Skip to main content
2010 -
Grant Awardees - Program

Spatiotemporal control of neuronal activity with holographic patterned illumination

CURTIS Jennifer (USA)

School of Physics/Molecular and Cell Biophysics Lab. - Georgia Institute of Technology - Atlanta - USA

EMILIANI Valentina (ITALY)

Photonics Department - Wavefront-engineering microscopy group - Vision Institute - Paris - FRANCE

ISACOFF Ehud Y. (USA)

Dept. of Molecular and Cell Biology - University of California Berkeley - Berkeley - USA

PEDARZANI Paola (ITALY)

UCL Neuroscience, Physiology and Pharmacology - University College London - London - UK

In recent years the use of advanced optical techniques has had a strong impact in neurobiology not only for visualizing neuronal structures and signaling processes, but also for controlling neuronal activity. This is possible thanks to a growing list of photosensitive tools that can modify their chemical or conformational structures upon illumination and precisely trigger the propagation of neuronal signals. In addition to the commonly used caged neuroactive compounds, genetically encoded light-sensitive proteins have recently been developed permitting neuronal activation and inactivation. In conjunction with spatiotemporally resolved photo-stimulation techniques, these photoswitchable proteins represent the most promising alternative to electrical stimulation, providing ways to control precisely in space and time the activity of specific types of brain cells.

These approaches require fast, flexible and precise illumination schemes, permitting the selective activation and imaging of sub-cellular regions or multi-cellular ensembles. The Emiliani group has recently developed the first one-photon scan-less holographic microscope where a liquid crystal spatial light modulator (LC-SLM) in the excitation path generates 2D and 3D multiple diffraction limited spots or shaped domains of excitation that can accurately cover a single cell or sub-cellular structures. They showed that shaped holographic illumination for large excitation areas has a significant higher axial resolution than a Gaussian beam, and concentrates all of the laser power on the desired region, thus providing the high intensity needed for fast photostimulation. They have also shown that further optical confinement can be achieved in two photon holographic illumination combined with temporal focusing.

The aim of this proposal is to use this innovative approach to study fundamental properties of central neurons. Holographic photolysis of caged compounds will be used as a novel means for mapping the subcellular distribution, modulation and function in neuronal signaling of ion channels in brain slices. Holographic photoactivation in vivo of the engineered light gated channel LiGluR will be used to elucidate the neural circuit basis of locomotion in zebrafish.

2010 -
Grant Awardees - Program

Mechanistic analysis of neuronal circuit structure and function

FRIEDRICH Rainer W. (GERMANY)

Dept. of Neurobiology - Friedrich Miescher Institute for Biomedical Research - Basel - SWITZERLAND

SEUNG H. Sebastian (USA)

Dept. of Brain and Cognitive Sciences - Massachusetts Institute of Technology - Cambridge - USA

YOSHIHARA Yoshihiro (JAPAN)

Lab. for Neurobiology of Synapse - RIKEN Brain Science Institute - Saitama - JAPAN

Neuroscientists have measured biophysical properties of individual nerve cells and monitored the activation of large brain areas in intact animals including humans. A coherent understanding of brain functions now requires quantitative insights into the interactions between individual neurons in order to understand how sophisticated computations are performed by neuronal circuits. This question raises a series of conceptual and technical challenges. First, connections between many or even all neurons in a circuit have to be determined by 3-dimensional imaging methods that achieve nanometer resolution throughout very large volumes. Second, activity patterns in the intact brain have to be measured with a resolution of individual nerve cells to quantitatively characterize neuronal computations. Combined measurements of activity and connectivity in the intact brain are then required to understand how the function of a circuit is determined by its structure. Third, activity and connectivity have to be manipulated in order to perturb a circuit and examine the effects onto neuronal computations and behaviors. Fourth, principles underlying neuronal circuit structure and function have to be extracted by mathematical approaches. To address these issues we will focus on the sense of smell in a small vertebrate, the zebrafish, which provides unique advantages. The connectivity among large numbers of neurons will be determined by novel 3-dimensional electron microscopy techniques and analysis methods based on machine learning. Activity patterns will be measured using non-linear optical methods that permit the visualization of activity across thousands of neurons in the intact brain. Advanced genetic methods will be used to manipulate the function of defined neurons in the intact brain. These approaches include optical techniques to precisely control the activity of individual neurons in space and time. Structure-function relationships of circuits will then be analyzed using mathematical methods and computational modelling. This interdisciplinary approach is expected to provide elementary insights into the mechanisms by which neuronal circuits process information and control behaviors, which is essential to understand how higher brain functions emerge from complex communications between large numbers of nerve cells.

2010 -
Grant Awardees - Program

The regulation of growth as a driving force for patterning and regeneration

DEUTSCH Andreas (GERMANY)

Centre for Information Services and High Performance Computing - Dresden University of Technology - Dresden - GERMANY

GALLIOT Brigitte (SWITZERLAND)

Dept. of Genetics and Evolution - University of Geneva - Geneva - SWITZERLAND

IRVINE Kenneth (USA)

Waksman Institute - Rutgers University - Piscataway - USA

MORATA Ginés (SPAIN)

- Centro de Biología Molecular CSIC-UAM - Madrid - SPAIN

TANAKA Elly M. (USA)

Molecular Mechanisms of Regeneration - Research Institute of Molecular Pathology - Vienna - AUSTRIA

A fascinating, unanswered question in biology is how some organisms respond to injury by regenerating the lost body structure. Two fundamental aspects of the problem are first, how does injury trigger a growth and patterning response? Second, how does the regenerating tissue “read” the overall size of the animal to produce an appropriately sized structure? While some molecular pathways have been implicated, a clear mechanism linking physical injury to cellular and molecular response is wholly lacking. Furthermore organ growth control remains a challenging problem in biology. Here we propose to couple the research efforts in three regeneration model organisms, Hydra, Axolotl (salamander) and Drosophila, to quantitative mathematical modeling as a means to address these issues in a novel way.

Drosophila has been a fertile system for identifying and analyzing molecular pathways involved in imaginal disc growth and patterning. We will therefore use the Drosophila imaginal disc as a starting point for identifying and analyzing mechanisms that link tissue injury to a growth and patterning response. Disadvantage: we cannot observe the regeneration in discs as it is happening. Therefore complementarily, we will use two classical regeneration models, Hydra and salamander (Axolotl) where regeneration can be exquisitely controlled and visualized in live animals to build on the physical link between injury and molecular signaling to cellular response. To harmonize the systems, we will all focus on the regeneration of epithelial structures where it is most likely that cellular responses may be similar: the Drosophila wing and leg discs are simple epithelia. In analogy, the Hydra body column consists of a bilayered myoepithelium, and finally, amputation of the Axolotl tail results in regeneration of the spinal cord through a neuroepithelium.

Therefore, we are not looking for molecules responsible for regenerating a particular body structure but for the general mechanisms used to control regeneration and size control of epithelial-based organs. Our specific aim is to come to a deeper mechanistic and integrative understanding of how signaling pathways respond to cell/tissue injury to initiate growth, patterning as well as size control response. For this reason mathematical modeling will help integrate the results obtained in the different phyla to identify shared mechanisms.

2010 -
Grant Awardees - Program

Chiral effects in DNA supercoiling

KORNYSHEV Alexei (RUSSIA)

Dept. of Chemistry - Imperial College London - London - UK

VAN DER HEIJDEN Gert (NETHERLANDS)

Dept. of Civil, Environmental and Geomatic Engineering - University College London - London - UK

WUITE Gijs J.L. (NETHERLANDS)

Molecular Biophysics Group / Faculty of Sciences/Division of Physics and Astronomy - Vrije University Amsterdam - Amsterdam - NETHERLANDS

ZECHIEDRICH Lynn E. (USA)

Dept. of Molecular Virology and Microbiology, Biochemistry, Pharmacology - Baylor College of Medicine - Houston - USA

This project will investigate the forces between two double-stranded DNA molecules in a protein-free environment. Due to the chiral nature of DNA, these interactions are also chiral and affect DNA conformation and, in turn, are affected by topological constraints imposed on the molecule. Existing (Kornyshev-Leikin) electrostatic theory provides an explanation for the homology-promoted attraction. This theory also predicts a chiral torque that tends to make long DNA molecules wind around each other. The effect will be stronger if the DNA tracts in close juxtaposition are physically identical (i.e., homologous).

The double-stranded helix of DNA in cells is slightly (~30%) underwound relative to relaxed DNAs such as those used to test the homology interaction theory mentioned above. Additionally, cellular forces (in the pN ranges) cause transient extreme under- and overwinding of DNA. These topological states are essential for DNA transactions and without them cells die. The complex interplay between homology and supercoiling is the core subject of this project.

The project will determine: (i) the degree to which homologous tracts attract each other more strongly than non-homologous tracts; (ii) the force keeping two homologous tracts in juxtaposition; (iii) how homology recognition is influenced by supercoiling. These questions will be answered by performing a complementary series of single-molecule experiments and biochemical experiments with circular DNAs of varying degrees of supercoiling. Experiements will then be move to inside live cells. The experiments will be supported and rationalised by a theory, to be developed. This new theory requires the merger of the continuum theory of elastic braids and the theory of chiral electrostatic DNA-DNA interactions. The theory will distinguish juxtapositions of homologous and non-homologous DNA tracts and will allow us to investigate the dependence of supercoiled structures of DNA on structural and environmental parameters, including homology length and type and concentration of counterions.

2010 -
Grant Awardees - Program

Synthetic Biology of cell division: reconstructing the bacterial division machinery in the test tube

MARGOLIN William (USA)

Dept. of Microbiology and Molecular Genetics - University of Texas Medical School at Houston - Houston - USA

RIVAS German (SPAIN)

Centro de Investigaciones Biológicas - CSIC - Madrid - SPAIN

SCHWILLE Petra (GERMANY)

Dept. of Cellular and Molecular Biophysics - Max Planck Institute of Biochemistry - Martinsried - GERMANY

VICENTE Miguel (SPAIN)

Dept. of Microbial Biotechnology - Centro Nacional de Biotecnología - Madrid - SPAIN

We will build the minimal molecular engine required to initiate bacterial division by reconstituting its molecular machinery (the divisome) and reproduce its vital functions in vitro. Cytokinesis is a fundamental process that is known in bacteria in sufficient detail to provide a comprehensive list of its molecular effectors.

Our novel idea is to reconstruct and track the operation of the minimal functional protein set needed to initiate division (the proto-ring) and the two main positioning mechanisms (Min system and nucleoid occlusion) that select the constriction site.

We will use complementary biochemical, biophysical, genetic and imaging methodologies to reconstitute the proto-ring components (FtsZ, FtsA and ZipA) into artificial membrane-bounded compartments that mimic the divisome organization at the cell membrane. The use of biomimetic materials, vesicles and nanodiscs, as substrates on which to reconstitute the machine will be complemented using biovessels to assemble the proto-ring within structures that reproduce the cellular environment with minimal genetic information. Ideal conditions regarding the composition and shape of the membrane compartments will be designed for divisome reconstruction in nanodiscs, biomimetic bilayers, giant vesicles, and membrane tubes pulled to various geometries.

Research will then focus on the reconstruction and stability of proto-rings in biovessels, the interactions and dynamics of proteins within the division ring and how ring constriction is regulated. In vivo top-down strategies will be designed to minimize the components necessary for cytokinesis. The mechanism of proto-ring positioning by Min proteins and the membrane deformation imposed by proteins responsible for its constriction will finally be investigated.

Because of their relative simplicity, bacteria will probably be the first cells to be fully understood at a systems level, and our results will help usher in the new field of synthetic biology of cell division and will open new horizons for pharmacological applications.

2010 -
Grant Awardees - Program

Viral docking and maturation in whole bacterial cells at near atomic resolution and in 4 dimensions

HOWARD Martin (UK)

Computational and Systems Biology - John Innes Centre - Norwich - UK

VIOLLIER Patrick (SWITZERLAND)

Dept. of Microbiology and Molecular Medicine - University of Geneva - Geneva 4 - -

WRIGHT Elizabeth R. (USA)

Biochemistry - University of Wisconsin-Madison - Madison - USA

One of the main motivations for examining host-pathogen relationships between viruses and cells is for the development of target-specific antimicrobials. Since many aspects of the complex nature of host-pathogen interactions are still enigmatic, even at the most fundamental level, an interdisciplinary and synergistic effort is proposed here to study viral infection and replication in the context of an intact host cell in four dimensions (4D, i.e. in 3D imaging as a function of time) and at a level of resolution never before achieved. Scientists from Geneva (CH), Atlanta (USA) and Norwich (UK) that are still early in their careers with expertise in genetics, live-cell fluorescence imaging, electron cryo-tomography (ECT, 3 dimensional electron microscopy) and mathematical modeling will use a bacterial model system (Caulobacter crescentus) to observe and quantitatively describe how viral particles (bacteriophages) infect and replicate in the live host. The emphasis will be on the mechanisms by which bacteriophages exploit both i) the surface structures on the host for infection (i.e. adsorption, adhesion and genome injection) and ii) the host cell cycle and/or its internal structures (cytoskeleton) for progeny production. C. crescentus is an ideal model system for these studies because it is small enough for whole-cell ECT imaging during bacteriophage infection and replication. Moreover, mutations can easily introduced into C. crescentus and it is easy to enrich for cells that are in a specific cell-cycle phase. These features provide an ideal experimental setting for a marriage of imaging and genetics to define external/internal structures and the cell-cycle parameters of the host that influence bacteriophage infection and replication. These studies, though fundamental in nature and of conceptual relevance for many aspects of biology, could potentially be useful in developing new antiviral treatments.

2010 -
Grant Awardees - Program

Central auditory processing: from single cells to perception and learning of complex sounds

BIALEK William S. (USA)

Dept. of Physics - Princeton University - Princeton, New Jersey - USA

DOUPE Allison (CANADA)

Dept. of Physiology and Psychiatry - University of California, San Francisco - San Francisco - USA

WILD John Martin (NEW ZEALAND)

Dept. of Anatomy with Radiology - University of Auckland - Auckland - NEW ZEALAND

How does the brain process the complex, dynamic signals that we encounter in the natural environment? And are there different central circuits active for processing the different aspects of sensory stimuli, or are shared neural substrates involved? In the case of vision, we have a handle on these questions, with an understanding of elementary features and their representation in the brain. In the auditory system, especially above the thalamus, the issues are complex and the organizing principles much less clear, with no overall framework yet to guide our understanding of auditory processing. With respect to these questions, songbirds provide a very useful model. They learn to sing using auditory feedback, and show lifelong perceptual learning of other birds’ songs. They possess a hierarchical network of central auditory areas, analogous to those in mammals, which likely subserves these auditory behaviors. The combination of a rich set of behaviorally relevant auditory stimuli and tasks and of an identified neural circuit opens up the possibility of fundamental understanding of how auditory networks function in perception and learning. To this end, we propose an international collaboration to combine neurophysiological recordings of multiple neurons in awake songbirds; new theory-based approaches to extracting complex feature selectivity and analyzing population coding in these neurons; and state-of-the art anatomical techniques to map the connectivity of neurons at electrophysiologically identified sites and to create a functional wiring diagram. Our work will be guided by, and test the following hypotheses: a) there is a systematic and hierarchical organization of feature response properties across the avian forebrain, including specialized pathways for different aspects of complex sounds, such as frequency and time, b) the collective activity of neuronal populations across these areas reveals a further organization of sound encoding not evident in the properties of neurons considered individually, and c) this network can be dynamically reconfigured by bottom-up influences (such as stimulus statistics), top-down influences (behavioral task), and learning (of adult song discriminations). Our goal is a principled picture of how this circuit is organized to analyze and learn complex natural sounds, with potential relevance to all vertebrates.

2010 -
Grant Awardees - Program

The calcified byssus of Anomia: a unique solution to underwater adhesion

BIRKEDAL Henrik (DENMARK)

Dept. of Chemistry and iNANO - Aarhus University - Aarhus - DENMARK

WAITE Herbert (USA)

Molecular Cell Developmental Biology Department - University of California-Santa Barbara - Santa Barbara - USA

Sedentary animals attach to substrates with glues that work underwater, a feat we do not master in our technology. A well-known example is the blue mussel who uses its beard, called the byssus, to attach the soft mussel tissue to the hard substrates it lives on. The blue mussel byssus is made of protein. In contrast to the multi-thread pure protein byssus of the blue mussel, its cousins called the Anomiidae attach via a byssus plug made from a single thread that is calcified, i.e. contains calcium carbonate crystals in addition to proteins. Almost nothing is known about this strange attachment system and in particular the adhesive. We will investigate this intriguing solution to sticking in place using an interdisciplinary approach where we will understand both the mechanical function and the biomolecules involved in the adhesion. This is done by joining the forces of materials chemists and biochemists.

In the project we will use state of the art tools to investigate the structure on the 1 nm to 10 µm length scale of the byssus with focus on the adhesive. In this range of distances molecules interact with each other and develop structures that have macroscopic functions. We will measure the strength of the byssus attachment system. By prodding the surface with a sharp needle, a technique called nanoindentation, we will measure the local mechanical performance on the µm scale. Thereby we will unravel the mechanical performance of the different structural elements. In particular, we can measure the mechanical properties of the adhesive layer itself. Through these experiments we will understand the function of the byssus but also how this function comes about. To do this we will investigate which molecules are involved and how they are built up from amino acids. We will also determine if the animal uses specially modified amino acids, as is sometimes the case in such systems in particular in the blue mussel. To fully understand how these biological glues function, we will measure the structure and self assembly capabilities of selected molecular moieties. We will also measure how they interact with specific surfaces to unravel their role in the adhesive process.

Through this interdisciplinary project we will develop a deep understanding of how these animals attach. This may provide inspiration for novel adhesives for use in e.g. biomedical applications.

2010 -
Grant Awardees - Program

Optical interrogation of motor cortex to provide insight into neuronal control of movement.

BOYDEN Edward (USA)

Media Lab and McGovern Institute - MIT - Cambridge - USA

MARTIN Kevan A. C. (UK)

Institute of Neuroinformatics - University of Zurich - Zurich - SWITZERLAND

MURPHY Timothy H. (USA)

Dept. of Psychiatry - Univ. of British Columbia - Vancouver - CANADA

Many times a second we engage the motor system: we evaluate evidence, plan options, decide, and act, all within 10’s of ms. Given fixed times for nerve conduction and synaptic delays there would appear to be little time for processing through multiple circuits. However, both primates and cats appear to have widespread and relatively unstructured lateral connectivity within their motor cortex. Such extensive local motor cortex connections may endow the system with flexibility needed to learn new tasks and coordinate a wide range of outputs. Our central question is how the local circuits in the primary rodent motor cortex process sensory information to produce a coherent, integrated, and specific pattern of corticospinal output. When faced with these less structured circuits we exploit genetics and develop mouse models where specific classes of neurons can be selectively excited or inhibited using light over both the timescale and landscape of the cortical area devoted to sensory-motor processing. The primary focus will be on the in vivo structure and function of single neurons and the local circuits they form in the rodent motor cortex M1, specifically in the forepaw representation that are associated with simple reaching tasks. Our hypothesis is that the motor cortex implements a decision making machine that operates in two main stages through the cortical lamina. The widely distributed connections in the superficial layers provide the circuits for broadcasting incoming patterns of sensory information for evaluation. The resulting decisions are then transmitted to the deep cortical layers, which coordinate the output patterns. While the basic input pathways and output neurons in the rodent forepaw representation of M1 are known, there are few data on functional properties of individual neurons, laminar differences in properties, or their collective activity. It is intended that the experiments outlined here will provide the functional and structural data for us to develop a biologically-realistic model of the mouse motor cortex.

2010 -
Grant Awardees - Program

Analysis of “cytoplasmic freezing” – preserving cellular architecture

BRUNNER Damian (SWITZERLAND)

Institute of Molecular Life Sciences - University of Zurich - Zurich - SWITZERLAND

FLORIN Ernst-Ludwig (GERMANY)

Center for Nonlinear Dynamics and Dept. of Physics - University of Texas at Austin - Austin - USA

HOENGER Andreas (SWITZERLAND)

Dept. of Molecular, Cellular and Developmental Biology - University of Colorado at Boulder - Boulder - USA

In response to nutrient starvation, yeast cells enter a quiescent state. This state is defined by a marked increase in the viscosity of the cell cytoplasm, a phenomenon we term “cell-freezing”. Here we propose a detailed structural and functional study of cell-freezing in fission yeast (S. pombe). Cell-freezing is a novel phenomenon discovered only recently through measurements of the viscoelastic properties of the cytoplasm of fission yeast cells. It leads to a dramatic immobilization of all visible sub-cellular structures and implies the presence of a mechanically stable network throughout the cell, we thus hypothesize that such a dense and homogeneous network develops when the cell enters the starvation state and that this network is sufficiently fine to immobilize all major cell components.

The four objectives of the proposed research are A: characterize details of cell-freezing, B: identify and dissect its molecular machinery, C: analyze its function, and D: search for cell-freezing in higher organisms. To address these objectives, we will follow an interdisciplinary approach that includes methods from cell and molecular biology, structural biology and (bio-)physics.

2010 -
Grant Awardees - Program

Dissecting the molecular mechanisms regulating somatic cell reprogramming

CALIFANO Andrea (ITALY)

Joint Centers for Systems Biology - Columbia University - New York - USA

COSMA Maria Pia (ITALY)

Reprogramming & Regeneration Group - Center for Genomic Regulation, CRG - Barcelona - SPAIN

Somatic nuclei can be reprogrammed to a pluripotent or embryonic stage. Somatic cell reprogramming has been shown to be possible via animal cloning, cell fusion, and ectopic expression of a few embryonic stem cell (ESC) factors. The reprogramming time of a cell nucleus is normally relatively long and it depends upon cell type, methods of reprogramming, and culture conditions used. These observations led us to envisage that the process according to which somatic cells change their fate involves many factors and sequential events. Thus, transcription factors, chromatin-modifying complexes and signalling molecules are likely to operate in a specific order to induce and maintain the reprogramming of a somatic nucleus. We have recently shown that the periodic activation of Wnt/beta-catenin signalling controls fusion-mediated somatic-cell reprogramming. Only this periodic and limited nuclear accumulation of beta-catenin allows ESCs to reprogramme somatic cells after fusion. Furthermore, in ESCs, either overexpression of Axin2, which is part of the beta-catenin destruction complex, or deletion of GSK3, which is devoted to the phosphorylation of beta-catenin, inhibits ESC-fusion-mediated somatic-cell reprogramming. In contrast, deletion of a specific beta-catenin modulator, in ESCs strongly enhances their reprogramming ability. Our hypothesis is that signalling dependent genes encode for key reprogramming factors, which we aim to identify. We will perform transcriptome, microRNAome and epigenome analyses of ESCs pre-treated with positive and negative modulators of the Wnt pathway. In addition, we will perform the same type of analysis using wild-type and mutant ESCs which carry the activated or inhibited Wnt signalling pathway. All of the collected data will be analysed by ARACNe, MINDy and MRA reverse engineering algorithms, to reconstruct the reprogramming regulatory network and to identify pathways and master reprogrammer genes. Finally, the reprogrammer candidates will be cloned and validated for their ability to induce fusion-mediated reprogramming or direct reprogramming.

2010 -
Grant Awardees - Program

Cycle-Quant: Defining Cell Cycle Progression and Responses to Perturbations

CARPENTER Anne (USA)

Imaging Platform - Broad Institute of Harvard and MIT - Cambridge - USA

CHANG Young-Tae (KOREA, REPUBLIC OF (SOUTH KOREA))

Dept. of Chemistry - National University of Singapore - Singapore - SINGAPORE

WOLTHUIS Rob (NETHERLANDS)

Dept. of Clinical Genetics - Section of Oncogenetics - Amsterdam - NETHERLANDS

Cycle-Quant: Developing Cell Cycle Profiles to Classify Responses and Regulators

Readily detectable changes in DNA content and cellular morphology are used to assign conventional cell cycle phases. However, cell division requires coordination of cell size, organelle duplication, cytoskeletal rearrangements, and changes in key proteins. Here, an interdisciplinary team of researchers will develop new methods for advanced cell cycle profiling. Novel landmarks will be identified in the continuum of the mammalian cell cycle, and in cycles of organelles and cell cycle proteins. An integrated and multi-disciplinary team of biologists, chemists, and computer scientists will establish new genetic and chemical cellular labels and machine learning-based computational techniques applicable to time-lapse microscopy.

The main aims are:

1. To develop advanced cell cycle profiling using a standardised and expandable set of non-transformed human cell lines. Different classes of genetically encoded fluorescent biological markers track, directly or indirectly reflecting cellular changes related to the cell cycle, will be tracked by time lapse fluorescence microscopy. Acquired image data will be processed by new computer-based data extraction methods for automated image recognition and adequate description of a spectrum of ‘sub-cellular cycles’ such as those of dividing organelles.

2. To develop novel, and uniform tools to identify flexible key cell-cycle landmarks, bypassing the requirement for the elaborate, intrusive introduction of genetic markers. This would create unprecedented flexibility for cell cycle profiling in any given cell type. We will a) use rich information from non-fluorescent cell images and, b) develop and validate permeable fluorescent cell-cycle-markers.

3. We will test cell cycle profiling by quantitatively comparing functions of key regulators of the G2-M transition and classify new regulators in focussed pathways.

We will combine our expertises to re-define cell cycle progression in a quantitative way, with the goal of assigning complex cell-cycle progression profiles. This coordinated effort will provide unprecedented opportunities to quantitatively compare genetic and chemical perturbants, essential for assigning functions to newly identified cell cycle regulators, and the development of robustly predictive models of cell cycle progression.

2010 -
Grant Awardees - Program

A microfluidic, small molecule approach to perturbation of the pluripotency transcription network

CHAMBERS Ian (UK)

MRC Centre for Regenerative Medicine/Institute for Stem Cell Research - School of Biological Sciences - Edinburgh - UK

LEE Luke P. (USA)

Dept. of Bioengineering - University of California, Berkeley - Berkeley - USA

SCHROEDER Timm (GERMANY)

Cell Systems Dynamics, Department of Biosystems Science and Engineering - ETH Zurich - Basel - SWITZERLAND

WANDLESS Thomas (USA)

Dept. of Chemical and Systems Biology - Stanford University - Stanford - USA

Stem cells possess two defining features; the capacity to maintain their identity during extended cell division (referred to as self-renewal) and the capacity to alter their identity to form specialised cells (referred to as differentiation). Preservation of a functional stem cell population requires a balance between self-renewal and differentiation. Although functionality operates on the stem cell population as a whole, it is the choices of individual cells that must be controlled to maintain such functionality. Self-renewal of embryonic stem (ES) cells is controlled by the action of a gene regulatory network centred around the transcription factors Oct4, Sox2 and Nanog. However, the mechanisms controlling operation of this network are not well understood. Recently, we demonstrated an heterogeneity in the expression of Nanog, which is strongly and directly correlated with self-renewal efficiency. This leads us to hypothesise that the cross-regulatory interactions between Oct4, Sox2, Nanog proteins and their target genes are only partly stable. This may allow a dynamic range of states to be explored by ES cells, only some of which enable differentiation. Here, our objective is to test this hypothesis by watching how the Oct4/Sox2/Nanog transcription factor network operates in individual stem cells. To do this we will put differently coloured “reporters” into each of the transcription factor genes (e.g. red at Oct4, blue at Sox2 and yellow at Nanog). These reporters will tell us when, where and to what extent each gene is switched on. We will then use precision engineering to allow ES cells, the developmentally more advanced epiblast stem cells and early mouse embryos to be cultured in small chambers with a surface area of less than 1 mm2 over which we can flow culture fluid with varying levels of “switch” chemicals. These “switch” chemicals will be used to alter the action of modified transcription factor genes that we have also inserted into the cells. Changes in expression of the network components can then be followed microscopically using the coloured reporters in real time. These studies will deliver insight into the regulatory dynamics underpinning stem cell identity. They will allow us to see the dynamic changes as they occur in stem cells that are maturing during development and will enhance our ability to manipulate stem cells behaviour at will.

2010 -
Grant Awardees - Program

An interaction map of C. elegans dauer pheromone compoments and chemoreceptors

CLARDY Jon (USA)

Dept. of Biological Chemistry & Molecular Pharmacology - Harvard Medical School - Boston - USA

SENGUPTA Piali (USA)

Dept. of Biology and National Center for Behavioral Genomics - Brandeis University - Waltham - USA

TOUHARA Kazushige (JAPAN)

Department of Applied Biological Chemistry - Graduate School of Agricultural and Life Sciences - Tokyo - JAPAN

Pheromones are chemicals that are produced by individual organisms, and that elicit highly stereotyped behavioral and developmental responses in other individuals of the same species. The chemical composition of pheromones is often complex, consisting of many small molecules of related or unrelated structures. These molecules can elicit distinct responses when acting individually or in combination with other pheromone components. Pheromones are recognized by members of large families of seven transmembrane domain receptor proteins in vertebrates and invertebrates, the ligand specificities of only a small subset of which have been described. The challenge of isolating and defining the chemical composition of pheromones, together with the presence of large numbers of putative pheromone receptor proteins, has made it difficult to describe the coding strategy used to respond to these biologically and ecologically important signaling molecules.

The goal of this project is to generate a comprehensive map of pheromone-receptor interactions in the C. elegans model system. Over twenty-five years ago, it was demonstrated that C. elegans produces a mixture of small molecules called dauer pheromone, high concentrations of which trigger entry into the stress-resistant and long-lived dauer larval stage. Recently, we and others have begun to elucidate the chemical composition of dauer pheromone, and have identified two G protein-coupled receptors required for the responses to a subset of pheromone components. The ability to cultivate C. elegans in sufficiently large quantities to extract, identify and synthesize pheromone components, and the genetic and genomic power of C. elegans together with its compact sensory nervous system, makes the generation of a complete pheromone-receptor interaction map a feasible goal in this organism.

The specific aims of this collaborative project are: 1) to define and chemically synthesize all components of C. elegans dauer pheromone; 2) to identify the neurons and chemoreceptors required for the responses to each of the components in vivo; and 3) to characterize the ligand response properties, selectivity, and affinity of the receptors via heterologous expression assays. These interdisciplinary aims integrate the expertise of J. Clardy (USA) in small molecule chemistry, of P. Sengupta (USA) in neurogenetics of C. elegans, and of K. Touhara (Japan) in biochemical mapping of ligand-receptor interactions.

2010 -
Grant Awardees - Program

Spatio-temporal Rho GTPase signaling to the cytoskeleton during neuronal development and repair

DANUSER Gaudenz (USA)

Lyda Hill Dept. of Bioinformatics and Celle Biology - UT Southwestern Medical Center - Dallas - USA

JEON Noo Li (USA)

Dept. of Mechanical Engineering - Seoul National University - Seoul - KOREA, REPUBLIC OF (SOUTH KOREA)

PERTZ Olivier (SWITZERLAND)

Institute of Cell Biology - University of Bern - Bern - SWITZERLAND

Neurons have to perform a wide variety of complex morphogenetic events in order to functionally wire the developing brain. In the adult brain, this process continues with changes in morphology that alters their functional connectivity upon learning and experience, or during the repair of injured neural tissues. Specific signaling events to the cytoskeleton are essential for the control of these processes and are likely to be highly regulated at fine spatiotemporal scales. Accordingly, novel cell biological tools such as fluorescent biosensors that enable dissection of these signaling events in time and space, have given novel insights in signaling complexity. The aim of this proposal is to visualize the dynamics of the activation of specific signaling molecules in a panel of prototypical neuronal behaviors with a strong emphasis on the axon. We will study: axonal specification (the establishment of an axon and a dendrite), axonal guidance (the response of a growth cone to attractive and repulsive cues) and inhibition of axonal regeneration after injury (the response of the axon to a glial scar). Our hypothesis is that these different morphogenetic behaviors involve distinct signaling programs with a tight crosstalk between the signaling molecules of interest. Comparing these in the prototypical cell behaviors should give essential insights in how a neuron can perform this diverse set of tasks using the same signaling machinery. We propose to engineer a new generation of highly sensitive fluorescent probes to monitor the spatio-temporal activation profiles of the specific signaling molecules in primary E18 embryonal hippocampal neurons. Microfluidic technology will be used to engineer highly defined extracellular environments to induce and orient the different axonal behaviors. This will evoke the asymmetric nature of extracellular cues observed in vivo, and recapitulate the associated signal amplification mechanisms that will lead to robust signaling. Finally, computer vision techniques will be devised that allow the pooling of measurements from many cells into statistically significant datasets for identification of the spatiotemporal relationships among different signaling programs and cell behavior. Multivariate time-series analysis methods will be devised to inform the model on the cross-coordination between the activation of the signaling molecules of interest and neuronal morphogenesis.

2010 -
Grant Awardees - Program

Mitotic Spindles in Nematodes: From Comparative Biophysics to Evolutionary Biology

DELATTRE Marie (FRANCE)

Lab. of Biology ad Modelling of the Cell - Ecole Normale Superieure de Lyon- CNRS - Lyon - FRANCE

MÜLLER-REICHERT Thomas (GERMANY)

Medical Theoretical Center - University of Technology, Dresden - Dresden - GERMANY

NEEDLEMAN Daniel Joseph (USA)

School of Engineering and Applied Sciences - Harvard University - Cambridge - USA

The architecture of cells and subcellular structures can show remarkable variability between tissues and organisms, but there is currently little understanding of the evolutionary basis of this diversity. Thus it is unclear why metaphase spindles in different Eukaryotes exhibit a range of morphologies and the volumes of these spindles vary over one thousand fold. Here we propose a comparative study to investigate the evolutionary forces shaping the spindle. Using one cell stage nematode embryos as a model system, we will characterize spindle structure in several strains of ~60 species exhibiting a range of cell sizes, karyotypes, and life histories. A more detailed analysis of microtubule dynamics and spindle architecture from ~10 select species will reveal the biophysical basis of variation in spindle morphology. This comparative data will be interpreted using phylogenies and quantitative models of phenotypic evolution. We will then test the developed biophysical and evolutionary models with perturbation. This unique comparative approach combines molecular methods, quantitative microscopy, and mathematical models, to gain insight into how the mitotic spindle is shaped through evolution by selection, drift, and biophysical constraints.

2010 -
Grant Awardees - Program

Deliberative decision-making in rats

DUDCHENKO Paul (USA)

Dept. of Psychology - University of Stirling - Stirling - UK

LAUWEREYNS Jan (BELGIUM)

Graduate School of Systems Life Sciences - Kyushu University - Fukuoka - JAPAN

REDISH A. David (USA)

Dept. of Neuroscience - University of Minnesota - Minneapolis - USA

TSUDA Ichiro (JAPAN)

Dept. of Mathematics, Faculty of Science - Hokkaido University - Sapporo - JAPAN

WOOD Emma (UK)

Centre for Cognitive and Neural Systems - University of Edinburgh - Edinburgh - UK

Our goal is to examine deliberative decision-making in rats from neurophysiological, cognitive, and mathematical perspectives. Deliberative decision-making entails behaviors in which agents explicitly consider multiple possibilities before acting. Early experiments identified behaviors in which a rat paused, looking back and forth, before acting (Tolman1948). This behavior occurred during learning and after changes in experimental contingencies, and was suggested to create expectancies of the consequences of the available choices, but the computational and experimental techniques needed to address this question were not available at the time. An interest in this behavior has been revived in the past few years based on new computational models of decision-making systems (Daw2005, Niv2006, Redish2007a, Redish2008), on the availability of new behavioral measures (e.g. Lauwereyns2006), and on recent discoveries of prospective encoding in the hippocampus (Wood2000; Johnson2007a) and other structures (Ramus2007, vanderMeer2009, vanDuuren2009). Our team brings together expertise in behavioral neuroscience, neurophysiology, computational neuroscience, cognitive psychology, and mathematics. Experiments will examine prediction, evaluation, and action-selection mechanisms in rats making deliberative decisions. Computational projects will examine mechanisms by which these systems can shift suddenly from consideration of one possibility to the other, based on models of chaotic attractors (Tsuda2001a).

2010 -
Grant Awardees - Program

Information processing by signal transduction and gene regulatory networks in mammalian cells

GUNAWARDENA Jeremy (UK)

Dept. of Systems Biology - Harvard Medical School - Boston - USA

HADJANTONAKIS Anna-Katerina (UK)

Developmental Biology Program - Sloan Kettering Institute - New York - USA

MARTINEZ-ARIAS Alfonso (SPAIN)

Dept. of Genetics, Lab. of the Structure and Function of Living Matter - University of Cambridge - Cambridge - UK

The aim of this project is to use mouse Embryonic Stem (ES) cells and preimplantation embryos, from which ES cells are derived, to understand how interactions between signal transduction pathways and gene regulatory networks regulate processes of cell fate assignment. We propose to take advantage of the operational simplicity of these developmental systems to test whether fate transitions are guided by digital additive genetic systems or by analog dynamical integrative devices. Specifically we shall focus on the emergence of the pluripotent epiblast (EPI) from a precursor population, the Inner Cell Mass (ICM). Ultimately we shall try to recreate this process in culture and thus determine the parameters that influence the decision to be one or the other cell type.

2010 -
Grant Awardees - Program

Optogenetics for small G-proteins and protein kinases in neuroscience

HAHN Klaus (USA)

Dept. of Pharmacology - University of North Carolina at Chapel Hill - Chapel Hill - USA

KASAI Haruo (JAPAN)

Lab. of Structual Physiology - Center for Disease Biology & Integrative Medicine - Tokyo - JAPAN

KUHLMAN Brian (USA)

Dept. of Biochemistry and Biophysics - University of North Carolina at Chapel Hill - Chapel Hill - USA

The brain is a constantly adapting organ, with connections between brain cells and the physiology of individual cells changing to reflect learning and life events. This dynamic reshaping is a critical element of human behavior and disease, but it has been difficult to study at the molecular level because, for most important ‘signaling’ molecules in brain cells, there are no tools to alter molecular dynamics in real time as changes in brain shape and interactions are taking place. In a recent development, Hahn lab demonstrated a new molecular engineering approach capable of rendering the activity of signaling molecules sensitive to light, and therefore subject to study during dynamic events. A protein fragment used by plants to turn their leaves towards the sun was engineered into a completely unrelated protein, a ‘small GTPase’ used by cells to control shape and movement. The fragment of light-sensitive plant protein could be used to control the activity of the GTPase. This enabled photomanipulation of protein activity in living cells, and was quite valuable in studies of metastasis and cell movement. Perhaps more importantly it promises a new general approach that can potentially make light sensitive versions of many molecules for their study in living cells. In this proposal, we combine the forces of a theoretician focused on understanding how protein structures can be altered to generate new function, Dr. Kuhlman, with the cell biologist and protein biochemist who developed the photoactivatable GTPase, Dr. Hahn. These two will devlope new caged proteins capable of shedding light on important and previously inaccessible questions in neurobiology. This will be accomplished via the development of generally applicable methods that can be used by other researchers to control protein activity with light. The selection of target proteins was guided by Dr. Kasai, a pioneer in the role of neuronal dynamics in brain function. He will use the new photoactivatable proteins to study brain tissues both in vitro and in vivo. The new effective PA-probes produced by our collaboration will have wide applications in various fields of biology, as these small GTPase are key to diverse cellular functions and diseases.

2010 -
Grant Awardees - Program

Nanoscale photoactivation and imaging of synaptic physiology

HELL Stefan (GERMANY)

Dept. of NanoBiophotonics - Max-Planck-Institute for Biophysical Chemistry - GOETTINGEN - GERMANY

LI Wen-Hong (USA)

Dept. of Cell Biology - University of Texas Southwestern Medical Center - Dallas - USA

NÄGERL Valentin (GERMANY)

Interdisciplinary Institute for Neuroscience - University of Bordeaux - Bordeaux - FRANCE

Synapses are highly complex and dynamic sites of intercellular communication, playing a pivotal role for learning and memory formation in the mammalian brain. As they are very small - usually smaller than what diffraction-limited light microscopy can resolve - and densely packed in light-scattering brain tissue, it has been extremely difficult to study their physiology in mechanistic terms. As a result, we still lack a clear understanding of the basic dynamic organization of neurotransmitter receptors and their molecular partners at mammalian synapses. While electron microscopy provided us with detailed snapshots of where glutamate receptors are located inside synapses, it does not convey dynamic information about their function. Therefore, a functional assay is required that non-invasively probes receptor localization and activation with nanoscale resolution. Since existing optical approaches such as 2-photon glutamate uncaging do not have sufficient spatial resolution, progress in this area relies on fundamental breakthroughs in live-cell-compatible techniques relying on focused visible light.

We seek to establish a new paradigm for controlling physiological processes, which reconciles instant non-invasive molecular control with nanoscale resolution in arbitrary locations inside a cell. Specifically, we will (1) engineer photosensitive compounds for superresolution-based photo-activation, (2) advance microscopy technology to concurrently activate and image beyond the diffraction limit, and (3) use this new methodology to probe synaptic physiology in brain slices with unprecedented resolution, addressing timely questions about the dynamic behavior of neurotransmitter receptors in synapses.

After breaking the diffraction barrier in imaging, diffraction-unlimited photomanipulation will be the next paradigmatic step in neuroscience methodology. Substantially departing from the state of the art, the project brings together three research groups working at the cutting edge of probe development, superresolution optics, and neuroscience in order to reach a goal that offers a huge potential for scientific breakthroughs. Beyond addressing key questions about the molecular physiology of synapses, the project will have a substantial impact in many other areas where the arbitrary release of molecules with nanometer spatial resolution will provide a paradigmatic shift.