprinsingly, it<br />

appeared that beta1 integrins, but not beta3, are essential to invadopodia formation.<br />

Using a reverse genetic approaches, we <strong>de</strong>termined the essential reisdues of the<br />

cytoplasmic domains of this class of integrin. In particular, we suggest that beta1<br />

activation is essential to recruit beta3 integrins and induce the formation of<br />

invadopodia. More surprinsingly, modulating beta1 integrins activity results in<br />

pertubation in the coupling between adhesion, actin remo<strong>de</strong>lling and the <strong>de</strong>gradation<br />

of the extracellular matrix. We proposed that invadopodia are the site of a complex<br />

cross talk between integrins and initiated by beta1 integrins.<br />

15


Leah EDELSTEIN-KESHET : keshet@math.ubc.ca<br />

Dept of Mathematics, University of British Columbia<br />

Vancouver, Canada<br />

The biochemical regulation of cell motility is a complex but fundamental process,<br />

without which cell polarization and directed motion would be impossible. We have<br />

explored aspects of eukaryotic motility regulation centered on the roles of small<br />

GTPases such as Cdc42, Rac, and Rho. We have shown the importance of the<br />

membrane-cytosolic cycling of such proteins in generating an internal “map” that<br />

<strong>de</strong>fines the front and the back of the cell, and that signals to the assembly of<br />

cytoskeletal elements that produce forces of protrusion at the front and retraction at<br />

the rear of the cell. I survey our mo<strong>de</strong>lling efforts in this presentation.<br />

Horatiu FANTANA : fantana@mpi-cbg.<strong>de</strong><br />

MPI of Molecular Cell Biology and Genetics, Dres<strong>de</strong>n, Germany<br />

TOWARDS MEASURING THE CENTERING FORCES ACTING ON THE MITOTIC<br />

SPINDLE IN THE C. ELEGANS EMBRYO.<br />

The mitotic spindle is a highly dynamic, microtubule-based structure, which is<br />

responsible for chromosome segregation and cleavage plane specification during cell<br />

division. At the beginning of mitosis, the spindle is stably positioned at the center of the<br />

cell and returns to the center if displaced by fluctuations.<br />

In this project, we want to measure the magnitu<strong>de</strong> of the restoring forces acting on the<br />

spindle in the one-cell C. elegans embryo by using magnetic tweezers. With our setup,<br />

we can apply calibrated forces of several 100 pN on 1-micron-diameter magnetic<br />

beads, which we introduce into the embryos by microinjection into the gonad of adult<br />

worms. To apply the force on the spindle, we push the beads against the spindle<br />

poles. Preliminary experiments indicate that the pole structure can withstand forces of<br />

at least a few tens of pN.<br />

Absolute force measurements will allow us to relate the mechanical properties of<br />

microtubule arrays in vivo with the properties of single molecules measured in vitro<br />

and to estimate the number of the force generators involved in centering, contributing<br />

to a better un<strong>de</strong>rstanding of the centering process.<br />

16


Sebastian FUERTHAUER : bastian@pks.mpg.<strong>de</strong><br />

Max Planck <strong>Institut</strong> für Physik komplexer Systeme, Dres<strong>de</strong>n, Germany<br />

ANTISYMMETRIC STRESS AND THE ROLE OF ANGULAR MOMENTUM<br />

CONSERVATION IN COMPLEX FLUIDS.<br />

The stress tensor of a Newtonian fluid is symmetric in the hydrodynamic limit.<br />

However, in complex fluids, such as nematic liquid crystals, the director field can exert<br />

a torque if it is locally rotated away from its undistorted configuration.<br />

This produces a reactive antisymmetric contribution to the stress tensor.<br />

Here, we provi<strong>de</strong> the <strong>de</strong>rivation of a hydrodynamic theory for a complex fluid based on<br />

i<strong>de</strong>ntifying the entropy production rate from the rate of change of the free energy.<br />

Analyzing the angular momentum balance, reveals that an additional dissipative<br />

contribution to the antisymmetric stress exists. We obtain an expression for the<br />

antisymmetric dissipative stress by expanding thermodynamic fluxes in terms of<br />

thermodynamic forces, which is crucial in un<strong>de</strong>rstanding the non-equilibrium dynamics<br />

of chiral complex fluids, such as the acto-myosin <strong>cytoskeleton</strong> or a fluid<br />

driven by beating cilia.<br />

17


Thomas GREVESSE : thomas.grevesse@gmail.com<br />

Laboratory InFluX Complex fluids and interfaces, Mons, Belgium<br />

RHEOLOGICAL BEHAVIOUR OF NERVE CELLS<br />

Following big head shocks (car crash, falls, blast, ,,,) the brain is severed, leading to<br />

dysfunctions called traumatic brain injury (TBI). It is well known that following TBI,<br />

neurons un<strong>de</strong>rgo mechanical damages, e.g. beading of axons, due to mechanical<br />

shearing and stress forces, un<strong>de</strong>rlying the importance of the mechanical properties of<br />

neurons. However how neurons behave and sustain mechanical loads is still poorly<br />

un<strong>de</strong>rstood. Therefore, in this work, we caracterized the mechanical properties of<br />

isolated neuronal cells in or<strong>de</strong>r to un<strong>de</strong>srtand and quantify their mechanical behaviour,<br />

Among other cells neurons show a very complex polarized structure with hundreds of<br />

processes (<strong>de</strong>ndrites and axons) arising from the cell body (the soma). These sub<br />

parts show very different structures and physiological properties. It was therefore<br />

necessary to be able to probe the mechanical properties of these sub parts<br />

in<strong>de</strong>pen<strong>de</strong>ntly of the others, hence looking at the sub cellular level. For this purpose<br />

we <strong>de</strong>velloped an experimental apparatus, the magnetic tweezers. Via a<br />

electromagnet filled with a sharpened ferromagnetic tip, we are able to apply local<br />

magnetic field gradient on micrometer sized (5 µm) paramagnetic beads fixed on<br />

nerve cells, hence applying forces to cells at the subcellular level.<br />

Using the magnetic tweezers apparatus we performed creep experiments on both the<br />

soma and neurites (<strong>de</strong>ndrites and axons) of PC 12 cells. We applied a constant force<br />

on a small time scale (~10 s) and observed the strain response of the neurites and the<br />

soma. At small time scales neurites exhibit a purely elastic response with Young<br />

modulus of 5 kPa whereas the cell body shows a viscoelastic solid behaviour with a<br />

rigidity of 1,5 kPa. These results show that when subjected to short impulses of great<br />

forces, the cell body can dissipate a part of the stress trough its viscous component.<br />

For the neurites however, all the stress is used to <strong>de</strong>form the neurite. This suggests<br />

that when subjected to short impulses of force, the neurite will un<strong>de</strong>rgo more damage<br />

and be more subject to rupture than the soma.This could bring some elements to<br />

explain the mechanical damage of axons during TBI.<br />

In future work we will probe the mechanical properties of structured networks of<br />

neurons and the influence of forces on nerve impulse transmission<br />

18


Philip GUTHARDT TORRES :<br />

p.guthardt.torres@tphys.uni-hei<strong>de</strong>lberg.<strong>de</strong><br />

Hei<strong>de</strong>lberg University, <strong>Institut</strong>e for Theoretical Physics, Hei<strong>de</strong>lberg,<br />

Germany<br />

FAILURE OF NETWORKS OF BIOMOLECULAR BONDS UNDER FORCE<br />

Structural stability of biological cells is provi<strong>de</strong>d by the <strong>cytoskeleton</strong>, a crosslinked<br />

polymer network that is mechanically connected to the environment through adhesion<br />

sites. Although often mo<strong>de</strong>led as static, the <strong>cytoskeleton</strong> is highly dynamic, with<br />

connections, so called bonds, continuously breaking and forming in time. In this<br />

contribution the <strong>cytoskeleton</strong> is mo<strong>de</strong>led as a two dimensional network consisting of N<br />

massless no<strong>de</strong>s and L massless links. The simplest assumption for the mechanical<br />

nature of a cytoskeletal link is a harmonic spring, but a more realistic mo<strong>de</strong>l is a cable,<br />

which represents the polymeric nature of the link. Cables differ from springs by the<br />

asymmetry of their force-extension curves. We analyze spring networks as well as<br />

cable networks.<br />

In or<strong>de</strong>r to introduce the possible rupture of links we adapt an earlier mo<strong>de</strong>l for the<br />

stochastic rupture dynamics of adhesion clusters. This leads to breaking of links with<br />

force-<strong>de</strong>pen<strong>de</strong>nt rates which is different from the usual approach to fracture in material<br />

science. The fracture of hard macroscopic materials, e.g. concrete, is often mo<strong>de</strong>led<br />

using spring networks with randomly distributed force thresholds for the network links<br />

(random spring mo<strong>de</strong>l).<br />

In our work, we first compare the equilibrium states for different mechanical mo<strong>de</strong>ls<br />

and network topologies. Then stochastic rupture with force-<strong>de</strong>pen<strong>de</strong>nt rates is used to<br />

compare the statistical properties of fracture obtained using this mo<strong>de</strong>l to those<br />

obtained using a random spring mo<strong>de</strong>l.<br />

19


Florian HUBER : florian.huber@uni-leipzig.<strong>de</strong><br />

University of Leipzig, Germany<br />

LINKER INDUCED ACTIN NETWORK FORMATION UNDER CELL-SIZED<br />

CONFINEMENT<br />

Cross-linked actin networks are <strong>de</strong>cisively involved in the overall mechanical<br />

properties of cells. The networks’ architecture ranges from <strong>de</strong>nsely packed bundles to<br />

networks with high crossing angles and is typically assigned to specific linker proteins.<br />

Recently, however, it was found that several cross-linkers give rise to both exten<strong>de</strong>d<br />

networks and bundles. We used multivalent ions as mo<strong>de</strong>l-linkers to study actin<br />

filament aggregation in cell-sized geometries. Small droplets close to cellular sizes<br />

were filled with actin filaments. We slowly increased multivalent ion concentration and<br />

at a critical threshold the ions’ potential turns attractive. This implies a phase transition<br />

from isotropic or nematic f-actin solutions to cross-linked actin networks.<br />

In addition to the well known bundle formation, we obtained regularly spaced networks<br />

of star-like astern pattern. These networks display many features of cellular networks<br />

in the actin cortex and may serve as a mo<strong>de</strong>l system for the cortical actin layer.<br />

Observed phase transitions are fast (seconds to few minutes) which is of high interest<br />

concerning the known ability of living cells to quickly modify their morphology.<br />

Robiya JOSEPH : rubsjo.jmj@gmail.com<br />

CEDAD-IMPRS, <strong>Institut</strong>e of Immunology, Munster, Germany<br />

THE INTRACELLULAR ROLE OF MRP8 AND MRP14 IN CELLULAR DYNAMICS<br />

OF PHAGOCYTES<br />

The migratory properties of phagocytes allow their rapid accumulation at sites of injury<br />

and infection. The complex transmigration processes are not fully un<strong>de</strong>rstood,<br />

especially the ability of phagocytes to rapidly reorganize their <strong>cytoskeleton</strong>. Calciumbinding<br />

proteins are key elements in these signal transduction processes and we<br />

investigated the role of the two phagocyte specific calcium-binding proteins S100A8<br />

(MRP8) and S100A9 (MRP14). Previously we could already show, that in activated<br />

monocytes complexes of S100A8/S100A9 co-localize with microtubules (MTs) in a<br />

calcium and phosphorylation <strong>de</strong>pen<strong>de</strong>nt manner. Furthermore, phagocytes from<br />

S100A9 <strong>de</strong>ficient mice show altered migratory properties compared to wild-type cells.<br />

To investigate these in more <strong>de</strong>tail we established stable transfected HEK293 cell<br />

lines, which express S100A8/S100A9, S100A8, S100A9, S100A8/S100A9(N69A)<br />

(unable to form tetramers) and S100A8/S100A9(T113A) (mutated phosphorylation<br />

site) and <strong>de</strong>termined the individual contributions of the subunits and mutations to<br />

cellular dynamics. Using S100A8/S100A9 expressing HEK cells we found a similar<br />

pattern of co-localization of the complex with microtubules comparable to monocytes.<br />

Remarkable differences in S100A8/S100A9 transfected HEK cells compared to mock<br />

transfected ones were seen during breakdown and rebuilding of the microtubule based<br />

<strong>cytoskeleton</strong> indicating that S100A8/S100A9 affects the dynamics of MTs. Cells from<br />

S100A9 <strong>de</strong>ficient mice show altered characteristics in migration rates. Investigating<br />

this in more <strong>de</strong>tail we found that S100A9 knockout cells are more adherent than wildtype<br />

cells. A similar pattern has also been observed for S100A8/S100A9 transfected<br />

and non-transfected HEK cells. These effects seem to be mediated by S100A8 but not<br />

by S100A9. Furthermore, the effects observed are possibly <strong>de</strong>pen<strong>de</strong>nt on<br />

phosphorylation of S100A9. Our results provi<strong>de</strong> evi<strong>de</strong>nce, that the S100A8/S100A9<br />

complexes fulfil a pivotal role in remo<strong>de</strong>lling cytoskeletal structures necessary for the<br />

migration of leukocytes.<br />

20


Svenja-Marei KALISCH : s.kalisch@amolf.nl<br />

Group Bio-Assembly and Organisation, FOM-<strong>Institut</strong>e AMOLF<br />

Amsterdam, Netherlands<br />

AGAINST THE WALL: MICROTUBULE DYNAMICS IS REGULATED BY FORCE<br />

AND END-BINDING PROTEINS<br />

The dynamics and organization of microtubules (MTs) are essential for cell<br />

morphogenesis. Microtubule plus-end dynamics is highly regulated by specific endbinding<br />

and end-tracking proteins, as well as by the forces generated when MTs grow<br />

against obstacles such as the cell cortex. However, the molecular mechanisms<br />

un<strong>de</strong>rlying the regulatory actions of end-binding proteins, especially un<strong>de</strong>r force,<br />

remain largely unknown. We therefore investigate the force-induced behavior of<br />

dynamic MTs in the presence of the fission yeast proteins Mal3, Tea2, and Tip1 in<br />

vitro. Head-on forces are achieved by growing the MTs against a micro-fabricated rigid<br />

barrier. Using TIRF microscopy we show that the protein complex in general enhances<br />

the MT growth speed and induces catastrophes. Moreover, when coming in contact<br />

with the barrier, the force experienced by the MT shortens the catastrophe time further<br />

by nearly an or<strong>de</strong>r of magnitu<strong>de</strong>. We use this experiment to investigate whether the<br />

presence of end-binding proteins qualitatively or quantitatively changes the way MT<br />

dynamics responds to force. This should allow us to improve our insight into the<br />

molecular <strong>de</strong>tails of (force-enhanced) MT regulation by end-binding proteins.<br />

Nada KHALIFAT : Nada.Khalifat@univ-montp2.fr<br />

LCVN, UMR 5587, cc 26, Université Montpellier II, France<br />

ACTIVATION AND MULTIMER FORMATION OF EZRIN INDUCED BY PIP2<br />

Ezrin protein acts as a crosslinker between the actin network near the cell surface and<br />

the cytoplasmic leaflet of the plasma membrane, and has been shown to exist in a<br />

dormant conformation in the cytoplasm. In its active conformation, the N-terminal<br />

FERM domain binds to the plasma membrane, and the C-terminal region binds to<br />

actin filaments. Transition of ezrin to its active conformation occurs by binding to a<br />

specific lipid, Phosphatidylinositol-4,5-biphosphate (PIP2).<br />

PIP2 is a minor but key phospholipid involved in the regulation of a wi<strong>de</strong> variety of<br />

processes such as membrane trafficking exocytosis and endocytosis. In this current<br />

work, we investigate the interaction of wild-type ezrin or a mutant known to not bind<br />

the membrane, and PIP2 in solution and in lipid bilayer membrane. We i<strong>de</strong>ntified the<br />

specifity of PIP2: (i) on the conformational transition of ezrin and the importance of<br />

PIP2 alkyl chain in this process; (ii) on the formation of wild-type ezrin multimers.<br />

21


Elena KREMNEVA : kremneva@mappi.helsinki.fi<br />

Pekka Lappalainen’s laboratory, <strong>Institut</strong>e of Biotechnology, Helsinki,<br />

Finland<br />

DISTINCT ROLES OF ADF/COFILIN ISOFORMS IN SARCOMERE ASSEMBLY<br />

AND MATURATION<br />

The actin <strong>cytoskeleton</strong> is necessary for a large number of cell biological processes,<br />

including morphogenesis, motility, endocytosis, cytokinesis and signal transduction.<br />

The structure and dynamics of the actin <strong>cytoskeleton</strong> are both spatially and temporally<br />

regulated by an array of proteins that interact with actin filaments and/or monomeric<br />

actin. The actin <strong>cytoskeleton</strong> in non-muscle cells is highly dynamic and actin filaments<br />

are thus constantly assembled and disassembled in response to various intracellular<br />

and extracellular signals. In contrast, the actin filaments in muscle cells are formed into<br />

well organized structures, sarcomeres, and are believed to be significantly less<br />

dynamic than the ones in other cell-types. However, many proteins promoting actin<br />

dynamics in non-muscle cells are expressed also in muscle cells. Thus, it is possible<br />

that the muscle-specific isoforms of these proteins promote actin dynamics differently<br />

from their non-muscle counterparts. ADF\cofilins are central actin-binding proteins that<br />

enhance turnover of actin filaments by severing and <strong>de</strong>polymerizing F-actin. This class<br />

of proteins consist of 3 different isoforms in mammals – cofilin-1, cofilin-2 and ADF.<br />

Our Western blot analysis reveald that cofilin-1 and cofilin-2 are expressed in cultured<br />

rat cardiomyocytes. Interestingly, the amount of cofilin-2 increases in cardiomyocytes<br />

during maturation, whereas level of cofilin-1 <strong>de</strong>creases. Furthermore, my preliminary<br />

studies using isoform specific antibodies revealed distinct localization patterns of these<br />

proteins. Cofilin 1 displays more diffuse distribution in sarcomeres compared to cofilin-<br />

2 which localizes to narrow stripes close to M- and Z-lines. Biochemical analyses<br />

revealed that cofilin-1 and cofilin-2 display different thermal stability and affinity to actin<br />

filaments in the presence and absence of tropomyosin. Together, these data suggest<br />

that cofilin-1 and cofilin-2 play different roles in regulation of actin dynamics in heart<br />

muscle cells.<br />

22


Dorothy KUIPERS : d.kuipers@ucl.ac.uk<br />

The UCL Ear <strong>Institut</strong>e & London Centre for Nanotechnology, London,<br />

UK<br />

CELL DEATH AND ACTIN DYNAMICS IN A MODEL EPITHELIUM<br />

Cell <strong>de</strong>ath within an epithelium could lead to loss of the barrier function and damage to<br />

surrounding tissues. Hence, epithelial cells have evolved mechanisms to preserve<br />

epithelial integrity. We are using a monolayer of MDCK cells to investigate the<br />

complex patterns of actin dynamics that occur at sites of cell <strong>de</strong>ath in a mo<strong>de</strong>l<br />

epithelium.<br />

Our imaging of MDCK cells stably-transfected with fluorescent actin reporters<br />

suggests that dying cells are internalised by the coordinated movement of surrounding<br />

cells. This coinci<strong>de</strong>s with the formation of an apical actin 'ring' which closes above the<br />

dying cell. In addition, lamellipodial crawling of surrounding cells closes the area<br />

basally, resulting in two distinct planes of actin enrichment. In or<strong>de</strong>r to <strong>de</strong>termine which<br />

cells contribute to the actin ring structure, we used mosaics of cells transfected with<br />

fluorescent actin reporter-variants. Analysis of 27 individual events from 4 different<br />

experiments always revealed actin rings when the dying cell was expressing a<br />

reporter. On the contrary, in 9 examples where the dying cell was not expressing a<br />

reporter a there was no clear evi<strong>de</strong>nce of actin ring formation in the surrounding cells.<br />

Opposing si<strong>de</strong>s of the actin ring contract towards one another with a speed of ~0.2<br />

microns/min, that remains surprisingly constant over the closure time of 15-30 min<br />

(n=18).<br />

Further live imaging with myosin reporters will reveal the role of myosin contraction in<br />

dying and surrounding cells. Mo<strong>de</strong>ling techniques will be employed to investigate the<br />

forces driving the epithelia repair and whether they are due to the dying or surrounding<br />

cells.<br />

23


Anne LEBAUDY : anne.lebaudy@ibgc.cnrs.fr<br />

IBGC-CNRS UMR5095, Université <strong>de</strong> Bor<strong>de</strong>aux 2, France<br />

ACTIN BODIES MOLECULAR ARCHITECTURE AND CELLULAR PROPERTIES<br />

Actively proliferating yeast cells display three F-actin containing structures: actin<br />

cables, which serve as tracks for the polarized transport of vesicles, organelles and<br />

mRNA; actin patches that are required for endocytosis; and an acto-myosin cytokinetic<br />

ring. A large set of conserved actin-binding proteins (ABPs) tightly orchestrates the<br />

formation and the maintenance of these highly dynamic F-actin containing structures.<br />

We have recently <strong>de</strong>scribed that quiescent yeast cells totally reorganize their<br />

actin <strong>cytoskeleton</strong> and assemble a specific F-actin containing structure that we have<br />

called Actin Bodies. Actin Bodies contain various ABPs, they are rather immobile and<br />

display a very slow actin filaments turn-over. Remarkably, Actin Bodies disassemble<br />

within seconds upon cells re-entry into the proliferation cycle.<br />

We now are interested in <strong>de</strong>ciphering Actin Bodies molecular architecture. For<br />

that purpose, we are using live cell imaging to follow Actin Bodies formation and<br />

disassembly in quiescent cells. We are also studying more in <strong>de</strong>pth the turn-over of<br />

the filaments that are embed<strong>de</strong>d into this specific structure. Finally, we are trying to<br />

<strong>de</strong>cipher the role of some specific ABPs in the formation, maintenance and<br />

dissociation of Actin Bodies. All together our data strongly suggest that Actin Bodies<br />

are just not an aggregate of actin filaments but a rather very organized structure that is<br />

very sensitive to environmental conditions.<br />

Jimmy Le DIGABEL : j<br />

jimmy.ledigabel@univ-paris-di<strong>de</strong>rot.fr<br />

Matière et Systèmes Complexes, Benoit Ladoux, Paris, France<br />

ACTIVE SUBSTRATES TO STUDY MECHANOTRANSDUCTION<br />

Cellular processes imply an important coordination of interactions with the extracellular<br />

medium. Accumulating evi<strong>de</strong>nces <strong>de</strong>monstrate that cell functions can be modulated by<br />

physical factors such as the mechanical forces acting on the cells and the extracellular<br />

matrix, as well as the topography or rigidity of the matrix. These extracellular signals<br />

can be sensed by mechanosensors on the cell surface or in the cell interior to induce<br />

various cell responses. We have <strong>de</strong>veloped an original approach based on microfabricated<br />

substrates of PolyDimethylSiloxane (PDMS) to study cell migration. We<br />

used a closely spaced array of flexible micropillars (diameter~1µm) to map the forces<br />

exerted by cells on their substrates. In this case, the micropillars act as passive force<br />

sensors. Here, we propose to analyze the cell response to an external applied stress<br />

by a well-controlled actuation of the substrate. To do so, we used magnetic pillars.<br />

Such substrates allow us to modify dynamic adhesion conditions of cells and to better<br />

un<strong>de</strong>rstand the coupling phenomena between mechanical sensing and biochemical<br />

activity of a living cell. Using polyacrylami<strong>de</strong> hydrogels doped with ferromagnetic iron<br />

oxi<strong>de</strong> particles or ferrofluids, we can make magnetic pillars with diameters of 5 to 10<br />

microns while a magnetic field can be locally applied with a magnetic needle. Such a<br />

technique will be helpful to study the mechanical response of cells to an external force<br />

or to local changes in their microenvironment.<br />

24


Chiu Fan LEE : cflee@pks.mpg.<strong>de</strong><br />

Max-Planck-<strong>Institut</strong>e for the Physics of Complex Systems, Dres<strong>de</strong>n,<br />

Germany<br />

PROTEIN AMYLOID SELF-ASSEMBLY<br />

Amyloids are insoluble fibrous protein aggregations stabilized by a network of<br />

hydrogen bonds and hydrophobic interactions and they are intimately related to many<br />

neuro<strong>de</strong>generative diseases such as the Alzheimer’s Disease, the Parkinson Disease<br />

and other prion diseases. Better characterization of the various properties of amyloid<br />

fibrils is therefore of high importance for the un<strong>de</strong>rstanding of the associated<br />

pathogenesis. Here, we present our recent results on various equilibrium and<br />

dynamical properties of protein amyloid self-assembly. In particular, we discuss the<br />

length distribution of amyloid fibrils in thermal equilibrium [1], the possibility of<br />

isotropic-nematic phase transition as monomer concentration is increased [2], and the<br />

dynamical processes of nucleation and fibril elongation [3,4]. Our methods of<br />

investigation consist of techniques in statistical mechanics and molecular dynamics<br />

simulations.<br />

References: [1] Lee (2009) Self-assembly of protein amyloid: a competition between<br />

amorphous and or<strong>de</strong>red aggregation. Phys. Rev. E 80, 031922. [2] Lee (2009)<br />

Isotropic-nematic phase transition in amyloid fibrilization. Phys. Rev. E 80, 031902. [3]<br />

Jean, Lee, Lee, Shaw and Vaux (2010) Competing discrete interfacial effects are<br />

critical for amyloidogenesis. To appear in the FASEB J. [4] Lee, Loken, Jean and Vaux<br />

(2009) Elongation dynamics of amyloid fibrils: a rugged energy landscape picture.<br />

Phys. Rev. E 80, 041906.<br />

Jonathan LEE TIN WAH :<br />

Jonathan.Lee-Tin-Wah@curie.fr<br />

Laboratoire Physico-Chimie Curie, UMR168, Paris, France<br />

COLLECTIONS OF MOLECULAR MOTORS UNDER ELASTIC LOADING<br />

The goal of this project will be to study the collective mechanical properties of a small<br />

group of molecular motors un<strong>de</strong>r elastic loading in a simple in-vitro system. A similar<br />

situation can be found in various biological systems, leading, both in-vivo and in-vitro<br />

to spontaneous oscillations. It has already been observed that the mechanosensitive<br />

hair cells of the inner ear are able to oscillate spontaneously <strong>de</strong>pending on the<br />

actomyosin protein complex. We will attempt to shed light on the mechanism behind<br />

the oscillatory activity of the acto-myosin system, in particular by <strong>de</strong>termining the<br />

parameters that control their frequency and amplitu<strong>de</strong> through a number of mechanical<br />

and biochemical means in-vitro. The main objective of this thesis will be to measure<br />

the dynamical responsiveness of molecular motors working in groups to external<br />

stimuli. An important challenge will be to <strong>de</strong>termine conditions un<strong>de</strong>r which motor<br />

assemblies can operate at timescales compatible with auditory frequencies ( > 1KHz )<br />

which are much shorter than the ATPase cycle of a single motor. To achieve this, we<br />

plan to make use of conventional and an unconventional myosins, in particular Myosin<br />

1c which is regulated by calcium and is involved in the oscillation of hair cells.<br />

25


Alexandre LEWALLE : a.lewalle@ucl.ac.uk<br />

London Centre for Nanotechnology, UK<br />

NEUTROPHIL MOTILITY IN CONFINED ENVIRONMENTS<br />

There is evi<strong>de</strong>nce that the <strong>de</strong>gree of confinement of a motile cell plays a role in<br />

<strong>de</strong>termining how the cell produces the forces that allow it to move. Traditionally, cell<br />

motility is studied on fibronectin-coated surfaces, to which a cell can adhere via focal<br />

contacts. These focal complexes act as anchor points that provi<strong>de</strong> a mechanical<br />

coupling between the cell and its environment. Motility is achieved when forces,<br />

generated within the cell, are exerted on the surface via the focal adhesions. However,<br />

it appears that motility is possible in the absence of focal adhesions when the cell is<br />

placed in a more confined geometry, e.g., (1) in the two-dimensional environment<br />

between a glass surface and a soft agarose gel, or (2) in narrow one-dimensional<br />

channels fabricated from the polymer pdms. In both cases, surfaces are passivated to<br />

prevent the formation of focal adhesions at the cell membrane.<br />

We examine actin dynamics in HL60 cells un<strong>de</strong>rgoing chemotaxis in these different<br />

confined environments. The cells contain actin that is doubly labelled: with RFP and<br />

with photo-activatable GFP (PAGFP), which becomes visible once excited with a burst<br />

of violet light applied to a small region of the cell. We monitor the time evolution, in a<br />

moving cell, of the RFP and of the PAGFP intensities simultaneously near the cell’s<br />

leading edge, with a view to measuring the actin turnover in this region, and<br />

un<strong>de</strong>rstanding the effect of confinement on actin-mediated force generation.<br />

Ofélia MANITI : ofemaniti@yahoo.com<br />

LMGP, CNRS-UMR 5628, Grenoble, France<br />

EZRIN INTERACTIONS WITH BIOMIMETIC VESICLES CONTAINING<br />

PHOSPHATIDYLINOSITOL (4,5) BISPHOSPHATE (PIP2)<br />

The plasma membrane-<strong>cytoskeleton</strong> interface is a dynamic structure, participating in a<br />

variety of cellular events including cell shape, polarization and motility. Among the<br />

proteins involved in the direct linkage between components of the <strong>cytoskeleton</strong> and<br />

the plasma membrane is the ezrin/radixin/moesin (ERM) family. The FERM domain in<br />

their N-terminus contains a phosphatidylinositol 4,5 bisphosphate (PIP2) binding site<br />

responsible for membrane-binding whereas their C-terminus bind actin. In this work,<br />

our aim was to characterize the interaction of ezrin with large unilamellar vesicles<br />

(LUVs) containing PIP2 and with giant unilamellar vesicles (GUVs) containing PIP2.<br />

We first synthesized human recombinant ezrin bearing a cysteine residue at its C-<br />

terminus for subsequent grafting with Alexa488 maleimi<strong>de</strong>. This allowed us to perform<br />

fluorescence spectroscopy and microscopy experiments. The<br />

functionality of labeled ezrin was checked by comparison with that of wild type (WT)<br />

ezrin. The affinity constant between ezrin and LUVs, <strong>de</strong>termined by co-sedimentation<br />

assays and fluorescence correlation spectroscopy, was found to be ~5 µM for PIP2-<br />

LUVs and 20 to 70 lower for PS-LUVs, <strong>de</strong>pending of the experimental technique. We<br />

found that the interaction is not cooperative for PIP2-LUVs. We prepared fluorescently<br />

labelled GUVs using PIP2 analogues used as tracers (0.1% of total lipids) to<br />

investigate ezrin/GUVs interactions by fluorescence microscopy. Zeta potential<br />

measurements confirmed that the effective incorporation of PIP2 in the GUVs. Finally,<br />

the interaction of ezrin with PIP2-containing GUVs was investigated. Using either<br />

labeled ezrin and unlabeled GUVs or both labeled ezrin and GUVs,<br />

we evi<strong>de</strong>nced that clusters containing both PIP2 and proteins are formed.<br />

26


Olga MARKOVA : markova@ibdm.univ-mrs.fr<br />

IBDML – UMR 6216, <strong>Institut</strong>e of Developmental Biology Marseilles<br />

France<br />

HYDRODYNAMIC DESCRIPTION OF TISSUE ELONGATION DURING<br />

DROSOPHILA EMBRYO MORPHOGENESIS: THE ROLE OF ACTIVE<br />

ANISOTROPIC CYTOSKELETAL STRESSES<br />

Olga Markova and Pierre-François Lenne<br />

<strong>Institut</strong>e of Developmental Biology Marseilles‐Luminy (IBDML), 13288 Marseille, France<br />

During tissue morphogenesis, cells are able to change shape and rearrange, yielding<br />

dramatic changes such as tissue elongation. In early Drosophila embryos, the<br />

elongation of the germ band (GBE) results from cell intercalation, a process driven by<br />

tensile actomyosin networks which generate anisotropic forces (Bertet et al. 2004,<br />

Rauze et al. 2008). In an attempt to <strong>de</strong>scribe quantitatively the dynamics of GBE, we<br />

used the hydrodynamics theory of viscous fluids (Bittig et al. 2008). We find that during<br />

the fast phase of GBE, cells flow like viscous fluids driven by internal anisotropic active<br />

stresses generated by actomyosin anisotropic contraction. From quantitative analysis<br />

of vector fields of cell motion we estimate the viscosity of the Drosophila germband.<br />

Our work proposes a non-invasive method - based on the analysis of kinetics of tissue<br />

movement in vivo - to <strong>de</strong>termine tissue mechanical properties.<br />

Bertet, C., Sulak, L. & Lecuit, T., 2004. Myosin-<strong>de</strong>pen<strong>de</strong>nt junction remo<strong>de</strong>lling controls<br />

planar cell intercalation and axis elongation. Nature, 429(6992), 667-671.<br />

Bittig, T., Wartlick, O., Kicheva, A., González-Gaitán, M., Jülicher, F. 2008. Dynamics<br />

of anisotropic tissue growth. NEW JOURNAL OF PHYSICS, 10.<br />

Rauzi, M., Verant, P., Lecuit, T., Lenne, PF. 2008. Nature and anisotropy of cortical<br />

forces orienting Drosophila tissue morphogenesis. Nature Cell Biology, 10(12), 1401-1410.<br />

27


Carolina MARTINEZ CINGOLANI : cmartinezci@unal.edu.co<br />

CURIE INSTITUT INSERM U 932, Paris, France<br />

PHD DIRECTOR: VASSILI SOUMELIS<br />

Lab Director: Sebastian AMIGORENA<br />

Inserm U932 « Immunité et cancer ». <strong>Institut</strong> Curie<br />

MOLECULAR MECHANISMS AND TUMORAL MICROENVIRONMENT ROLE IN HUMAN<br />

DENDRITIC CELL MIGRATION IN CONFINED SPACE<br />

Dendritic cells (DCs) have an essential role due to their participation in the innate and<br />

acquired immune system responses. The DCs that resi<strong>de</strong> in the peripheral tissues must be<br />

able to leave this environment and to migrate to secondary lymphoid organs, where they<br />

exert their function as professional antigen presenting cells. (Banchereau et al, 2000). To do<br />

so, they have to pass through very narrow spaces, such as the tight epithelial intercellular<br />

junctions, the extracellular matrix, the basal membrane and the endothelium. The velocity of<br />

the movement must be optimal, because the DCs must induce transitory <strong>de</strong>formations of the<br />

microenvironment, without leaving collateral lesions, and at the same time, they must sense<br />

and integrate the signals around them. DCs have to be able to maintain their sentinel and<br />

phagocyte functions in the most efficient way (Faure-André, 2008). This migration occurs in<br />

a three-dimensional microenvironment (confined microenvironment). Its direction is<br />

<strong>de</strong>termined mainly by chemokine gradients (Rossi, and Zlotnik, 2000). However, there are<br />

other factors that may be implied in this process. In fact, some physical parameters, such as<br />

the tissue geometry, can affect the type of movement and the necessary conditions for the<br />

migration itself (Lämmermann, Ba<strong>de</strong>r, et al, 2008).<br />

Our recent findings show that DCs can be instructed to migrate by the inflamed tissues. This<br />

is, in particular, the case of Thymic Stromal Lymphopoietin (TSLP). This pro-inflammatory<br />

cytokine secreted by epithelial keratynocytes, is responsible for Langerhans cell <strong>de</strong>pletion<br />

that characterizes the Human Papilloma Virus infected tissues. This molecule was revealed<br />

as an inductor of DC <strong>cytoskeleton</strong> polarization and migration, in a chemokine-in<strong>de</strong>pen<strong>de</strong>nt<br />

and a myosin II-<strong>de</strong>pen<strong>de</strong>nt way (Fernan<strong>de</strong>z, et al, Immunity, submitted).<br />

Now, we intend to <strong>de</strong>termine which is the relative contribution of different molecular<br />

(chemokinesis, chemo-attraction) and physical parameters (tissue geometry, confinement<br />

and lymphatic flux) in the DC migration, both in physiological and pathological conditions<br />

with particular attention to cancer. It has been reported that reduced DC numbers and DC<br />

migration alterations occur in solid tumours (melanoma and uterine cervical intraepithelial<br />

neoplasia, Halliday, Le, et al 2001, Fernan<strong>de</strong>z, et al, submitted and Villablanca et al 2010).<br />

We want to evaluate the DC migration alterations in a microenvironment that favours the<br />

acquisition of migratory capacity by the non-immunological metastatic cells (Sahai, 2007).<br />

The relative contribution of different molecular and physical parameters will be <strong>de</strong>termined<br />

after DC treatment with TSLP and other migration stimulating and inhibiting factors. To this<br />

purpose we will use a micro-channel system <strong>de</strong>veloped by M. Piel ´s team at the Curie<br />

<strong>Institut</strong>e (Faure-André, 2008). The <strong>cytoskeleton</strong> rearrangements will be assessed by<br />

immunofluorescence for each condition using anti-actin, anti-tubulin and anti-myosin II<br />

antibodies. To evaluate the effects induced by the tumoral microenvironment we will use the<br />

supernatants <strong>de</strong>rived from both tumoral cell line and primary tumour cultures. The cytokine<br />

composition of these supernatants will be analysed ELISA and immunohistochemistry.<br />

This is a multi-disciplinar project that involves physics, human immunology, tumoral<br />

and cellular biology. We believe that our work will highlight the fundamental mechanisms<br />

used by DC to migrate. Our results could be useful for the establishment of novel therapeutic<br />

strategies to promote antitumoral immunity by favouring DC migration.<br />

28


Helen MATTHEWS : h.matthews@ucl.ac.uk<br />

MRC Laboratory for Molecular Cell Biology, London; UK<br />

When a cell enters mitosis, it dramatically rearranges its actin network to assume a<br />

stiff, roun<strong>de</strong>d shape. Recent work from our lab has shown that mitotic rounding is<br />

driven by ERM-induced cortical stiffening and is essential for proper spindle formation<br />

and successful mitosis (Kunda et al). Although much is known about the regulation of<br />

entry into mitosis by mitotic kinases, it has not been <strong>de</strong>termined how this regulation<br />

could feed into the actin <strong>cytoskeleton</strong>. We are interested in i<strong>de</strong>ntifying molecular<br />

regulators of mitotic rounding and, ultimately, un<strong>de</strong>rstanding how they generate the<br />

mechanical forces required to drive cell rounding.<br />

The spatial and temporal dynamics of mitotic cell rounding were analysed in two<br />

human cell lines, Hela and retinal pigment epithelial (RPE1) cells. Rounding begins<br />

early in mitosis, preceding nuclear envelope breakdown, and continues into<br />

metaphase. It consists of two steps; an initial <strong>de</strong>tachment from the substrate and<br />

retraction of lamellipodia, followed by the construction of a meshwork of cortical actin,<br />

during which stage phosphorylated ERM proteins accumulate at the membrane.<br />

Interestingly microtubules are not required for either stage as nocodozole-treated cells<br />

round normally. Myosin contractility contributes to mitotic rounding, with the inhibition<br />

of myosin/ROK/RhoA slowing the process, although cells do eventually achieve a<br />

roun<strong>de</strong>d shape.<br />

I am currently taking two approaches to i<strong>de</strong>ntify novel regulators of mitotic rounding.<br />

Firstly, I am using small molecule inhibitors of the mitotic kinases to try to i<strong>de</strong>ntify the<br />

kinase(s) driving shape change at mitosis. Secondly I am carrying out an RNAi screen<br />

using a targeted library of actin regulators, to i<strong>de</strong>ntify genes affecting mitotic cell<br />

shape. Here I will present some of the preliminary data from these analyses.<br />

Kunda et al (2008). Moesin controls cortical rigidity, cell rounding and spindle morphogenesis during<br />

mitosis. Current Biology 18(2), 91-101<br />

29


Manos MAVRAKIS : mavrakis@ibdm.univ-mrs.fr<br />

IBDML / CNRS UMR 6216, Marseille, France<br />

AN EMERGING ROLE OF SEPTINS IN TUNING ACTOMYOSIN CONTRACTILITY<br />

The formation of the primary epithelium in the Drosophila embryo involves a dramatic<br />

morphogenetic process during which plasma membrane invaginates between<br />

cortically-anchored nuclei to produce 6000 polarized epithelial cells within less than an<br />

hour. A set of proteins including F-actin, myosin-II, anillin and septins partition into the<br />

tips of the invaginating membrane front. This actomyosin-II network is maintained<br />

coinci<strong>de</strong>nt with the plasma membrane growth throughout cellularization, and its<br />

perturbation has been reported to disrupt cellularization. However, how the actomyosin<br />

network assembles, how it is stabilized and how its contractile properties are regulated<br />

in space and time so that it is coupled with invagination without premature constriction<br />

are largely unknown. The role of septins during this process is also entirely unclear.<br />

We imaged myosin-II dynamics during cellularization in embryos expressing functional<br />

myosin-II regulatory light chain–GFP. In wild-type embryos myosin-II is initially found<br />

as small myosin-II particles lining the invagination front. This distribution changes<br />

progressively into a continuous ring-like distribution, which is maintainted until the end<br />

of cellularization. When we treat embryos with forchlorfenuron, a drug which disrupts<br />

septin assembly, we observed that myosin-II prematurely constricts, and the<br />

invagination front closes off in an untimely fashion at the very onset of cellularization.<br />

We observed the same effect when we knocked down septin expression using RNAi.<br />

Increasing myosin-II activity with calyculin A, a myosin phosphatase inhibitor, leads<br />

also to an untimely hypercontractile ring phenotype. Our results suggest an interplay<br />

between septin organization and myosin-II activity and/or stabilization on F-actin.<br />

30


Tomas MAZEL : tomas.mazel@mpi-dortmund.mpg.<strong>de</strong><br />

Technische Universität Dortmund, Chemische Biologie, Germany<br />

CYTOSKELETON DYNAMICS DURING NEURITE INITIATION - MODELING<br />

APPROACH<br />

Tomáš Mazel, Anja Biesemann, Olga Müller, Leif Dehmelt*<br />

Technische Universität, Fakultät Chemie, D-44227 Dortmund, Germany<br />

e-mail: mazel@mpi-dortmund.mpg.<strong>de</strong><br />

Neurites enable targeted, long distance connections in the nervous system.<br />

Many signaling pathways are important for neurite <strong>de</strong>velopment, but little is known,<br />

how these signals are transformed into morphological features. Deeper un<strong>de</strong>rstanding<br />

of the un<strong>de</strong>rlying mechanisms could lead to new strategies in treatment of stroke,<br />

spinal cord injuries, or neuro<strong>de</strong>velopmental and neuro<strong>de</strong>generative diseases, in which<br />

neurite <strong>de</strong>velopment or regeneration is often compromised.<br />

In or<strong>de</strong>r to get an insight into these complex cellular mechanisms, we started to<br />

<strong>de</strong>velop an in silico mo<strong>de</strong>l of neurite initiation. This mo<strong>de</strong>l consists of several layers: 1)<br />

a regulatory network, including plasma membrane receptors and their downstream<br />

signaling casca<strong>de</strong>s, including Rho family GTPases. 2) effectors, controlled by this<br />

regulatory network, 3) the <strong>cytoskeleton</strong>, microtubuli and actin, which is controlled and<br />

rearranged by these effectors, 4) the cell bor<strong>de</strong>r, which is un<strong>de</strong>r the influence of forces<br />

generated by the <strong>cytoskeleton</strong>.<br />

We started with a highly simplified system, in which we can directly measure the<br />

behavior of many of the system components directly: the intracellular motility of<br />

MAP2c-induced microtubule bundles. By combining high-resolution TIRF microscopy<br />

with object tracking, we can estimate dynamic properties of microtubules and<br />

interacting motor complexes, which are then integrated into our mo<strong>de</strong>l system.<br />

Comparison of experimental observation with computer-based simulations is then<br />

used to i<strong>de</strong>ntify missing components or hid<strong>de</strong>n parameters in our mo<strong>de</strong>l. We will<br />

subsequently extend both our computational mo<strong>de</strong>l and our experimental observations<br />

to approach our goal of mo<strong>de</strong>ling neurite initiation in a more complex and more<br />

realistic cellular system.<br />

31


Xavier MEZANGES : xavier.mezanges@curie.fr<br />

<strong>Institut</strong> Curie UMR 168 Physicochimie, Paris, France<br />

SPERM CELL CRAWLING IN THE NEMATODE CAENORHABDITIS ELEGANS<br />

Cell motility is important in biological processes, such as the immune response,<br />

and in pathological states, such as cancer cell metastasis. Actin is implicated in most<br />

amoeboid cell movement, but Caenorhabditis elegans sperm cells lack actin, and their<br />

motility is driven by the Major Sperm Protein (MSP) <strong>cytoskeleton</strong>. Both MSP and actin<br />

form filament systems, but there is no biochemical or structural similarity between the<br />

two molecules. Little is known as to how MSP polymerization creates movement, or<br />

what biochemical partners are necessary for motility.<br />

In vitro systems, where cellular actin polymerization is recreated on the surface<br />

of a bead, giving actin comet tails and movement, have been useful in <strong>de</strong>fining the<br />

biochemical and physical parameters of actin-based motility. Our goal is to apply this<br />

technology to the MSP motility system. The first step is to i<strong>de</strong>ntify the activator(s) of<br />

MSP polymerization. The activator(s) appears to be a membrane-bound protein that<br />

contains a phosphorylated tyrosine residue in its active form. Our approach is to<br />

perform immunoprecipitation from sperm cell extracts with an anti-phosphotyrosine<br />

antibody, followed by mass spectroscopy to i<strong>de</strong>ntify the activator or activator complex.<br />

Once the activator(s) is in hand, we will find conditions for absorbing it to<br />

bead surfaces to produce MSP comets in sperm cell extracts. These comets will<br />

permit a characterization of MSP-based movement (speed, comet structure, comet<br />

elasticity etc). The comets will be further analyzed by mass spectrometry to i<strong>de</strong>ntify<br />

other components of the MSP motility system. In parallel, we will provi<strong>de</strong> a first<br />

characterization of MSP filaments formed in solution in vitro using TIRF and confocal<br />

microscopy, and microrheology and AFM to measure the elastic properties of MSP<br />

networks and single filaments.<br />

In the long term, the comparison of MSP and actin based movement should<br />

lead to a better un<strong>de</strong>rstanding of the fundamental principles cell motility.<br />

32


Francesca MILANESI :<br />

francesca.milanesi@ifom-ieo-campus.it<br />

Membrane and actin dynamics in the control of migratory and<br />

invasive strategies Laboratory, Milan, Italy<br />

MOLECULAR BASIS FOR THE DUAL FUNCTION OF EPS8 ON ACTIN<br />

DYNAMICS: BUNDLING AND CAPPING<br />

Actin capping and cross-linking proteins regulate the dynamics and architectures of<br />

different cellular structures by controlling the number of free actin growing ends and<br />

organizing filaments into higher or<strong>de</strong>r structures, respectively. EPS8 is the founding<br />

member of a unique family of capping proteins capable of binding and bundling actin<br />

filaments. The structural basis through which EPS8 binds actin filaments remains,<br />

however, unexplored. We combined biochemical and molecular approaches with<br />

electron microscopy image analysis, to dissect the molecular mechanism responsible<br />

for the distinct activities of Eps8. We propose that Eps8 caps by wrapping around<br />

filament ends, inserting its amphiphatic H1 helix into the hydrophobic pocket of the<br />

barbed end unit, acting as a clamp that prevents further addition of monomers.<br />

Whereas, actin crosslinking is primarily mediated by contacts between the filament<br />

and the Eps8 globular helical bundle. Mutantions in either the amphipatic helix or in<br />

the globular helical core of Eps8 permitted to dissect the capping and bundling<br />

activities, respectively, thus validating the mo<strong>de</strong>l, both in vitro and in vivo. Thus, Eps8<br />

controls actin-based motility through its capping activity, while, as a bundler, is<br />

essential for proper intestinal morphogenesis of <strong>de</strong>veloping Caenorhabditis elegans.<br />

Makito MIYAZAKI :<br />

miyazaki@chem.scphys.kyoto-u.ac.jp<br />

Department of Physics, Graduate School of Science, Kyoto University,<br />

Japan<br />

UNCOVERING THE HIDDEN STRUCTURE OF THE PROTEIN USING BAYESIAN<br />

INFERENCE<br />

In single-molecule experiments of proteins, the observable variables are restricted<br />

within a small fraction of the whole <strong>de</strong>grees of freedom in general.<br />

For instance, in the case of motor proteins, attaching a large probe particle to the<br />

molecule is a wi<strong>de</strong>ly adopted strategy. In this case, the motion of the probe is the only<br />

observable while the motion of the protein itself is completely hid<strong>de</strong>n.<br />

Therefore, in or<strong>de</strong>r to investigate the protein in <strong>de</strong>tail, we need to infer the internal<br />

structure of the protein (ex. shape of the Interaction potential between the motor<br />

protein and the “track”), only referring to the motion of accessible <strong>de</strong>grees of freedom.<br />

We formulated this problem on the basis of Bayesian framework, which can be<br />

applicable for various complex systems in non-equilibrium, and we obtained the simple<br />

and general evi<strong>de</strong>nce that this framework actually works. From careful numerical<br />

studies, although we confirmed that the performance of our method is better than the<br />

conventional method by or<strong>de</strong>rs of magnitu<strong>de</strong>, we found that there is a critical line in the<br />

original parameter space below which the precision of the estimate is abruptly lost.<br />

We will illustrate the basic feature of the proposed method including the property of<br />

“loss-of-precision transition” using a simple but nontrivial mo<strong>de</strong>l and we will propose<br />

the application for the single molecule experiments.<br />

33


Nikola OJKIC : nro207@lehigh.edu<br />

Physics Department, Bethlehem, USA<br />

Kinetics of self assembly of the contractile ring from a broad band of cortical no<strong>de</strong>s<br />

During cytokinesis of fission yeast, a contractile ring forms through the con<strong>de</strong>nsation of a<br />

broad band of ~ 65 cortical “no<strong>de</strong>s”. Each no<strong>de</strong> is a protein complex which contains myosin-<br />

II motors and formins. A mechanistic mo<strong>de</strong>l <strong>de</strong>scribing the aggregation of no<strong>de</strong>s into a ring<br />

is the search, capture, pull and release mo<strong>de</strong>l (Vavylonis et al. Science 97:319, 2008).<br />

According to this mo<strong>de</strong>l, formin polymerizes actin filaments in random directions along the<br />

cell cortex. These filaments establish transient connections among no<strong>de</strong>s and serve as links<br />

for myosin to exert the force required to pull the no<strong>de</strong>s towards one another. Simulations<br />

and experiments with mutant cells (Hachet, Simanis, Genes. Dev. 22, 2008) have<br />

suggested that no<strong>de</strong>s may form clumps instead of ring when the parameter values are<br />

different to those corresponding to wild type cells. To better un<strong>de</strong>rstand the conditions for<br />

successful ring assembly, we used scaling arguments, coarse grained stability analysis of<br />

homogeneous no<strong>de</strong> distributions, and Monte Carlo simulations. We found that initial uniform<br />

distribution of no<strong>de</strong>s is stable over short times due to randomness of connection among the<br />

no<strong>de</strong>s. Poisson fluctuations in the initial no<strong>de</strong> distribution lead to clump instabilities at long<br />

times. Uniform ring forms when the width of the broad band is less or approximately equal<br />

to the average length of actin filaments: in this case the lateral shrinking occurs faster than<br />

clump formation. We used scaling arguments to calculate the timescales for clump<br />

formation and for lateral shrinking. We used these theoretical results to <strong>de</strong>scribe clump<br />

formation process observed in the formin mutant cells.<br />

34


Didier PORTRAN : didierportran@aol.com<br />

IRTSV/LPCV , Grenoble, France<br />

STUDY OF DYNAMIC ASSEMBLY AND ORGANIZATION OF MICROTUBULE<br />

BUNDLE ARRAYS IN VITRO<br />

In eukaryotes, microtubule (MT) arrays provi<strong>de</strong> a molecular framework for<br />

various cellular processes including cell morphogenesis, establishment of cell polarity<br />

and cell division. Powering these cellular functions often hinges on the ability of the MT<br />

arrays to auto-organize in higher or<strong>de</strong>r structures. This is particularly true for noncentrosomal<br />

cells, such as plant cells, polarized epithelial cells, neural cells,.... where<br />

MT arrays do not emanate from a centrosome. Instead, they are mainly self-organized<br />

as MT bundles highly dispersed within the cell cortex. Thus, assembly of these cortical<br />

MT bundles is a key feature of cortical array organization and function in numerous<br />

cells. To un<strong>de</strong>rstand physical laws and molecular mechanisms that un<strong>de</strong>rlie the<br />

dynamic assembly that control the spatial organization of these MT arrays, we are<br />

going to reconstitute MT bundle arrays in minimal biochemical systems (with zippering<br />

MAPs such as Arabidopsis MAP65 members wich are homolog to PRC1 and Ase1p<br />

proteins respectively in mammals and in yeast). Observations of MT dynamic, bundle<br />

growth, bundle encountering… will use Total Internal Reflection Fluorescence<br />

Microscopy (TIRFM). The biomimetic assays will be further complexify by adding other<br />

molecules (stabilizing proteins such as CLASP and/or kinesins and/or +Tips proteins<br />

such as EB1). This biomimetic system will be further completed by controlling the<br />

spatial self-organization networks of MTs using micro- and nano-surface technic<br />

(micro-pattern) allowing spatial constrained architectures. The aim is to be able to<br />

<strong>de</strong>fine rules that govern MT behaviour when growing in specific environments or when<br />

encountering other MTs, and also to get insights into the links between MT bundle<br />

organization and physical constraints.<br />

Amsha PROAG : amsha.proag@univ-paris-di<strong>de</strong>rot.fr<br />

Laboratoire Matière et systèmes complexes UMR 7057, Paris, France<br />

MICROSTRUCTURED CHAMBERS TO STUDY THE SENSITIVITY OF CELLS<br />

TO THE GEOMETRY AND STIFFNESS OF THEIR ENVIRONMENT<br />

In or<strong>de</strong>r to study the behaviour of a living cell constrained in a tridimensional<br />

environment, we <strong>de</strong>signed microstructured substrates with controlled geometry and<br />

stiffness.Using microfabricated moulds, we produce arrays of elastomer and hydrogel<br />

microwells (poly(dimethylsiloxane), polyacrylami<strong>de</strong>, agarose), in which we can culture<br />

cells.The size of one well (typically from 20μm to 250μm in diameter) <strong>de</strong>termines the<br />

numberof cells per well and the extent of their spreading; its height restricts the<br />

number of possible layers. The shape of the well can be adapted to mo<strong>de</strong>l the cell<br />

physiological environment, <strong>de</strong>pending on the cell type, which makes it possible to<br />

examine in vitro the behaviour of cells in their natural geometrical configuration.<br />

In addition, the Young modulus of the substrate material can be tuned over several<br />

or<strong>de</strong>rs of magnitu<strong>de</strong> (from 0.1kPa to 1MPa): we may thus observe the response of<br />

cells to the substrate stiffness.<br />

We also investigate surface treatment protocols to enable protein grafting on the<br />

substrates before adding cells. We are currently <strong>de</strong>veloping a method for differential<br />

coating of the bottom surface, insi<strong>de</strong> walls and top surface of the microwells.<br />

35


Thomas PUJOL : thomas.pujol@espci.fr<br />

PMMH UMR 7636, ESPCI, Paris, France<br />

Mechanical characterization of actin gels by a magnetic colloids technique.<br />

The mechanism of the branched actin gel growing at the leading edge of moving cells<br />

is a major topic in biophysics. Un<strong>de</strong>rstanding the mechanical properties of actin<br />

networks can give insights into <strong>cytoskeleton</strong> rigidity and cell migration.<br />

We synthetize a branched actin network around colloids using the Arp2/3 protein<br />

machinery. The particles are super-paramagnetic and display an important dipole<br />

moment when an external magnetic field is applied. They attract each other via dipoledipole<br />

interaction and form chains. By increasing the magnetic field, we increase the<br />

force between the colloids and therefore apply a increasing stress on the actin gel<br />

between two beads. The gel <strong>de</strong>formation is measured optically by observing the<br />

distance between particles. We obtain stress-strain curves for actin networks, and<br />

measure its Young modulus. This technique can also be used as a micro-rheomoter<br />

by applying a sinusoidal stress with different frequency to obtain the dynamic modulus.<br />

As each pair of bead in a chain is a sensor in itself, this technique allows us to monitor<br />

a large number of gels and acquire convincing statistics.<br />

We plan to characterize different actin networks by varying the concentration of the<br />

capping and branching proteins and by using other actin binding proteins.<br />

Michal REICHMAN-FRIED : mreichm@uni-muenster.<strong>de</strong><br />

<strong>Institut</strong>e of Cell Biology, ZMBE, Munster, Germany<br />

MIGRATION OF PRIMORDIAL GERM CELLS IN ZEBRAFISH EMBRYSO<br />

The migration of Primordial Germ Cells (PGCs) in zebrafish relies on directional cues<br />

provi<strong>de</strong>d by the chemokine SDF1a and its receptor CXCR4b. These cells serves as an<br />

excellent mo<strong>de</strong>l system for un<strong>de</strong>rstanding long-range cell migration in <strong>de</strong>velopment<br />

and disease. Using time-lapse microscopy we have been able to monitor the<br />

behaviour of PGCs from early stages of <strong>de</strong>velopment to later stages when they have<br />

reached their target. We show that following their specification the cells un<strong>de</strong>rgo a<br />

series of morphological alterations that prece<strong>de</strong> the acquisition of motility and<br />

responsiveness to attractive cues. The cells then migrate towards their target while<br />

correcting their path following phases of loss of morphological cell polarity. In the<br />

following stages the cells gather at specific locations and move as cell clusters<br />

towards their final target. In all of these stages, zebrafish PGCs individually respond to<br />

alterations in the shape of the sdf-1a expression domain by directed migration towards<br />

their target, the somatic part of the gonad.<br />

To <strong>de</strong>termine the molecular basis for PGC polarization and migration we have followed<br />

the distribution of cytoskeletal elements in the migrating cells. Interestingly, unlike<br />

findings in some other cell types, this analysis did not support the i<strong>de</strong>a that actin<br />

polymerization propels cellular protrusions. Rather, we could <strong>de</strong>monstrate that<br />

zebrafish primordial germ cells generate bleb-like protrusions that are powered by<br />

cytoplasmic flow. Protrusions are formed at sites of higher levels of free calcium where<br />

activation of myosin contraction occurs. Separation of the acto-myosin cortex from the<br />

plasma membrane at these sites is followed by a flow of cytoplasm into the forming<br />

bleb. We propose that polarized activation of the receptor CXCR4 leads to a rise in<br />

free calcium that in turn activates myosin contraction in the part of the cell responding<br />

to higher levels of the ligand SDF-1. The biased formation of new protrusions in a<br />

particular region of the cell in response to SDF-1 <strong>de</strong>fines the leading edge and the<br />

direction of cell migration.<br />

36


Derek REVILL : phy5djr@leeds.ac.uk<br />

Contractility Group, Astbury Centre for Structural Molecular Biology,<br />

University of Leeds, UK<br />

MECHANICAL PROPERTIES OF MYOSIN LEVERS<br />

Our work studies the mechanical properties of myosin lever domains, using myosin 5<br />

as a mo<strong>de</strong>l system. Myosins are motor proteins that participate in key cellular<br />

functions including muscle contraction, intracellular cargo transport and cell migration.<br />

I<strong>de</strong>ntified by a motor domain that binds actin and hydrolyses ATP, myosins share the<br />

ability to translocate or move cargo along actin filaments. Their structure is<br />

characterised by three domains: motor, lever and tail. Mechanical properties of the<br />

lever are especially important in function. This domain, a single α-helix stabilised by<br />

light chains that bind IQ motifs, must be both rigid enough to rotate as a lever and<br />

generate force, whilst being flexible enough to accommodate functionally important<br />

strained conformations. Variety across the myosin family suggests evolution of lever<br />

structure to match mechanical function. Despite its functional importance, we lack a<br />

<strong>de</strong>tailed un<strong>de</strong>rstanding of the mechanical properties of myosin levers and how they<br />

<strong>de</strong>rive from internal substructure. Work is presented that combines negative stain<br />

electron microscopy and single-particle image processing to examine thermally-driven<br />

lever conformations of myosin 5 molecules. Preliminary results show averages with<br />

smoothly varying conformations, suggesting isotropic flexibility, but contrastingly, a<br />

smaller subset of conformations were also found with sharp bends in the lever<br />

indicating potential pliant points. A Metropolis Monte Carlo simulation technique was<br />

<strong>de</strong>veloped to validate mechanical lever mo<strong>de</strong>ls against EM image averages. Future<br />

plans are outlined to investigate the relationship between myosin lever mechanics and<br />

substructure using various experimental and analytical techniques.<br />

37


Anne-Cécile REYMAN : anne-cecile.reymann@cea.fr<br />

iRTSV-LPCV,Bat C2, pièce 227D, CEA Grenoble, France<br />

Physique du Cytosquelette et <strong>de</strong> la Morphogenese<br />

GEOMETRICAL CONTROL OF ACTIN NUCLEATION GOVERNS ACTIN<br />

NETWORK ARCHITECTURE<br />

Cellular architecture needs to perform highly complex mechanical transformations in<br />

or<strong>de</strong>r to achieve efficient morphogenesis, cell motility or any cell shape changes.<br />

Perpetual dynamics, organization, regulation or rapid reconstruction are only a few of<br />

the properties required for these morphological features which are supported by the<br />

actin <strong>cytoskeleton</strong>. To achieve their cellular functions, actin filaments can assemble<br />

into a large variety of super-structures The formation of these structures has been<br />

studied biochemically however, how the localization of the nucleation zones affects the<br />

structure of the actin networks is still an open question. Here we have combined UV<br />

based substrate micro-patterning techniques and in vitro actin polymerization to <strong>de</strong>sign<br />

a versatile and powerful biomimetic mo<strong>de</strong>l system allowing actin dynamics study at the<br />

cellular scale. Interestingly, actin polymerization occurs on this innovative biomimetic<br />

<strong>de</strong>vice with conventional biochemical properties. Significantly, actin filament networks<br />

assembled on these nucleating <strong>de</strong>vices adopt a collective and highly reproducible<br />

organization. Our study <strong>de</strong>monstrated that the spatial distribution of the boundary<br />

conditions of actin nucleation sites controls the dynamics and mechanics of actin<br />

assembly. Consequently, these boundary conditions give rise to specific network<br />

architectures and hence affect the force production location. Through extensive<br />

analysis of actin networks assembled on various geometries of nucleation zones, we<br />

conclu<strong>de</strong> that basic physical and probabilistic laws govern the spatial arrangements of<br />

anti-parallel and filopodia-like parallel filaments. Importantly, the geometry of different<br />

nucleation zones results in an entanglement of actin filaments into networks that can<br />

control their length<br />

Olena RIABININA : oriabinina@googlemail.com<br />

UCL Ear <strong>Institut</strong>e, London, UK<br />

TUNED BY LOVE: MATING SONGS AND EARS OF FRUIT FLIES.<br />

Drosophila melanogaster is an established mo<strong>de</strong>l species in the studies of genetic<br />

basis of sensory processing and behaviour. Here we investigate the components of<br />

the auditory communication in the flies of 9 Drosophila species. Auditory<br />

communication constitutes a crucial part of the flies mating ritual: while a male fly<br />

pursuits a female, he vibrates his wings in a characteristic manner to produce a<br />

species-specific “love song”. The frequency content of the songs differs greatly even<br />

between flies of closely related species. The spectral variability of the songs suggest<br />

corresponding differential tuning of the flies’ ears. Here we are bringing the signal and<br />

the receiver aspects of this story together. Employing the laser-Doppler vibrometry<br />

technique, we characterise the tuning of the antennal ears of the flies, and compare<br />

the peaks of the hearing sensitivity to the maxima in the males’ songs. We<br />

<strong>de</strong>monstrate, that the positive correlation holds between the characteristics of the ears<br />

of the awake, but not sedated, flies. In the sedated state, the active amplification of the<br />

hearing processes is likely to be affected. Thus, it is the active processes in the ears,<br />

rather than the passive characteristics like mass and size, that has been shaped<br />

during the evolution.<br />

38


Serge RINCON : Sergio.Rincon@curie.fr<br />

<strong>Institut</strong> Curie UMR 144, Anne Paoletti / Phong Tran Lab, Paris, France<br />

SPATIO-TEMPORAL CONTROL OF CELL DIVISION BY MEDIAL CORTICAL<br />

NODES IN FISSION YEAST CELLS.<br />

Schizosaccharomyces pombe is a unicellular eukaryotic organism whose easy<br />

genetics and reproducible cell shape make of it a good mo<strong>de</strong>l for studying cell polarity<br />

and division. Fission yeast cells are rod shaped, grow by tip extension and divi<strong>de</strong> at a<br />

constant length of about 14 μm by the formation of a contractile ring and septum in the<br />

middle of the cell. Recent work has revealed that cortical no<strong>de</strong>s organized at the<br />

medial cortex by the Wee1 regulatory kinase Cdr2 play a key role in the spatiotemporal<br />

control of cell division (Moseley et al. 2009; Almonacid et al. 2009; Martin et<br />

al. 2009). These no<strong>de</strong>s recruit the anillin-like protein Mid1 and influence the position of<br />

the division plane in parallel to Mid1 export to the nucleus. In addition, medial cortical<br />

no<strong>de</strong>s contain a second Wee1 regulatory kinase Cdr1 as well as a fraction of Wee1<br />

kinase. The distribution of medial cortical no<strong>de</strong>s and the activity of Cdr2 kinase are<br />

negatively regulated by Pom1 kinase. This polarity factor forms a gradient of<br />

concentration from the cell tips to the cell middle. As cells grow, Pom1 concentration in<br />

the medial region <strong>de</strong>creases, alleviating Cdr2 inhibition and promoting Wee1 inhibition<br />

and entry into mitosis.Therefore, Cdr2-mediated assembly of cortical no<strong>de</strong>s and their<br />

restricted localization to the medial cortex are key factors for the correct coordination<br />

of cell size and mitotic entry and for the <strong>de</strong>finition of the division plane. We are now<br />

investigating how Cdr2 associates with the cortex to promote cortical no<strong>de</strong>s assembly<br />

and how anchoring is inhibited by Pom1. These studies will help un<strong>de</strong>rstanding how<br />

the medial cortical region controlling cell division is <strong>de</strong>fined.<br />

Bibliography<br />

- Moseley, J. B., A. Mayeux, et al. (2009). "A spatial gradient coordinates cell size and mitotic<br />

entry in fission yeast." Nature 459(7248): 857-60.<br />

- Almonacid, M., J. B. Moseley, et al. (2009). "Spatial control of cytokinesis by Cdr2 kinase<br />

and Mid1/anillin nuclear export." Curr Biol 19(11): 961-6.<br />

- Martin, S. G. and M. Berthelot-Grosjean (2009). "Polar gradients of the DYRK-family kinase<br />

Pom1 couple cell length with the cell cycle." Nature 459(7248): 852-6.<br />

39


Jérémy ROLAND : roland.jeremy@gmail.com<br />

Laboratoire TIMC/IMAG, CNRS UMR 5525, La Tronche, France<br />

HOW FAR DO WE UNDERSTAND ADF/COFILIN-DEPENDENT ACTIN FILAMENT<br />

SEVERING?<br />

Direct visualization of ADF/cofilin-<strong>de</strong>corated F-actin shows that most actin filament<br />

severing events occur at the boundaries between <strong>de</strong>corated and non-<strong>de</strong>corated<br />

sections of actin filaments. Consistent with this behavior, bare actin filaments and<br />

those fully saturated with cofilin sever less easily than those partially saturated with<br />

cofilin. However, the mechanical basis for preferential filament fracture at boundaries<br />

has not been i<strong>de</strong>ntified. It has been hypothesized that severing arises from the intense<br />

and localized shearing of the filament due to an accumulation of stress at boundaries<br />

between ADF/cofilin-<strong>de</strong>corated and non-<strong>de</strong>corated segments.<br />

We have previously characterized the existence of twist-bend coupling in actin<br />

filaments and hypothesize that twist-bend coupling in actin filaments is a plausible<br />

candidate for converting bending filament undulation into stress accumulation at<br />

boundaries that generates filament severing.<br />

In the present work, we investigate the classical equations for elastic rods<br />

supplemented with twist-bend coupling terms. By simulating actin filament dynamics in<br />

different conditions (terminal load or twist; random forces) and in the presence or<br />

absence of ADF/cofilin bound to the filament, we <strong>de</strong>monstrated that twist-bend<br />

coupling is directly responsible for localized and important twist shear at boundaries of<br />

ADF/cofilin <strong>de</strong>corated domains. Additionally, more than half of the elastic energy<br />

<strong>de</strong>nsity is associated with twist-bend coupling. Finally, numerical simulations of<br />

filaments subjected to random forces provi<strong>de</strong> a coherent qualitative and qualitative<br />

<strong>de</strong>scription of the mechanical events leading to filament severing.<br />

In conclusion, we suggest that actin filaments are nanomachines that convert long<br />

range bending fluctuations into intense and localized twist shearing that contribute to a<br />

greater probability of filament severing. This energy processing, which consists of<br />

transforming elastic energy into work to break actin subunit bonds, is of crucial<br />

importance in the dynamics of complex filament architectures, such as the<br />

<strong>cytoskeleton</strong>.<br />

40


Florian RUCKERL : Florian.Ruckerl@curie.fr<br />

<strong>Institut</strong> Curie, CNRS, UMR 168, Paris, France<br />

DYNAMICS OF ACTIN ASSEMBLY PROMOTED BY FORMIN<br />

The shape and function of a cell is mainly <strong>de</strong>termined by its <strong>cytoskeleton</strong>, especially by<br />

the networks and bundles of polymerized actin. Force generation by actin<br />

polymerization is, together with the contraction by myosin motors, essential for cell<br />

motility. The branched actin networks are assembled by the Arp2/3 complex, and are<br />

already well un<strong>de</strong>rstood. Other structures like contractile rings and filapodia, which are<br />

both polarized actin bundles, are organized by the multi domain protein formin. It acts<br />

as a so-called leaky capper and remains bound to barbed end of the filament during<br />

the polymerization process.<br />

While the general mechanism of actin filament assembly by formin is known, the data<br />

on binding rates, potential <strong>de</strong>ad times and stall forces is still scarce.<br />

Using functionalized microbeads in a precise optical tweezer setup with high lateral<br />

and temporal resolution enables to measure the addition of single or multiple<br />

monomers to a bundle or filament. This allows to measure the rate and frequency of<br />

the assembly and gives insight to the molecular workings of the formin complex.<br />

Especially the stepsize is interesting, as it is unclear whether formin adds single<br />

monomers or pre assembled oligomers to the filament. Applying additional force to the<br />

fibre with the optical trap enables to measure the stall force . This is of particular<br />

interest, as the force created by formin mediated actin assembly might exceed the<br />

normal polymerization forces.<br />

41


Sara SADI : sara.sadi@wgi.su.se<br />

Department of Cell Biology, Stockholm University, Suè<strong>de</strong><br />

THE ROLE OF PROFILIN FOR SRF/MAL CONTROLLED GENE EXPRESSION<br />

Actin filaments form the basic structure of the force-generating microfilament<br />

system, which is essential for fundamental cellular processes such as cell motility,<br />

adhesion and cytokinesis. Profilin is a key regulator of actin polymerization; it binds to<br />

actin monomers and forms the profilin:actin (P:A) complex and <strong>de</strong>livers actin<br />

monomers to the growing (+)-end of the filament via nuclear promoting factors like<br />

Ena/Vasp and the formins. Profilin binds a number of other proteins and also lipids of<br />

the phosphatidylinositol family. It has been reported that both actin and profilin are<br />

present in the nucleus, however it is less clear to what extent the P:A complex is<br />

formed and functions in the nucleus.<br />

Actin is implicated in gene expression, e.g. the transcription factor SRF which<br />

controls a large number of genes, including many encoding microfilament associated<br />

proteins like actin itself and profilin, ‘senses’ cytoplasmic changes in actin dynamics<br />

through its co-activator MAL. MAL is an actin monomer binding protein un<strong>de</strong>r constant<br />

nucleocytoplasmatic shuttling. It competes with profilin for actin binding and upon<br />

serum stimulation it dissociates from actin and accumulates in the nucleus. (Vartiainen<br />

M).<br />

In this project I am studying the connection between SRF/MAL and actin<br />

dynamics with respect to the role of profilin and P:A and I have observed that downregulation<br />

of profilin expression using siRNA interferes with the nuclear accumulation<br />

of MAL fused to GFP. Furthermore, <strong>de</strong>pletion of profilin also represses SRF/MALcontrolled<br />

transcription as seen using a plasmid reporter system. It remains to be seen<br />

if this is a concequence of the competition between profilin and MAL for actin or if<br />

other mechanisms are involved. Other projects in my laboratory focus on microfilament<br />

organization and dynamics at the leading edge in non-muscle cells.<br />

42


Isabelle SAGOT : isabelle.sagot@ibgc.u-bor<strong>de</strong>aux2.fr<br />

IBGC-CNRS UMR5095, Université <strong>de</strong> Bor<strong>de</strong>aux , France<br />

ACTIN BODIES COMPOSTION: A COMPLEX PROBLEM<br />

Actin Bodies are F-actin containing structures that are specifically assembled in<br />

quiescent yeast cells. Actin Bodies are remarkably stable structures but disassemble<br />

within seconds upon cells re-entry into the proliferation cycle. Actin Bodies formation<br />

and molecular properties can result either to the specific expression in quiescent cells<br />

of one or more ABPs or can be due to the specific post-translational modification of<br />

one or more ABPs that lead to a change in its/theirs biochemical properties. In or<strong>de</strong>r to<br />

<strong>de</strong>cipher the molecular basis of Actin Bodies formation and maintenance, colocalization<br />

studies have been un<strong>de</strong>rtaken and have shown that specific Actin Binding<br />

Proteins are associated with these structures. Further genetic approaches have<br />

<strong>de</strong>monstrated that some of these ABPs are required for Actin Bodies formation and/or<br />

maintenance. However, these two approaches are not exhaustive and some critical<br />

components of the Actin Bodies could have been missed. Although the actin filaments<br />

that are embed<strong>de</strong>d in Actin Bodies have a rather slow turn-over in living cells, Actin<br />

Bodies are extremely sensitive structures and their purification turn out to be rather<br />

difficult. To circumvent this problem, we have un<strong>de</strong>rtaken two proteomic approaches;<br />

one based on 2D-DiGE and the other based on 2D BN / SDS – PAGE followed by<br />

Mass Spectromerty. Partial preliminary results show that in<strong>de</strong>ed, one ABP that colocalize<br />

with Actin Bodies and is required for their formation and/or maintenance is<br />

specifically modified in quiescent yeast cells. Our global approach may lead to the<br />

<strong>de</strong>ciphering of the complex interplay between ABPs that causes actin <strong>cytoskeleton</strong><br />

reorganization in quiescent cells.<br />

Carsten SCHULDT : schuldt@physik.uni-leipzig.<strong>de</strong><br />

University of Leipzig Faculty of Physics and Earth Science, UK<br />

DYNAMICS OF FORMIN PROMOTED ACTIN POLYMERIZATION<br />

In vivo the semiflexible polymer actin is organized preferentially in networks or<br />

bundles. These structures contribute to the <strong>cytoskeleton</strong>, whose inherent properties<br />

<strong>de</strong>termine the cell’s morphology, both mechanically and functionally, and facilitate<br />

motility via protrusions and contractions. The assembly of large and polarized<br />

cytoskeletal actin bundles (contractile ring, filopodia) far from thermodynamic<br />

equilibrium is driven by a multi-domain protein called formin. This ’leaky capper’ is<br />

known to remain bound to the growing barbed ends of filaments and is capable of<br />

accelerating the polymerization rate.<br />

We employ an optical tweezer setup in interaction with functionalized microbeads to<br />

measure formin’s stall force and step size in vitro. Determining the stall force will yield<br />

further insight into formin’s ability to produce forces from biochemical energy. In<br />

particular, formin may be able to overri<strong>de</strong> the force limit of normal actin polymerization.<br />

Measuring the step size may elucidate the assembly process itself. Does formin<br />

preform actin oligomers and insert them to the existing filament at once?<br />

The application of the sophisticated force clamp method seems to be an apropriate<br />

technique to measure step size and examine the behavior of formin with and without<br />

external applied tension.<br />

43


Rama SHESHKA : sheshka@lms.polytechnique.fr<br />

Laboratoire <strong>de</strong> mécanique <strong>de</strong>s soli<strong>de</strong>s, École polytechique, Orsay, France<br />

THE ROLE OF POWER STROKE IN MOLECULAR MOTOR MYOSIN II<br />

The motor protein such as myosin II produce directional motion and force by<br />

consuming the chemical energy stored in ATP. We try to un<strong>de</strong>rstand the mechanism<br />

used by molecular proteins to convert this chemical energy directly into the mechanical<br />

work. We propose the mathematical mo<strong>de</strong>l based on directional Brownian motion.<br />

The hydrolysis of ATP plays an essential role in creating the directed motion<br />

observed in motor proteins. Many experimental observations <strong>de</strong>monstrate the coupling<br />

between ATP hydrolysis and the force-displacement generation. The binding of ATP to<br />

the actin-myosin complex leads to rapid dissociation of the crossbridge from actin,<br />

then myosin hydrolyze ATP and forms a stable ADP-Pi complex, the myosin attaches<br />

to the actin filament. As ADP is realized, the motor protein passed in the rigor state in<br />

which it is locked in its position on the filament until one other molecule of ATP is<br />

bound. Binding cause a myosin- actin complex change its state so as to move the<br />

actin approximately 5 nm, it is the powerstroke.<br />

The attachment-<strong>de</strong>tachment process can be viewed as a mo<strong>de</strong>l of Brownian<br />

ratchets. The motor (myosin) rectify the randomly fluctuating stochastic forces using<br />

the periodic asymmetric potential which we mo<strong>de</strong>l the actin filament.<br />

The Power Stroke we presented entirely in mechanical framework by using a<br />

bistable element. We combine the bistable element with the motor based on brownian<br />

ratchets and also we try to explain and to reconstruct the Lymnn-Taylor cycle of<br />

muscle contraction with four states mo<strong>de</strong>l.<br />

We apply this mathematical framework in the case of cooperative motor for un<strong>de</strong>rstanding<br />

the experimental curves during isometric and isotonic contraction using the numerical<br />

computation of correspond system of Langevin equations<br />

44


Tomita Vasilica STIRBAT :<br />

tstirbat@lpmcn.univ-lyon1.fr<br />

Laboratoire <strong>de</strong> Physique <strong>de</strong> la Matière Con<strong>de</strong>nsée et Nanostructures,<br />

Lyon, France<br />

The interest in the rheological and mechanical characterization of embryonic<br />

cell aggregates is due to the fact that these “spheroids”, through their complex<br />

properties are proven to be mo<strong>de</strong>ls of embryonic tissues. Their fluid like behavior can<br />

help for the un<strong>de</strong>rstanding of the process of tissue organization in the fields of<br />

embryology, oncology and tissue engineering.<br />

The team is interested in studying the physical properties of aggregates by<br />

measurements of aggregate’s surface tension (using a compression tissue<br />

tensiometer based on the Laplace formula applied to compressed aggregates), by<br />

measurements of apparent viscosity (knowing the fusion time of two aggregates) and<br />

by measurements of the elastic, viscous and plastic timescales (aggregates<br />

compression-relaxation experiments).<br />

We now want to go further in the rheological characterization of these aggregates.<br />

The stress-strain relationship will be studied in <strong>de</strong>tails using a parallel-plate rheometer.<br />

Moreover, in or<strong>de</strong>r to test their potential not-Newtonian properties we plan to measure<br />

the velocity profiles of cells by forcing the aggregates to flow in microfluidic channels.<br />

Björn STUHRMANN : stuhrmann@amolf.nl<br />

FOM <strong>Institut</strong>e for Atomic and Molecular Physics , Amsterdam,<br />

NetherlandS<br />

BIOMIMETIC MODELLING OF CELLULAR MORPHOGENESIS<br />

Migration and division of living cells are ultimately generated by the coupled<br />

morphogenesis of the cell <strong>cytoskeleton</strong> and the plasma membrane. Despite<br />

impressive advances in the i<strong>de</strong>ntification of the cell molecular inventory, the un<strong>de</strong>rlying<br />

processes are still poorly un<strong>de</strong>rstood. We strive to discern biophysical principles of<br />

cytoskeletal and cell morphogenesis. To this end, we construct a biomimetic mo<strong>de</strong>l<br />

system of the <strong>cytoskeleton</strong> by confining to liposomes cross-linked actin biopolymers<br />

driven by the active processes of polymerization and motor sliding. The key innovation<br />

of this project lies in its systematic biomimetic approach alongsi<strong>de</strong> quantitative<br />

morphological and mechanical examination and theoretical mo<strong>de</strong>lling.<br />

45


Cristian SUAREZ : suarezcristian38@yahoo.fr<br />

iRTSV-LPCV,Bat C2, pièce 227D CEA Grenoble, France<br />

Physique du Cytosquelette et <strong>de</strong> la Morphogenese<br />

Cell motility, based on actin polymerisation, requires a tight regulation of the dynamics<br />

of Actin Binding Proteins (ABPs). ADF/cofilin is one of three ABPs that precisely<br />

choreograph actin polymerization and organization to generate “comet-tail” motility in<br />

vitro. ADF/cofilin severs actin filaments and then enhances disassembly of actin<br />

filaments. During actin polymerisation, hydrolysis of the nucleoti<strong>de</strong> (ATP to ADP-Pi)<br />

bound to actin subunits occurs rapidly (k - = 0.35 s -1 ) whereas Pi release is much more<br />

slower (k - = 0.0019 s -1 ). ADF/cofilin has a strong affinity only for ADP-actin, by<br />

consequence its binding is exclu<strong>de</strong>d from newly polymerised zone of an actin filament.<br />

We use TIRF (Total Internal Reflection Fluorescence) microscopy to follow in real time<br />

the association of fluorescently-labeled ADF/cofilin on growing actin filaments. Our<br />

results <strong>de</strong>monstrate that labeled ADF/cofilin is a marker of the nucleoti<strong>de</strong> state of actin<br />

filaments. We show also that fragmentation occurs near the interface between actin<br />

subunits free of ADF/cofilin or bound to it. Finally, FRAP experiments allow us to<br />

measure directly the dynamic of interaction of ADF/cofilin with actin filament.<br />

Olga SYTINA : sytina@amolf.nl<br />

Laboratory of Bio-organization, FOM <strong>Institut</strong>e AMOLF, Amsterdan,<br />

Netherlands<br />

MEASURING SINGLE MICROTUBULE DYNAMICS IN VITRO WITH NEAR<br />

MOLECULAR RESOLUTION.<br />

OLGA SYTINA, SVENJA-MAREI KALISCH AND MARILEEN DOGTEROM<br />

Microtubules are one of the main cytoskeletal polymers in all eukaryotic cells.<br />

The experimental method to study molecular mechanisms of microtubule (MT) selfassembly,<br />

force generation and regulation by MAPs (microtubule associated proteins)<br />

with nanometer resolution will be presented. The technique integrates optical<br />

tweezers with line optical trap coupled to the DIC high-resolution imaging microscope<br />

and microfluidic cell with incorporated microfabricated barriers. In the experiment a<br />

microbead with bound to it axoneme, serving as a nucleation site for MT growth, is<br />

trapped in the laser beam and can be directed against a rigid barrier. MT growth from<br />

axoneme is initiated by injection of the reaction mix and the displacement of the bead<br />

upon MT length change is monitored with nanometer resolution. The new special<br />

feature being introduced now is a force feedback system to un<strong>de</strong>rstand how MT<br />

dynamic properties change un<strong>de</strong>r constant load. Preliminary results of the MT growth<br />

against rigid barriers and shrinkage dynamics will be presented.<br />

46


Nessy TANIA : ntania@math.ubc.ca<br />

Department of Mathematics The University of British Columbia<br />

Vancouver, Canada<br />

A Mathematical Mo<strong>de</strong>l of Cell Motility Regulation by the Cofilin Pathway<br />

Directed cell movement is an integral part of a wi<strong>de</strong> range of physiological<br />

processes. Cofilin is an important regulator of the actin <strong>cytoskeleton</strong>. Recent<br />

experimental data from our collaborator, John Con<strong>de</strong>elis, on mammary<br />

carcinoma cells reveal that upon stimulation by epi<strong>de</strong>rmal growth factor<br />

(EGF), a pool of active cofilin is generated leading to the production of new<br />

barbed-ends of actin filaments. These fast growing barbed-ends lead to<br />

protrusive forces as the filaments extend. In carcinoma cells, cofilin thus plays<br />

a primary role in initiating cell protrusion and <strong>de</strong>termining cell direction. In this<br />

presentation, we discuss and show results from a mathematical mo<strong>de</strong>l of<br />

cofilin regulatory pathway and its response to stimulation by EGF. The mo<strong>de</strong>l<br />

consists of a set of differential equations <strong>de</strong>scribing the dynamics of various<br />

cofilin forms due to regulations by membrane lipids (PIP2), F-actin binding,<br />

and inactivation/phosphorylation by LIM kinase (LIMK). In addition, F-actin<br />

<strong>de</strong>nsity is tracked and cofilin-induced barbed end generation is quantified.<br />

Results from our mo<strong>de</strong>l capture the dynamics of barbed end generation upon<br />

the first minute of EGF stimulation as observed experimentally on mammary<br />

carcinoma cells. Interestingly, we find that increasing cofilin inactivation via<br />

LIMK promotes barbed end production. We show how this effect can be<br />

explained in terms of the resting level of the membrane-bound cofilin form.<br />

Hirokazu TANIMOTO :<br />

thirokazu@daisy.phys.s.u-tokyo.ac.jp<br />

Sano Laboratory, Department of Physics, Graduate School of Science,<br />

the University of Tokyo, Japan<br />

CELLULAR SHAPE, MOTION AND TRACTION FORCES ON VARIOUS ADHESIVE<br />

SURFACES<br />

Although many biophysical studies about cell migration have been done, the<br />

quantitative relationship between cellular shape, motion and its forces is still unclear.<br />

To elucidate the relationship, experiments with controlling physical conditions are<br />

nee<strong>de</strong>d.<br />

We used Dictyostelium cells as the mo<strong>de</strong>l of fast moving cells and controlled the cellsubstrate<br />

adhesion strength by changing collagen <strong>de</strong>nsity on the poly-acrylami<strong>de</strong> gels.<br />

With this setup, we measured cellular shape pattern, centroid motion and traction<br />

forces exerted to the substrate. The results are as follows.<br />

(1): Dictyostelium’s velocity takes its maximum value at the intermediate adhesion<br />

strength.<br />

(2): Cellular shape becomes or<strong>de</strong>red and oscillating in time with increasing adhesion<br />

strength.<br />

(3): The magnitu<strong>de</strong> of the traction forces becomes larger with increasing adhesion<br />

strength.<br />

In this poster session, we will show these results and discuss how cells<br />

coordinate their shapes, motions and forces in space and time.<br />

47


Ulrike THEISEN : U.Theisen@warwick.ac.uk<br />

Cytoskeletal Organization Centre for Mechanochemical Cell Biology<br />

Warwick Medical School, UK<br />

KIF1C, A KINESIN-3 FAMILY MEMBER, IS INVOLVED IN CELL MIGRATION,<br />

POLARITY AND FUSION<br />

Upon differentiation, myoblasts leave the cell cycle and change their morphology to a<br />

bipolar spindle shape before they fuse predominantly at their tips to form a muscle<br />

fiber. Microtubules are crucial in this process, forming a parallel array with the plus<br />

ends extending towards the tips of elongating cells. Maintenance of cell polarity and<br />

acquisition of fusion competence is therefore likely to involve kinesin-mediated<br />

transport of important, as yet unknown factors towards the cell tips.<br />

In an RNAi screen highly expressed Kinesins were <strong>de</strong>pleted in C2C12 cells, and<br />

monitored these cells for morphological <strong>de</strong>fects upon differentiation. In this assay, 10<br />

Kinesins were nee<strong>de</strong>d for proper cell elongation and subsequent cell-cell fusion.<br />

Among these is the poorly characterized Kinesin-3 family member Kif1c. In addition to<br />

causing <strong>de</strong>fects in cell elongation and fusion, Kif1c <strong>de</strong>pleted cells exhibit difficulties in<br />

establishing or maintaining cell polarity and cell motility. In undifferentiated and moving<br />

cells, Kif1c-GFP localizes to the centrosomal region and the cell periphery, especially<br />

cell extensions. Upon differentiation, Kif1c concentrates at the tips of the elongating<br />

cells, and when cells fuse redistributes to the tips of the recently fused myofibres.<br />

Although initial studies indicated an involvement of Kif1c in cell adhesion in<br />

macrophages, information on Kif1c’s function in cell polarity and fusion is lacking.<br />

Current work to i<strong>de</strong>ntify Kif1c’s cargo will hopefully reveal novel factors that are<br />

important for the establishment and/or maintenance of cell polarity, and will help to<br />

un<strong>de</strong>rstand how their correct spatial distribution supports changes in cell morphology.<br />

Anastasiya TRUSHKO trushko@mpi-cbg.<strong>de</strong><br />

Max Planck <strong>Institut</strong>e of Molecular Cell Biology and Genetics, Dres<strong>de</strong>n,<br />

Germany<br />

INTERACTION OF XMAP215 WITH DYNAMIC MICROTUBULES STUDIED WITH<br />

OPTICAL TWEEZERS<br />

Anastasiya Trushko, Erik Schäffer 2 , Jonathon Howard<br />

Microtubules have a crucial organizing role in all eukaryotic cells. They are long<br />

and relatively stiff hollow tubes of protein that consist of tubulin dimers and can rapidly<br />

disassemble in one location and reassemble in other. Cell can regulate the<br />

microtubule growth rate and their location with help of MAPs (microtubule associated<br />

proteins). One member of this large family is XMAP215.<br />

The protein XMAP215 binds to the end of growing microtubules and changes the<br />

polymerization rate, promoting faster elongation. To investigate the addition of tubulin<br />

dimmers to the plus end of the microtubule by XMAP215 and the <strong>de</strong>pen<strong>de</strong>nce of the<br />

addition on the applied force, XMAP215 is tethered to a microsphere held in optical<br />

trap. Because XMAP215 remains at the microtubule end for several rounds of tubulin<br />

addition, one can use it as a handle to hold the microtubule tip. In this way one can<br />

assess the force exerted by polymerizing microtubule and explore changing its growth<br />

dynamics un<strong>de</strong>r applied load with high temporal and spatial resolution.<br />

48


Feng-Ching TSAI : tsai@amolf.nl<br />

Biological Soft Matter Lab, Amsterdam, Netherlands<br />

SYNTHETIC CYTOSKELETAL NETWORKS IN SOFT CONFINEMENT<br />

Konstantinos TSEKOURAS : Konstantinos.Tsekouras@curie.fr<br />

<strong>Institut</strong> Curie, UMR 168, Paris, France<br />

ACTIN FILAMENT BUNDLE PUSHING AGAINST A WALL<br />

Motivated by recent experiments of Israelachvili et al., we construct a theoretical<br />

mo<strong>de</strong>l of a bundle of parallel actin filaments growing together from a nucleating<br />

location towards a wall upon which they exert force. We incorporate in our mo<strong>de</strong>l actin<br />

polymerization and <strong>de</strong>polymerization as well as hydrolysis of ATP-actin directly to<br />

ADP-actin. A phase diagram is <strong>de</strong>rived and the dynamics of the system theoretically<br />

investigated. All results are validated with extensive Monte-Carlo simulations and<br />

areas of agreement or <strong>de</strong>viation with experimental results discussed.<br />

49


Stanilas VINOPAL : vinopal@img.cas.cz<br />

<strong>Institut</strong>e of Molecular Genetics ASCR, Prague, Czech Republic<br />

LOCALIZATION OF GAMMA-TUBULIN IN NUCLEOLUS AND ITS DYNAMICS<br />

Gamma-tubulin is the key protein for nucleation of microtubules. We have shown<br />

previously that in human gliomas and glioblastoma cell lines γ-tubulin aberrantly<br />

accumulated in the cytoplasm (J. Neuropathol Exp. Neurol. 65: 465, 2006).<br />

Immunofluorescence microscopy of samples prepared un<strong>de</strong>r various fixation<br />

conditions disclosed that besi<strong>de</strong> microtubule organizing centers, γ-tubulin was also<br />

located in the cytoplasm in the form of large aggregates. Moreover, un<strong>de</strong>r some<br />

fixation conditions, bright staining for γ-tubulin was observed in nuclei with anti-pepti<strong>de</strong><br />

monoclonal antibodies against γ-tubulin. Immunofluorescence with nucleolar markers<br />

and marker of Cajal bodies <strong>de</strong>monstrated accumulation of γ-tubulin in nucleoli but not<br />

in Cajal bodies. Nucleolar staining was inhibited when antibodies were preabsorbed<br />

with immunizing pepti<strong>de</strong>s. Surprisingly, nucleolar staining of interphase cells was not<br />

limited to glioblastoma cells, but was also <strong>de</strong>tected in the other tested cultured cells.<br />

The presence of nucleolar γ-tubulin was further confirmed by TEM, immunoblotting<br />

and immunofluorescence of isolated nucleoli. To assess whether exogenous γ-tubulin<br />

is associated with nucleoli, various tagged versions of human γ-tubulin were prepared.<br />

In all tested cases exogenous γ-tubulin was <strong>de</strong>tected in the nucleoli. After treatment<br />

with leptomycin B, γ-tubulin did not accumulate in the nucleus suggesting the<br />

in<strong>de</strong>pen<strong>de</strong>nce of its nuclear export/transport on the RanGTP/Crm1 pathway. Using<br />

green-to-red photoconvertible γ-tubulin-DendraII fusion, it was shown that γ-tubulin<br />

does not rapidly shuttle between cytoplasm and nucleus. Confocal imaging of mitotic<br />

cells revealed the association of γ-tubulin with nucleoli remnats in prophase and with<br />

newly forming nucleoli in telophase as well. These results suggest that γ-tubulin might<br />

get into the nucleolus during mitotis. In conclusion, we have shown the novel γ-tubulin<br />

localization in the nucleolus. Our findings indicate that γ-tubulin can fulfill, in<br />

transformed as well as non-transformed cells, other functions in addition to<br />

microtubule nucleation.<br />

50


Simone WIEGAND : s.wiegand@fz-juelich.<strong>de</strong><br />

Research center Juelich, IFF - Soft con<strong>de</strong>nsed Matter, Germany<br />

MOVEMENT OF BIO- AND SOFT MATTER IN A TEMPERATURE GRADIENT<br />

In or<strong>de</strong>r to explain how the concentration problem of early evolution in the highly<br />

diluted ocean could have been solved often the importance of temperature gradients is<br />

discussed as an origin of life concept [1]. Also in the reproduction process of<br />

mammalians temperature gradients gui<strong>de</strong> the sperms from the cooler reservoir site,<br />

towards the warmer fertilization site [2]. Very recently it could be shown the convective<br />

flow both drives the DNA replication polymerase chain reaction while concurrent<br />

thermophoresis accumulates the replicated DNA in bulk solution. In or<strong>de</strong>r to gain a<br />

<strong>de</strong>eper un<strong>de</strong>rstanding of the un<strong>de</strong>rlying mechanism we study systematically bio- and<br />

soft matter in a temperature gradient.<br />

Various low molecular weight structures such as sugar molecules, sugar surfactant<br />

and microemulsions we studied by a holographic grating method called infrared<br />

thermal diffusion forced Rayleigh scattering (IR-TDFRS) setup [3]. Additionally we<br />

<strong>de</strong>veloped a thermal diffusion cell in or<strong>de</strong>r to study synthetic and biological colloids<br />

un<strong>de</strong>r a microscope. The new <strong>de</strong>veloped set-up is presented and its strength and<br />

weakness in comparison with other methods is discussed. As first system we<br />

investigate colloidal suspensions of silica particles and of fd-virus in a temperature<br />

gradient. Open questions such as the radial <strong>de</strong>pen<strong>de</strong>nce of the thermal diffusion<br />

coefficient and its relation with the interfacial tension are consi<strong>de</strong>red. Additionally,<br />

empirical correlations with the ratio of the thermal expansion coefficient and the<br />

kinematic viscosity for polar and non-polar substances are discussed.<br />

Marija ZANIC : zanic@mpi-cbg.<strong>de</strong><br />

Max Planck <strong>Institut</strong>e of Molecular Cell Biology and Genetics, Dres<strong>de</strong>n,<br />

Germany<br />

EB1 RECOGNIZES THE NUCLEOTIDE STATE OF TUBULIN IN THE<br />

MICROTUBULE LATTICE<br />

Plus-end-tracking proteins (+TIPs) are localized at the fast-growing, or plus end, of<br />

microtubules, and link microtubule ends to cellular structures. One of the best studied<br />

+TIPs is EB1, which forms comet-like structures at the tips of growing microtubules.<br />

The molecular mechanisms by which EB1 recognizes and tracks growing microtubule<br />

ends are largely unknown. However, one clue is that EB1 can bind directly to a<br />

microtubule end in the absence of other proteins. Here we use an in vitro assay for<br />

dynamic microtubule growth with two-color total-internal-reflection-fluorescence<br />

imaging to investigate binding of mammalian EB1 to both stabilized and dynamic<br />

microtubules. We find that un<strong>de</strong>r conditions of microtubule growth, EB1 not only tip<br />

tracks, as previously shown, but also preferentially recognizes the GMPCPP<br />

microtubule lattice as opposed to the GDP lattice. The interaction of EB1 with the<br />

GMPCPP microtubule lattice <strong>de</strong>pends on the E-hook of tubulin, as well as the amount<br />

of salt in solution. The ability to distinguish different nucleoti<strong>de</strong> states of tubulin in<br />

microtubule lattice may contribute to the end-tracking mechanism of EB1.<br />

51


Lecturers and organizers<br />

e-mail<br />

Laurent BLANCHOIN<br />

Enrique DE LA CRUZ<br />

Jonathon HOWARD<br />

Fanck JULICHER<br />

Marko KAKSONEN<br />

Gijsje KOEDERING<br />

David KOVAR<br />

Pekka LAPPALAINEN<br />

Matthias RIEF<br />

Phong TRAN<br />

Mike SIXT<br />

Cécile SYKES<br />

laurent.blanchoin@cea.fr<br />

enrique.<strong>de</strong>lacruz@yale.edu<br />

howard@mpi-cbg.<strong>de</strong><br />

julicher@mpipks-dres<strong>de</strong>n.mpg.<strong>de</strong><br />

kaksonen@embl.<strong>de</strong><br />

gkoen<strong>de</strong>rink@amolf.nl<br />

dkovar@uchicago.edu<br />

pekka.lappalainen@helsinki.fi<br />

mrief@ph.tum.<strong>de</strong><br />

tranp@mail.med.upenn.edu<br />

sixt@biochem.mpg.<strong>de</strong><br />

cecile.sykes@curie.fr<br />

52


FEBS Journal offers:<br />

The journal for rapid publication<br />

of full-lengthlength research papers and<br />

reviews in all areas of the<br />

Molecular Life Sciences<br />

www.febsjournal.org<br />

• Fast and helpful peer review (normally

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!