Project acronym ADOS
Project AMPA Receptor Dynamic Organization and Synaptic transmission in health and disease
Researcher (PI) Daniel Georges Gustave Choquet
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS5, ERC-2013-ADG
Summary AMPA glutamate receptors (AMPAR) play key roles in information processing by the brain as they mediate nearly all fast excitatory synaptic transmission. Their spatio-temporal organization in the post synapse with respect to presynaptic glutamate release sites is a key determinant in synaptic transmission. The activity-dependent regulation of AMPAR organization is at the heart of synaptic plasticity processes underlying learning and memory. Dysfunction of synaptic transmission - hence AMPAR organization - is likely at the origin of a number of brain diseases.
Building on discoveries made during my past ERC grant, our new ground-breaking objective is to uncover the mechanisms that link synaptic transmission with the dynamic organization of AMPAR and associated proteins. For this aim, we have assembled a team of neurobiologists, computer scientists and chemists with a track record of collaboration. We will combine physiology, cellular and molecular neurobiology with development of novel quantitative imaging and biomolecular tools to probe the molecular dynamics that regulate synaptic transmission.
Live high content 3D SuperResolution Light Imaging (SRLI) combined with electron microscopy will allow unprecedented visualization of AMPAR organization in synapses at the scale of individual subunits up to the level of intact tissue. Simultaneous SRLI and electrophysiology will elucidate the intricate relations between dynamic AMPAR organization, trafficking and synaptic transmission. Novel peptide- and small protein-based probes used as protein-protein interaction reporters and modulators will be developed to image and directly interfere with synapse organization.
We will identify new processes that are fundamental to activity dependent modifications of synaptic transmission. We will apply the above findings to understand the causes of early cognitive deficits in models of neurodegenerative disorders and open new avenues of research for innovative therapies.
Summary
AMPA glutamate receptors (AMPAR) play key roles in information processing by the brain as they mediate nearly all fast excitatory synaptic transmission. Their spatio-temporal organization in the post synapse with respect to presynaptic glutamate release sites is a key determinant in synaptic transmission. The activity-dependent regulation of AMPAR organization is at the heart of synaptic plasticity processes underlying learning and memory. Dysfunction of synaptic transmission - hence AMPAR organization - is likely at the origin of a number of brain diseases.
Building on discoveries made during my past ERC grant, our new ground-breaking objective is to uncover the mechanisms that link synaptic transmission with the dynamic organization of AMPAR and associated proteins. For this aim, we have assembled a team of neurobiologists, computer scientists and chemists with a track record of collaboration. We will combine physiology, cellular and molecular neurobiology with development of novel quantitative imaging and biomolecular tools to probe the molecular dynamics that regulate synaptic transmission.
Live high content 3D SuperResolution Light Imaging (SRLI) combined with electron microscopy will allow unprecedented visualization of AMPAR organization in synapses at the scale of individual subunits up to the level of intact tissue. Simultaneous SRLI and electrophysiology will elucidate the intricate relations between dynamic AMPAR organization, trafficking and synaptic transmission. Novel peptide- and small protein-based probes used as protein-protein interaction reporters and modulators will be developed to image and directly interfere with synapse organization.
We will identify new processes that are fundamental to activity dependent modifications of synaptic transmission. We will apply the above findings to understand the causes of early cognitive deficits in models of neurodegenerative disorders and open new avenues of research for innovative therapies.
Max ERC Funding
2 491 157 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym C.NAPSE
Project TOWARDS A COMPREHENSIVE ANALYSIS OF EXTRACELLULAR SCAFFOLDING AT THE SYNAPSE
Researcher (PI) Jean-Louis BESSEREAU
Host Institution (HI) UNIVERSITE LYON 1 CLAUDE BERNARD
Call Details Advanced Grant (AdG), LS5, ERC-2015-AdG
Summary Synaptic scaffolding molecules control the localization and the abundance of neurotransmitter receptors at the synapse, a key parameter to shape synaptic transfer function. Most characterized synaptic scaffolds are intracellular, yet a growing number of secreted proteins appear to organize the synapse from the outside of the cell. We recently demonstrated in C. elegans that an evolutionarily conserved protein secreted by motoneurons specifies the excitatory versus inhibitory identity of the postsynaptic domains at neuromuscular synapses. We propose to use this system as a genetically tractable paradigm to perform a comprehensive characterization of this unforeseen synaptic organization.
Specifically, this project will pursue 4 complementary aims:
1) Identify and characterize a comprehensive set of genes that organize and control the formation and maintenance of these scaffolds through a series of genetic screens based on the direct visualization of fluorescent acetylcholine and GABA receptors in living animals.
2) Solve the spatial synaptic organization of these scaffolds at a nanoscale resolution using super-resolutive and correlative light and electron microscopy, and analyze their dynamic behavior in vivo by implementing Single Particle Tracking imaging in living worms.
3) Decipher the role of the synaptomatrix in the organization of synaptic extracellular scaffolds and evaluate its functional contribution at the physiological and molecular levels using a candidate gene strategy and innovative imaging.
4) Analyze the formation and decline of these scaffolds at the lifetime scale and evaluate the role of synaptic activity and aging in these processes by taking advantage of the possibility to follow identified synapses over the entire life of C. elegans.
Using powerful genetics in combination with cutting-edge in vivo imaging and electrophysiology, we anticipate to identify new genes and new mechanisms at work to regulate normal and pathological synaptic function.
Summary
Synaptic scaffolding molecules control the localization and the abundance of neurotransmitter receptors at the synapse, a key parameter to shape synaptic transfer function. Most characterized synaptic scaffolds are intracellular, yet a growing number of secreted proteins appear to organize the synapse from the outside of the cell. We recently demonstrated in C. elegans that an evolutionarily conserved protein secreted by motoneurons specifies the excitatory versus inhibitory identity of the postsynaptic domains at neuromuscular synapses. We propose to use this system as a genetically tractable paradigm to perform a comprehensive characterization of this unforeseen synaptic organization.
Specifically, this project will pursue 4 complementary aims:
1) Identify and characterize a comprehensive set of genes that organize and control the formation and maintenance of these scaffolds through a series of genetic screens based on the direct visualization of fluorescent acetylcholine and GABA receptors in living animals.
2) Solve the spatial synaptic organization of these scaffolds at a nanoscale resolution using super-resolutive and correlative light and electron microscopy, and analyze their dynamic behavior in vivo by implementing Single Particle Tracking imaging in living worms.
3) Decipher the role of the synaptomatrix in the organization of synaptic extracellular scaffolds and evaluate its functional contribution at the physiological and molecular levels using a candidate gene strategy and innovative imaging.
4) Analyze the formation and decline of these scaffolds at the lifetime scale and evaluate the role of synaptic activity and aging in these processes by taking advantage of the possibility to follow identified synapses over the entire life of C. elegans.
Using powerful genetics in combination with cutting-edge in vivo imaging and electrophysiology, we anticipate to identify new genes and new mechanisms at work to regulate normal and pathological synaptic function.
Max ERC Funding
2 492 750 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym CDK6-DrugOpp
Project CDK6 in transcription - turning a foe in a friend
Researcher (PI) Veronika SEXL
Host Institution (HI) VETERINAERMEDIZINISCHE UNIVERSITAET WIEN
Call Details Advanced Grant (AdG), LS7, ERC-2015-AdG
Summary "Translational research aims at applying mechanistic understanding in the development of "precision medicine", which depends on precise diagnostic tools and therapeutic approaches. Cancer therapy is experiencing a switch from non-specific, cytotoxic agents towards molecularly targeted and rationally designed compounds with the promise of greater efficacy and fewer side effects.
The two cell-cycle kinases CDK4 and CDK6 normally facilitate cell-cycle progression but are abnormally activated in certain cancers. CDK6 is up-regulated in hematopoietic malignancies, where it is the predominant cell-cycle kinase. The importance of CDK4/6 for tumor development is underscored by the fact that the US FDA selected inhibitors of the kinase activity of CDK4/6 as "breakthrough of the year 2013". Our recent findings suggest that the effects of the inhibitors may be limited as CDK6 is not only involved in cell-cycle progression: ground-breaking research in my group and others has shown that CDK6 is involved in regulation of transcription in a kinase-independent manner thereby driving the proliferation of leukemic stem cells and tumor formation. We have now identified mutations in CDK6 that convert it from a tumor promoter into a tumor suppressor. This unexpected outcome is accompanied by a distinct transcriptional profile. Separating the tumor-promoting from the tumor suppressive functions may open a novel therapeutic avenue for drug development. We aim at understanding which domains and residues of CDK6 are involved in rewiring the transcriptional landscape to pave the way for sophisticated inhibitors. The idea of turning a cancer cell's own most potent weapon against itself is novel and would represent a new paradigm for drug design. Finally, the understanding of CDK6 functions in tumor promotion and maintenance will also result in better patient stratification and improved treatment decisions for a broad spectrum of cancer types."
Summary
"Translational research aims at applying mechanistic understanding in the development of "precision medicine", which depends on precise diagnostic tools and therapeutic approaches. Cancer therapy is experiencing a switch from non-specific, cytotoxic agents towards molecularly targeted and rationally designed compounds with the promise of greater efficacy and fewer side effects.
The two cell-cycle kinases CDK4 and CDK6 normally facilitate cell-cycle progression but are abnormally activated in certain cancers. CDK6 is up-regulated in hematopoietic malignancies, where it is the predominant cell-cycle kinase. The importance of CDK4/6 for tumor development is underscored by the fact that the US FDA selected inhibitors of the kinase activity of CDK4/6 as "breakthrough of the year 2013". Our recent findings suggest that the effects of the inhibitors may be limited as CDK6 is not only involved in cell-cycle progression: ground-breaking research in my group and others has shown that CDK6 is involved in regulation of transcription in a kinase-independent manner thereby driving the proliferation of leukemic stem cells and tumor formation. We have now identified mutations in CDK6 that convert it from a tumor promoter into a tumor suppressor. This unexpected outcome is accompanied by a distinct transcriptional profile. Separating the tumor-promoting from the tumor suppressive functions may open a novel therapeutic avenue for drug development. We aim at understanding which domains and residues of CDK6 are involved in rewiring the transcriptional landscape to pave the way for sophisticated inhibitors. The idea of turning a cancer cell's own most potent weapon against itself is novel and would represent a new paradigm for drug design. Finally, the understanding of CDK6 functions in tumor promotion and maintenance will also result in better patient stratification and improved treatment decisions for a broad spectrum of cancer types."
Max ERC Funding
2 497 520 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym Dyn-Syn-Mem
Project Dynamic mechanisms and functional roles of synaptic plasticity in memory
Researcher (PI) Daniel Choquet
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS5, ERC-2017-ADG
Summary Activity-dependent plasticity of synaptic transmission together with refinement of neural circuits connectivity are amongst the core mechanisms underlying learning and memory. While there is already extensive knowledge on some of the mechanisms of synaptic plasticity, fundamental questions remain on the dynamics of the underlying molecular events and the functional roles of various forms of synaptic plasticity in information processing, learning and behavior.
We previously uncovered basic features of glutamate receptor movements and their role in excitatory synaptic transmission. Our new ground-breaking objectives are: 1) to uncover, in a physiological context, the dynamic mechanisms through which synapses modulate their strength in response to neuronal activity by integrating on different space and time scales the properties of receptor traffic pathways and associated stabilization mechanisms, 2) to use our knowledge and innovative tools to interfere with these trafficking mechanisms in order to decipher the specific roles of different forms of synaptic plasticity in given brain functions and behavioral tasks. For this aim, I lead a team of neurobiologists, physicists and chemists with a collaborative record of accomplishment. We will combine imaging, cellular neurobiology, physiology and behavior to probe the mechanisms and roles of different forms of synaptic plasticity.
New in tissue high-resolution imaging combined with innovative molecular reporters and electrophysiology will allow analysis of receptor traffic during short and long-term synaptic plasticity in physiological conditions. We will probe the interplay between activity-dependent changes in synaptic strength and circuit function with new photo-activable modifiers of receptor traffic with an unprecedented time and space resolution. Use of these tools in vivo will allow identifying the roles of synaptic plasticity in sensory information processing and the various phases of spatial memory formation.
Summary
Activity-dependent plasticity of synaptic transmission together with refinement of neural circuits connectivity are amongst the core mechanisms underlying learning and memory. While there is already extensive knowledge on some of the mechanisms of synaptic plasticity, fundamental questions remain on the dynamics of the underlying molecular events and the functional roles of various forms of synaptic plasticity in information processing, learning and behavior.
We previously uncovered basic features of glutamate receptor movements and their role in excitatory synaptic transmission. Our new ground-breaking objectives are: 1) to uncover, in a physiological context, the dynamic mechanisms through which synapses modulate their strength in response to neuronal activity by integrating on different space and time scales the properties of receptor traffic pathways and associated stabilization mechanisms, 2) to use our knowledge and innovative tools to interfere with these trafficking mechanisms in order to decipher the specific roles of different forms of synaptic plasticity in given brain functions and behavioral tasks. For this aim, I lead a team of neurobiologists, physicists and chemists with a collaborative record of accomplishment. We will combine imaging, cellular neurobiology, physiology and behavior to probe the mechanisms and roles of different forms of synaptic plasticity.
New in tissue high-resolution imaging combined with innovative molecular reporters and electrophysiology will allow analysis of receptor traffic during short and long-term synaptic plasticity in physiological conditions. We will probe the interplay between activity-dependent changes in synaptic strength and circuit function with new photo-activable modifiers of receptor traffic with an unprecedented time and space resolution. Use of these tools in vivo will allow identifying the roles of synaptic plasticity in sensory information processing and the various phases of spatial memory formation.
Max ERC Funding
2 499 505 €
Duration
Start date: 2019-02-01, End date: 2024-01-31
Project acronym EnergyMemo
Project Dynamic Interplay between Energy and Memory
Researcher (PI) Thomas Jules Henri PREAT
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS5, ERC-2016-ADG
Summary Understanding the links between neuronal plasticity which underlies memory and energy metabolism is a major goal of brain studies. The brain is a main energy consumer and the central regulator of energy homeostasis, and it prioritizes its own supply over peripheral organs. Interestingly, our work demonstrates that the brain is also able to regulate its own activity under energy shortage to favor survival.
The EnergyMemo project proposes to perform in drosophila an original integrated study of the interplay between energy metabolism and olfactory memory at the molecular, cellular and circuit levels. On the ground of important preliminary results, we will investigate in vivo how and why the energy flux increases during long-term memory encoding, and how brain plasticity is regulated by the energy supply. We will focus on three major challenges:
* Objective 1: to improve our understanding of brain physiology, we will characterize in drosophila neuronal circuits that integrate information about the brain energy status.
* Objective 2: to understand how abnormal levels of energy can affect the brain, we will analyze how the energy level shapes the functioning of the olfactory memory center.
* Objective 3: to characterize how energy stores are mobilized during memory formation, we will investigate how the neuronal and glial networks interact to manage the energy fluxes.
This multidisciplinary project will benefit from our team's longstanding experience in behavioral studies and leadership in live brain imaging, in addition to the unmatched descriptive power of drosophila neuronal circuits at the single-neuron resolution. Successful completion of this program will surely uncover mechanisms of brain function conserved across species, and should bring-up new ideas about how deregulation of energy metabolism can affect cognitive functions in human. Thus the EnergyMemo project could have a major impact in neuroscience from fundamental research to human applications.
Summary
Understanding the links between neuronal plasticity which underlies memory and energy metabolism is a major goal of brain studies. The brain is a main energy consumer and the central regulator of energy homeostasis, and it prioritizes its own supply over peripheral organs. Interestingly, our work demonstrates that the brain is also able to regulate its own activity under energy shortage to favor survival.
The EnergyMemo project proposes to perform in drosophila an original integrated study of the interplay between energy metabolism and olfactory memory at the molecular, cellular and circuit levels. On the ground of important preliminary results, we will investigate in vivo how and why the energy flux increases during long-term memory encoding, and how brain plasticity is regulated by the energy supply. We will focus on three major challenges:
* Objective 1: to improve our understanding of brain physiology, we will characterize in drosophila neuronal circuits that integrate information about the brain energy status.
* Objective 2: to understand how abnormal levels of energy can affect the brain, we will analyze how the energy level shapes the functioning of the olfactory memory center.
* Objective 3: to characterize how energy stores are mobilized during memory formation, we will investigate how the neuronal and glial networks interact to manage the energy fluxes.
This multidisciplinary project will benefit from our team's longstanding experience in behavioral studies and leadership in live brain imaging, in addition to the unmatched descriptive power of drosophila neuronal circuits at the single-neuron resolution. Successful completion of this program will surely uncover mechanisms of brain function conserved across species, and should bring-up new ideas about how deregulation of energy metabolism can affect cognitive functions in human. Thus the EnergyMemo project could have a major impact in neuroscience from fundamental research to human applications.
Max ERC Funding
2 499 500 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym EPIC
Project Enabling Precision Immuno-oncology in Colorectal cancer
Researcher (PI) Zlatko TRAJANOSKI
Host Institution (HI) MEDIZINISCHE UNIVERSITAT INNSBRUCK
Call Details Advanced Grant (AdG), LS7, ERC-2017-ADG
Summary Immunotherapy with checkpoints blockers is transforming the treatment of advanced cancers. Colorectal cancer (CRC), a cancer with 1.4 million new cases diagnosed annually worldwide, is refractory to immunotherapy (with the exception of a minority of tumors with microsatellite instability). This is somehow paradoxical as CRC is a cancer for which we have shown that it is under immunological control and that tumor infiltrating lymphocytes represent a strong independent predictor of survival. Thus, there is an urgent need to broaden the clinical benefits of immune checkpoint blockers to CRC by combining agents with synergistic mechanisms of action. An attractive approach to sensitize tumors to immunotherapy is to harness immunogenic effects induced by approved conventional or targeted agents.
Here I propose a new paradigm to identify molecular determinants of resistance to immunotherapy and develop personalized in silico and in vitro models for predicting response to combination therapy in CRC. The EPIC concept is based on three pillars: 1) emphasis on antitumor T cell activity; 2) systematic interrogation of tumor-immune cell interactions using data-driven modeling and knowledge-based mechanistic modeling, and 3) generation of key quantitative data to train and validate algorithms using perturbation experiments with patient-derived tumor organoids and cutting-edge technologies for multidimensional profiling. We will investigate three immunomodulatory processes: 1) immunostimulatory effects of chemotherapeutics, 2) rewiring of signaling networks induced by targeted drugs and their interference with immunity, and 3) metabolic reprogramming of T cells to enhance antitumor immunity.
The anticipated outcome of EPIC is a precision immuno-oncology platform that integrates tumor organoids with high-throughput and high-content data for testing drug combinations, and machine learning for making therapeutic recommendations for individual patients.
Summary
Immunotherapy with checkpoints blockers is transforming the treatment of advanced cancers. Colorectal cancer (CRC), a cancer with 1.4 million new cases diagnosed annually worldwide, is refractory to immunotherapy (with the exception of a minority of tumors with microsatellite instability). This is somehow paradoxical as CRC is a cancer for which we have shown that it is under immunological control and that tumor infiltrating lymphocytes represent a strong independent predictor of survival. Thus, there is an urgent need to broaden the clinical benefits of immune checkpoint blockers to CRC by combining agents with synergistic mechanisms of action. An attractive approach to sensitize tumors to immunotherapy is to harness immunogenic effects induced by approved conventional or targeted agents.
Here I propose a new paradigm to identify molecular determinants of resistance to immunotherapy and develop personalized in silico and in vitro models for predicting response to combination therapy in CRC. The EPIC concept is based on three pillars: 1) emphasis on antitumor T cell activity; 2) systematic interrogation of tumor-immune cell interactions using data-driven modeling and knowledge-based mechanistic modeling, and 3) generation of key quantitative data to train and validate algorithms using perturbation experiments with patient-derived tumor organoids and cutting-edge technologies for multidimensional profiling. We will investigate three immunomodulatory processes: 1) immunostimulatory effects of chemotherapeutics, 2) rewiring of signaling networks induced by targeted drugs and their interference with immunity, and 3) metabolic reprogramming of T cells to enhance antitumor immunity.
The anticipated outcome of EPIC is a precision immuno-oncology platform that integrates tumor organoids with high-throughput and high-content data for testing drug combinations, and machine learning for making therapeutic recommendations for individual patients.
Max ERC Funding
2 460 500 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym FRONTEX
Project Decision-making and prefrontal executive function
Researcher (PI) Etienne Koechlin
Host Institution (HI) ECOLE NORMALE SUPERIEURE
Call Details Advanced Grant (AdG), LS5, ERC-2009-AdG
Summary The prefrontal cortex (PFC) subserves decision-making and executive control, i.e. the ability to make decisions and to regulate behavior according to external events, mental models of situations, internal drives and subjective preferences. Our overall aim is to understand the functional architecture of the human PFC and computational mechanisms of PFC function. The PFC function is known to operate along three major dimensions, namely the affective, motivational and cognitive control of action subserved by the orbital, medial and lateral sectors of the PFC, respectively. In this project, our specific objectives are to solve the following three open issues of critical theoretical significance: (1) the functional organization of motivational control in the medial prefrontal cortex; (2) the mechanisms that enables the PFC to control the learning of representational sets required for cognitive control; (3) the functional interactions between the medial and lateral prefrontal cortex, i.e. the integration of motivational and cognitive control into a unitary decision-making and control system. We will address these theoretically and methodologically challenging issues by elaborating computational models that integrate learning and control mechanisms, and in relation to these models, by conducting functional magnetic resonance imaging experiments in healthy humans. The project is expected to significantly improve our knowledge of the human PFC function. This basic project has potential major implications especially in medicine, because alterations of the prefrontal function is observed in aging and most neuropsychiatric diseases, as well as in technology for developing artificial and robotics intelligence with human-like adaptive reasoning and decision-making abilities.
Summary
The prefrontal cortex (PFC) subserves decision-making and executive control, i.e. the ability to make decisions and to regulate behavior according to external events, mental models of situations, internal drives and subjective preferences. Our overall aim is to understand the functional architecture of the human PFC and computational mechanisms of PFC function. The PFC function is known to operate along three major dimensions, namely the affective, motivational and cognitive control of action subserved by the orbital, medial and lateral sectors of the PFC, respectively. In this project, our specific objectives are to solve the following three open issues of critical theoretical significance: (1) the functional organization of motivational control in the medial prefrontal cortex; (2) the mechanisms that enables the PFC to control the learning of representational sets required for cognitive control; (3) the functional interactions between the medial and lateral prefrontal cortex, i.e. the integration of motivational and cognitive control into a unitary decision-making and control system. We will address these theoretically and methodologically challenging issues by elaborating computational models that integrate learning and control mechanisms, and in relation to these models, by conducting functional magnetic resonance imaging experiments in healthy humans. The project is expected to significantly improve our knowledge of the human PFC function. This basic project has potential major implications especially in medicine, because alterations of the prefrontal function is observed in aging and most neuropsychiatric diseases, as well as in technology for developing artificial and robotics intelligence with human-like adaptive reasoning and decision-making abilities.
Max ERC Funding
2 500 000 €
Duration
Start date: 2010-05-01, End date: 2016-04-30
Project acronym FRU CIRCUIT
Project Neural basis of Drosophila mating behaviours
Researcher (PI) Barry Dickson
Host Institution (HI) FORSCHUNGSINSTITUT FUR MOLEKULARE PATHOLOGIE GESELLSCHAFT MBH
Call Details Advanced Grant (AdG), LS5, ERC-2008-AdG
Summary How does information processing in neural circuits generate behaviour? Answering this question requires identifying each of the distinct neuronal types that contributes to a behaviour, defining their anatomy and connectivity, and establishing causal relationships between their activity, the activity of other neurons in the circuit, and the behaviour. Here, I propose such an analysis of the neural circuits that guide Drosophila mating behaviours. The distinct mating behaviours of males and females are genetically pre-programmed, yet can also be modified by experience. The set of ~2000 neurons that express the fru gene have been intimately linked to both male and female mating behaviours. This set of neurons includes specific sensory, central, and motor neurons, at least some of which are directly connected. Male-specific fruM isoforms configure this circuit developmentally for male rather than female behaviour. In females, mating triggers a biochemical cascade that reconfigures the circuit for post-mating rather than virgin female behaviour. We estimate that there are ~100 distinct classes of fru neuron. Using genetic and optical tools, we aim to identify each distinct class of fru neuron and to define its anatomy and connectivity. By silencing or activating specific neurons, or changing their genetic sex, we will assess their contributions to male and female behaviours, and how these perturbations impinge on activity patterns in other fru neurons. We also aim to define how a specific experience can modify the physiological properties of these circuits, and how these changes in turn modulate mating behaviour. These studies will define the operating principles of these neural circuits, contributing to a molecules-to-systems explanation of Drosophila s mating behaviours.
Summary
How does information processing in neural circuits generate behaviour? Answering this question requires identifying each of the distinct neuronal types that contributes to a behaviour, defining their anatomy and connectivity, and establishing causal relationships between their activity, the activity of other neurons in the circuit, and the behaviour. Here, I propose such an analysis of the neural circuits that guide Drosophila mating behaviours. The distinct mating behaviours of males and females are genetically pre-programmed, yet can also be modified by experience. The set of ~2000 neurons that express the fru gene have been intimately linked to both male and female mating behaviours. This set of neurons includes specific sensory, central, and motor neurons, at least some of which are directly connected. Male-specific fruM isoforms configure this circuit developmentally for male rather than female behaviour. In females, mating triggers a biochemical cascade that reconfigures the circuit for post-mating rather than virgin female behaviour. We estimate that there are ~100 distinct classes of fru neuron. Using genetic and optical tools, we aim to identify each distinct class of fru neuron and to define its anatomy and connectivity. By silencing or activating specific neurons, or changing their genetic sex, we will assess their contributions to male and female behaviours, and how these perturbations impinge on activity patterns in other fru neurons. We also aim to define how a specific experience can modify the physiological properties of these circuits, and how these changes in turn modulate mating behaviour. These studies will define the operating principles of these neural circuits, contributing to a molecules-to-systems explanation of Drosophila s mating behaviours.
Max ERC Funding
2 492 164 €
Duration
Start date: 2009-07-01, End date: 2013-09-30
Project acronym FUSIMAGINE
Project A new neuroimaging modality: from bench to bedside
Researcher (PI) Mickaël Tanter
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary "NeuroImaging systems are invaluable tools in the understanding of the brain both for fundamental research and clinical diagnosis. However, recent improvements in deep brain imaging technology have been somewhat limited because most of them are based on incremental innovation of mature techniques (EEG, PET and fMRI) instead of breakthrough.
In FUSIMAGINE, a genuinely new functional brain imaging modality will be developed and validated whose performances could have a major impact in neuroscience from fundamental research to clinical applications.
This new modality is based on the use of ultrafast ultrasound scanners able to reach more than 10 000 frames per second (fps) compared to the usual 50 fps in conventional ultrasound scanners. This concept relies on compounded plane wave transmissions introduced by my team and demonstrates up to 100-fold increase in the sensitivity of blood flow measurements. It enables to image the subtle hemodynamic changes in small brain vessels and thus brain activity thanks to neurovascular coupling. Functional Ultrasound (fUS by analogy to fMRI) is a real breakthrough in brain imaging as our project will demonstrate that:
in neuroscience, fUS provides a unique real time, portable and deep brain functional imaging technique for awake and even freely moving small animal imaging, moreover with unprecedented spatiotemporal resolution (~100µm, 50ms).
in clinical diagnosis, fUS provides a unique bedside neuro-imaging system of newborns brain activity through the fontanel window. Such real time system will permit to monitor and better understand neonatal seizures and hemorrhages. On adults, fUS provides a unique functional imaging modality during neurosurgery to predict the cortical mapping remodeling resulting of tumor development (such as low-grade gliomas). Finally, new adaptive skull bone correction techniques implemented on the system will enable us to perform non invasive transcranial fUS imaging on human adults through the temple bone."
Summary
"NeuroImaging systems are invaluable tools in the understanding of the brain both for fundamental research and clinical diagnosis. However, recent improvements in deep brain imaging technology have been somewhat limited because most of them are based on incremental innovation of mature techniques (EEG, PET and fMRI) instead of breakthrough.
In FUSIMAGINE, a genuinely new functional brain imaging modality will be developed and validated whose performances could have a major impact in neuroscience from fundamental research to clinical applications.
This new modality is based on the use of ultrafast ultrasound scanners able to reach more than 10 000 frames per second (fps) compared to the usual 50 fps in conventional ultrasound scanners. This concept relies on compounded plane wave transmissions introduced by my team and demonstrates up to 100-fold increase in the sensitivity of blood flow measurements. It enables to image the subtle hemodynamic changes in small brain vessels and thus brain activity thanks to neurovascular coupling. Functional Ultrasound (fUS by analogy to fMRI) is a real breakthrough in brain imaging as our project will demonstrate that:
in neuroscience, fUS provides a unique real time, portable and deep brain functional imaging technique for awake and even freely moving small animal imaging, moreover with unprecedented spatiotemporal resolution (~100µm, 50ms).
in clinical diagnosis, fUS provides a unique bedside neuro-imaging system of newborns brain activity through the fontanel window. Such real time system will permit to monitor and better understand neonatal seizures and hemorrhages. On adults, fUS provides a unique functional imaging modality during neurosurgery to predict the cortical mapping remodeling resulting of tumor development (such as low-grade gliomas). Finally, new adaptive skull bone correction techniques implemented on the system will enable us to perform non invasive transcranial fUS imaging on human adults through the temple bone."
Max ERC Funding
2 497 603 €
Duration
Start date: 2014-05-01, End date: 2019-04-30
Project acronym GENE FOR CURE
Project Expanding and extending gene therapy of monogenic diseases of the haematopoietic system
Researcher (PI) Marina Cavazzana
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2015-AdG
Summary Given that (i) not all patients with a monogenic disease affecting the lymphohaematopoietic system have an HLA-genoidentical sibling donor and (ii) severe immunological complications worsen the outcome in HLA-partially-matched hematopoietic stem cell transplantation (HSCT), the genetic modification of autologous hematopoietic stem cells (HSCs) has become a powerful curative treatment option for these individuals. The present project seeks to further consolidate the rationale for replacing HLA-partially-matched HSCT with a gene therapy approach. Wiskott-Aldrich syndrome is a primary immunodeficiency whose severity is due to impairment of both lymphoid and myeloid cell function. We have reported robust evidence showing that the infusion of gene-corrected autologous HSCs enables the restoration of the T cell function. However, we are still cautious with regard to claiming the stable correction of autoimmunity and thrombocytopenia. Accordingly, we plan to thoroughly monitor long-term B cell functional outcomes and the platelet count in our treated patients. Moreover, we wish to extend the gene therapy approach to the SCID caused by mutations in the DLREIC gene, since the long-term post-HSCT outcomes are particularly poor. The preclinical work has been completed; initiation of a clinical protocol is the next step. Immune-dysregulation polyendocrinopathy enteropathy X-linked (IPEX) and sickle cell anaemia (SCA) are the last two target diseases. IPEX is a devastating disease caused by mutation of FOXP3 transcription factor; it may be possible to correct it by infusing gene-modified CD4+Tcells. We intend to accumulate the data required to prove our working hypothesis. SCA is a worldwide public health issue. We are seeking to improve the conventional gene therapy procedure and to evaluate the efficacy of CrisP/Cas9-mediated disruption of the CIS-regulatory elements required for HbF silencing. This disruption may provide a cure for SCA.
Summary
Given that (i) not all patients with a monogenic disease affecting the lymphohaematopoietic system have an HLA-genoidentical sibling donor and (ii) severe immunological complications worsen the outcome in HLA-partially-matched hematopoietic stem cell transplantation (HSCT), the genetic modification of autologous hematopoietic stem cells (HSCs) has become a powerful curative treatment option for these individuals. The present project seeks to further consolidate the rationale for replacing HLA-partially-matched HSCT with a gene therapy approach. Wiskott-Aldrich syndrome is a primary immunodeficiency whose severity is due to impairment of both lymphoid and myeloid cell function. We have reported robust evidence showing that the infusion of gene-corrected autologous HSCs enables the restoration of the T cell function. However, we are still cautious with regard to claiming the stable correction of autoimmunity and thrombocytopenia. Accordingly, we plan to thoroughly monitor long-term B cell functional outcomes and the platelet count in our treated patients. Moreover, we wish to extend the gene therapy approach to the SCID caused by mutations in the DLREIC gene, since the long-term post-HSCT outcomes are particularly poor. The preclinical work has been completed; initiation of a clinical protocol is the next step. Immune-dysregulation polyendocrinopathy enteropathy X-linked (IPEX) and sickle cell anaemia (SCA) are the last two target diseases. IPEX is a devastating disease caused by mutation of FOXP3 transcription factor; it may be possible to correct it by infusing gene-modified CD4+Tcells. We intend to accumulate the data required to prove our working hypothesis. SCA is a worldwide public health issue. We are seeking to improve the conventional gene therapy procedure and to evaluate the efficacy of CrisP/Cas9-mediated disruption of the CIS-regulatory elements required for HbF silencing. This disruption may provide a cure for SCA.
Max ERC Funding
2 445 268 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym GIANTSYN
Project Biophysics and circuit function of a giant cortical glutamatergic synapse
Researcher (PI) Peter Jonas
Host Institution (HI) INSTITUTE OF SCIENCE AND TECHNOLOGYAUSTRIA
Call Details Advanced Grant (AdG), LS5, ERC-2015-AdG
Summary A fundamental question in neuroscience is how the biophysical properties of synapses shape higher network
computations. The hippocampal mossy fiber synapse, formed between axons of dentate gyrus granule cells
and dendrites of CA3 pyramidal neurons, is the ideal synapse to address this question. This synapse is accessible
to presynaptic recording, due to its large size, allowing a rigorous investigation of the biophysical
mechanisms of transmission and plasticity. Furthermore, this synapse is placed in the center of a memory
circuit, and several hypotheses about its network function have been generated. However, even basic properties
of this key communication element remain enigmatic. The ambitious goal of the current proposal, GIANTSYN,
is to understand the hippocampal mossy fiber synapse at all levels of complexity. At the subcellular
level, we want to elucidate the biophysical mechanisms of transmission and synaptic plasticity in the
same depth as previously achieved at peripheral and brainstem synapses, classical synaptic models. At the
network level, we want to unravel the connectivity rules and the in vivo network function of this synapse,
particularly its role in learning and memory. To reach these objectives, we will combine functional and
structural approaches. For the analysis of synaptic transmission and plasticity, we will combine direct preand
postsynaptic patch-clamp recording and high-pressure freezing electron microscopy. For the analysis of
connectivity and network function, we will use transsynaptic labeling and in vivo electrophysiology. Based
on the proposed interdisciplinary research, the hippocampal mossy fiber synapse could become the first synapse
in the history of neuroscience in which we reach complete insight into both synaptic biophysics and
network function. In the long run, the results may open new perspectives for the diagnosis and treatment of
brain diseases in which mossy fiber transmission, plasticity, or connectivity are impaired.
Summary
A fundamental question in neuroscience is how the biophysical properties of synapses shape higher network
computations. The hippocampal mossy fiber synapse, formed between axons of dentate gyrus granule cells
and dendrites of CA3 pyramidal neurons, is the ideal synapse to address this question. This synapse is accessible
to presynaptic recording, due to its large size, allowing a rigorous investigation of the biophysical
mechanisms of transmission and plasticity. Furthermore, this synapse is placed in the center of a memory
circuit, and several hypotheses about its network function have been generated. However, even basic properties
of this key communication element remain enigmatic. The ambitious goal of the current proposal, GIANTSYN,
is to understand the hippocampal mossy fiber synapse at all levels of complexity. At the subcellular
level, we want to elucidate the biophysical mechanisms of transmission and synaptic plasticity in the
same depth as previously achieved at peripheral and brainstem synapses, classical synaptic models. At the
network level, we want to unravel the connectivity rules and the in vivo network function of this synapse,
particularly its role in learning and memory. To reach these objectives, we will combine functional and
structural approaches. For the analysis of synaptic transmission and plasticity, we will combine direct preand
postsynaptic patch-clamp recording and high-pressure freezing electron microscopy. For the analysis of
connectivity and network function, we will use transsynaptic labeling and in vivo electrophysiology. Based
on the proposed interdisciplinary research, the hippocampal mossy fiber synapse could become the first synapse
in the history of neuroscience in which we reach complete insight into both synaptic biophysics and
network function. In the long run, the results may open new perspectives for the diagnosis and treatment of
brain diseases in which mossy fiber transmission, plasticity, or connectivity are impaired.
Max ERC Funding
2 677 500 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym GT-SKIN
Project Gene Therapy for Inherited Skin Adhesion Disorders
Researcher (PI) Fulvio Mavilio
Host Institution (HI) ASSOCIATION GENETHON
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary Epidermolysis bullosa (EB) is a family of severe skin adhesion defects characterized by disfiguring blistering, infections, visual impairment, and a high risk of cancer. We showed through pre-clinical and clinical investigation that transplantation of genetically corrected epithelia leads to long-term functional correction of EB. The current vectors, however, integrate in an uncontrolled fashion in the human genome, a cause of genotoxicity and potentially severe adverse events. The objective of this project is the development and pre-clinical evaluation of new gene targeting and gene correction technology, aimed at integrating therapeutic transgenes at specific loci or at correcting genetic defects by homologous recombination. Epidermal stem cells (EpSCs) and EB are ideal targets for the design and development of such technology, which is expected to have an impact on gene therapy of many other genetic diseases. We will design and test novel viral vectors for homologous recombination based on site-specific integrases and Zn-finger nucleases, and non-viral vectors for integrating large genes and complex regulatory sequences based on vertebrate transposons. All vectors will be tested in repopulating human EpSCs in vitro and in vivo, in a pre-clinical model of xenotransplantation of human skin on immunodeficient mice. A parallel, basic research program will aim at applying existing and novel genomic approaches to the definition of transcription factors, regulatory regions and gene expression programs involved in self renewal, commitment and differentiation of EpSCs. The anticipated output of the project is the development of next-generation technology for gene transfer, and the establishment of a knowledge base for a better utilization of EpSCs in gene therapy.
Summary
Epidermolysis bullosa (EB) is a family of severe skin adhesion defects characterized by disfiguring blistering, infections, visual impairment, and a high risk of cancer. We showed through pre-clinical and clinical investigation that transplantation of genetically corrected epithelia leads to long-term functional correction of EB. The current vectors, however, integrate in an uncontrolled fashion in the human genome, a cause of genotoxicity and potentially severe adverse events. The objective of this project is the development and pre-clinical evaluation of new gene targeting and gene correction technology, aimed at integrating therapeutic transgenes at specific loci or at correcting genetic defects by homologous recombination. Epidermal stem cells (EpSCs) and EB are ideal targets for the design and development of such technology, which is expected to have an impact on gene therapy of many other genetic diseases. We will design and test novel viral vectors for homologous recombination based on site-specific integrases and Zn-finger nucleases, and non-viral vectors for integrating large genes and complex regulatory sequences based on vertebrate transposons. All vectors will be tested in repopulating human EpSCs in vitro and in vivo, in a pre-clinical model of xenotransplantation of human skin on immunodeficient mice. A parallel, basic research program will aim at applying existing and novel genomic approaches to the definition of transcription factors, regulatory regions and gene expression programs involved in self renewal, commitment and differentiation of EpSCs. The anticipated output of the project is the development of next-generation technology for gene transfer, and the establishment of a knowledge base for a better utilization of EpSCs in gene therapy.
Max ERC Funding
2 210 000 €
Duration
Start date: 2011-12-01, End date: 2017-05-31
Project acronym HAIRBUNDLE
Project Assembling the puzzle of the operating auditory hair bundle
Researcher (PI) Christine Petit
Host Institution (HI) INSTITUT PASTEUR
Call Details Advanced Grant (AdG), LS5, ERC-2011-ADG_20110310
Summary The hair bundles of the sensory cells play a central role in the processing of sounds by the cochlea. Various performances of auditory perception depend upon what biophysical and physiological environment the operating hair bundle can provide to its mechanoelectrical transduction (MET) channels. The project aims at building a more integrated view of the way the hair bundle works. Reaching this goal, requires to understand the interdependence of its functional building blocks and their dynamic interplay.
To this purpose, the objectives of the work are :
1. To assemble components of the basal MET machinery
The work aims at
a) identifying the molecular players of the basal MET machinery, most of which are still unknown
b) deciphering their functional interactions,
c) developing a simple and efficient tool to validate candidate components of the MET machinery.
2. To elucidate the coupling between MET and stereocilia F-actin polymerisation
The work aims at
a) characterising this coupling, both in development and in steady-state condition,
b) deciphering the role of tip-link tension and Ca+2 influx through the MET channel,
c) developing a coarse-grained mathematical model of this coupling, experimentally testable.
3. To understand the interplay between MET, waveform distortions, and masking
The work aims at
a) deciphering biochemical characteristics of the top connectors,
b) explaining why large waveform distortions vanish in the absence of top connectors,
c) determining how the operating outer hair cell (OHC) hair bundle contributes to the masking effect in auditory perception.
The hair bundle is a highly vulnerable structure, actually the main target structure of noise-induced hearing loss and hereditary deafness forms. As such, advances in the understanding of the mechanisms underlying its functioning will pave the way for the development of therapeutic approaches.
Summary
The hair bundles of the sensory cells play a central role in the processing of sounds by the cochlea. Various performances of auditory perception depend upon what biophysical and physiological environment the operating hair bundle can provide to its mechanoelectrical transduction (MET) channels. The project aims at building a more integrated view of the way the hair bundle works. Reaching this goal, requires to understand the interdependence of its functional building blocks and their dynamic interplay.
To this purpose, the objectives of the work are :
1. To assemble components of the basal MET machinery
The work aims at
a) identifying the molecular players of the basal MET machinery, most of which are still unknown
b) deciphering their functional interactions,
c) developing a simple and efficient tool to validate candidate components of the MET machinery.
2. To elucidate the coupling between MET and stereocilia F-actin polymerisation
The work aims at
a) characterising this coupling, both in development and in steady-state condition,
b) deciphering the role of tip-link tension and Ca+2 influx through the MET channel,
c) developing a coarse-grained mathematical model of this coupling, experimentally testable.
3. To understand the interplay between MET, waveform distortions, and masking
The work aims at
a) deciphering biochemical characteristics of the top connectors,
b) explaining why large waveform distortions vanish in the absence of top connectors,
c) determining how the operating outer hair cell (OHC) hair bundle contributes to the masking effect in auditory perception.
The hair bundle is a highly vulnerable structure, actually the main target structure of noise-induced hearing loss and hereditary deafness forms. As such, advances in the understanding of the mechanisms underlying its functioning will pave the way for the development of therapeutic approaches.
Max ERC Funding
2 495 000 €
Duration
Start date: 2012-12-01, End date: 2018-08-31
Project acronym HEP
Project Epilepsies of the temporal lobe: emergence,
basal state and paroxysmal transitions
Researcher (PI) Richard Miles
Host Institution (HI) INSTITUT DU CERVEAU ET DE LA MOELLE EPINIERE
Call Details Advanced Grant (AdG), LS5, ERC-2012-ADG_20120314
Summary This proposal focuses on a human disease: epilepsy of the temporal lobe. This syndrome has a relatively stereotyped history. Focal seizures emerge, with a delay of several years in the human, after an initial insult that results in neuronal death. We will use physiology, imaging, anatomy and transcriptomic techniques to ask how an epileptic brain emerges, to define different neuronal cell types in the basal epileptic state and to examine factors associated with the paroxysmal transition to a seizure. Specifically we will ask whether a loss of cholesterol homeostasis is involved in the initial sclerotic neuronal death. We will ask whether proteoglycans deposited in the extracellular space as a generalised wound healing response force the establishment of aberrant synaptic contacts and so facilitate the slow process of epileptogenesis. We will then study the basal state of an epileptic brain. We will establish the physiology, anatomy, connectivity and transcriptome of functionally distinct neurons defined by different contributions to epileptiform activities in human epileptic brain slices. Finally we will seek to establish what sudden changes in field potential, ionic homeostasis, cellular firing and synaptic interactions are associated with the paroxysmal moment of transition to seizure.
Summary
This proposal focuses on a human disease: epilepsy of the temporal lobe. This syndrome has a relatively stereotyped history. Focal seizures emerge, with a delay of several years in the human, after an initial insult that results in neuronal death. We will use physiology, imaging, anatomy and transcriptomic techniques to ask how an epileptic brain emerges, to define different neuronal cell types in the basal epileptic state and to examine factors associated with the paroxysmal transition to a seizure. Specifically we will ask whether a loss of cholesterol homeostasis is involved in the initial sclerotic neuronal death. We will ask whether proteoglycans deposited in the extracellular space as a generalised wound healing response force the establishment of aberrant synaptic contacts and so facilitate the slow process of epileptogenesis. We will then study the basal state of an epileptic brain. We will establish the physiology, anatomy, connectivity and transcriptome of functionally distinct neurons defined by different contributions to epileptiform activities in human epileptic brain slices. Finally we will seek to establish what sudden changes in field potential, ionic homeostasis, cellular firing and synaptic interactions are associated with the paroxysmal moment of transition to seizure.
Max ERC Funding
1 985 057 €
Duration
Start date: 2013-10-01, End date: 2018-09-30
Project acronym HEPCENT
Project Molecular Analysis of Hepatitis C Virus Neutralization and Entry For the Development of Novel Antiviral Immunopreventive Strategies
Researcher (PI) François-Loic Cosset
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2008-AdG
Summary Hepatitis C virus (HCV) infection is a leading cause of chronic liver disease world-wide. HCV-induced end-stage liver disease such as liver cirrhosis and hepatocellular carcinoma represent a major concern in global health. Treatment options for chronic hepatitis C are limited and no vaccine against HCV infection is available. Vaccine development is hampered by several obstacles. High viral variability and escape from host immune responses render antigen selection a major challenge. Antigen selection requires thorough studies to identify conserved T cell and neutralization epitopes and to decipher neutralization mechanisms, aiming to discover the optimal viral target for immune responses counteracting HCV escape strategies. At the same time it is important to develop antigen presentation systems that are efficient in patients with impaired antiviral immune responses, as often observed during chronic hepatitis C. While most vaccine development programs are based on improving HCV cellular immunity, it is essential to associate, in a same vaccine formulation, immunogens able to induce broad spectrums neutralizing and cellular responses. Owing to recent progresses in the field, here we propose a project aiming to overcome the current limitations in vaccine development by addressing the improvement of B cell responses targeting HCV infection. This will be achieved by a detailed investigation of: 1) mechanisms of antibody-mediated neutralization and escape, 2) impact of lipoproteins associating with the viral particle during assembly/release and counteracting neutralization and 3) cell entry steps that can potentially be targeted by antibodies, including those that are not induced naturally. Thus, through the combined expertise of the team in molecular virology, immunology, clinical hepatology and vectorology, we aim to rationalize the development of B cell immunogens and neutralizing antibodies for novel antiviral immunopreventive strategies targeting HCV infection.
Summary
Hepatitis C virus (HCV) infection is a leading cause of chronic liver disease world-wide. HCV-induced end-stage liver disease such as liver cirrhosis and hepatocellular carcinoma represent a major concern in global health. Treatment options for chronic hepatitis C are limited and no vaccine against HCV infection is available. Vaccine development is hampered by several obstacles. High viral variability and escape from host immune responses render antigen selection a major challenge. Antigen selection requires thorough studies to identify conserved T cell and neutralization epitopes and to decipher neutralization mechanisms, aiming to discover the optimal viral target for immune responses counteracting HCV escape strategies. At the same time it is important to develop antigen presentation systems that are efficient in patients with impaired antiviral immune responses, as often observed during chronic hepatitis C. While most vaccine development programs are based on improving HCV cellular immunity, it is essential to associate, in a same vaccine formulation, immunogens able to induce broad spectrums neutralizing and cellular responses. Owing to recent progresses in the field, here we propose a project aiming to overcome the current limitations in vaccine development by addressing the improvement of B cell responses targeting HCV infection. This will be achieved by a detailed investigation of: 1) mechanisms of antibody-mediated neutralization and escape, 2) impact of lipoproteins associating with the viral particle during assembly/release and counteracting neutralization and 3) cell entry steps that can potentially be targeted by antibodies, including those that are not induced naturally. Thus, through the combined expertise of the team in molecular virology, immunology, clinical hepatology and vectorology, we aim to rationalize the development of B cell immunogens and neutralizing antibodies for novel antiviral immunopreventive strategies targeting HCV infection.
Max ERC Funding
2 447 357 €
Duration
Start date: 2009-04-01, End date: 2014-12-31
Project acronym HEPCIR
Project Cell circuits as targets and biomarkers for liver disease and cancer prevention
Researcher (PI) Thomas Baumert
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2014-ADG
Summary Chronic liver diseases such as liver cirrhosis and hepatocellular carcinoma (HCC) are major challenges for global health. HCC is the second leading and fastest rising cause of cancer death worldwide. The limited availability of therapeutic options reflects our poor understanding of the molecular and clinical mechanisms involved in progression of liver disease. Chronic hepatitis C virus (HCV) infection is a main risk factor for HCC. Although HCC may be avoided by addressing the underlying cause in early stage disease, strategies to prevent HCC in patients with established cirrhosis and advanced fibrosis, in which the risk of HCC persists despite treatment of the underlying cause are lacking. Indeed, even HCV cure does not eliminate the risk of HCC development when advanced fibrosis is already present. Since fibrosis/cirrhosis-driven carcinogenesis is the mechanism of HCC development common to all major etiologies, we propose to use HCV-induced liver disease as a model to decipher the pan-etiology sequence of molecular events underlying disease progression and HCC. Our own data provide solid evidence that HCV infection alters pathways implicated in liver disease progression, including cirrhosis deterioration, HCC development, and overall and liver-specific death. Thus, the molecular investigation of these pathways will identify key cell circuits for the understanding of the pathogenesis of liver disease and HCC in general, and as broadly applicable pan-etiology diagnostic and therapeutic targets. Using a novel patient-derived cell culture model system for liver disease biology combined with advanced functional genomics, novel animal models and clinical investigation, we aim to uncover the cell circuits that are of clinical relevance for liver disease progression and cancer. By providing novel targets and biomarkers for liver disease and HCC prevention, this proposal will have a marked impact on the management and prognosis of patients with liver disease and HCC.
Summary
Chronic liver diseases such as liver cirrhosis and hepatocellular carcinoma (HCC) are major challenges for global health. HCC is the second leading and fastest rising cause of cancer death worldwide. The limited availability of therapeutic options reflects our poor understanding of the molecular and clinical mechanisms involved in progression of liver disease. Chronic hepatitis C virus (HCV) infection is a main risk factor for HCC. Although HCC may be avoided by addressing the underlying cause in early stage disease, strategies to prevent HCC in patients with established cirrhosis and advanced fibrosis, in which the risk of HCC persists despite treatment of the underlying cause are lacking. Indeed, even HCV cure does not eliminate the risk of HCC development when advanced fibrosis is already present. Since fibrosis/cirrhosis-driven carcinogenesis is the mechanism of HCC development common to all major etiologies, we propose to use HCV-induced liver disease as a model to decipher the pan-etiology sequence of molecular events underlying disease progression and HCC. Our own data provide solid evidence that HCV infection alters pathways implicated in liver disease progression, including cirrhosis deterioration, HCC development, and overall and liver-specific death. Thus, the molecular investigation of these pathways will identify key cell circuits for the understanding of the pathogenesis of liver disease and HCC in general, and as broadly applicable pan-etiology diagnostic and therapeutic targets. Using a novel patient-derived cell culture model system for liver disease biology combined with advanced functional genomics, novel animal models and clinical investigation, we aim to uncover the cell circuits that are of clinical relevance for liver disease progression and cancer. By providing novel targets and biomarkers for liver disease and HCC prevention, this proposal will have a marked impact on the management and prognosis of patients with liver disease and HCC.
Max ERC Funding
2 305 000 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym HOMEOSIGN
Project HOMEOSIGN : Homeoprotein signaling during development and in the adult
Researcher (PI) Alain, Louis Prochiantz
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS5, ERC-2013-ADG
Summary "Homeoprotein (HP) signaling designates the ability of HP transcription factors to transfer between cells, thus acting as signaling molecules. We will investigate several properties of this novel signaling pathway in the CNS and how it can shed light on brain development and physiology, also neurological and psychiatric pathologies. The studies will concentrate on 4 HPs (Engrailed1/2, Otx2 and Pax6) illustrative of the HP family and of high physiological interest. Mouse lines will be developed in which the extracellular expression of single chain antibodies against these HPs can be induced at specific places and times, allowing us to follow the consequences of neutralizing extracellular HPs while preserving their cell autonomous functions. With this tool (and others) we shall study if (and how) HP transfer regulates boundary position in the neuroepithelium and direct cell/axon migration and guidance. In postnatal development, we will dissect how Otx2 regulates the opening, closure and reopening of critical periods in the cortex and controls the behavior of neuronal networks throughout life. Also in the adult we will follow how HPs regulate the physiology and survival of distinct neuronal groups, including midbrain dopaminergic neurons. We shall analyze the physiological interactions between HPs and classical signaling molecules and identify HP targets, in particular those regulating the cell energy metabolism, and explore their ability to modulate the epigenetic state of the chromatin. Using Otx2 and Engrailed as baits, we shall identify the complex sugars with which they interact at the cell surface to get an entry into the ""sugar code"" that confers specificity to HP capture by distinct cell populations. This project should identify new HP functions and highlight them as part of a major mode of signal transduction in the developing and mature CNS. It is expected that this will modify the way we look at many developmental, evolutionary and physiological mechanisms."
Summary
"Homeoprotein (HP) signaling designates the ability of HP transcription factors to transfer between cells, thus acting as signaling molecules. We will investigate several properties of this novel signaling pathway in the CNS and how it can shed light on brain development and physiology, also neurological and psychiatric pathologies. The studies will concentrate on 4 HPs (Engrailed1/2, Otx2 and Pax6) illustrative of the HP family and of high physiological interest. Mouse lines will be developed in which the extracellular expression of single chain antibodies against these HPs can be induced at specific places and times, allowing us to follow the consequences of neutralizing extracellular HPs while preserving their cell autonomous functions. With this tool (and others) we shall study if (and how) HP transfer regulates boundary position in the neuroepithelium and direct cell/axon migration and guidance. In postnatal development, we will dissect how Otx2 regulates the opening, closure and reopening of critical periods in the cortex and controls the behavior of neuronal networks throughout life. Also in the adult we will follow how HPs regulate the physiology and survival of distinct neuronal groups, including midbrain dopaminergic neurons. We shall analyze the physiological interactions between HPs and classical signaling molecules and identify HP targets, in particular those regulating the cell energy metabolism, and explore their ability to modulate the epigenetic state of the chromatin. Using Otx2 and Engrailed as baits, we shall identify the complex sugars with which they interact at the cell surface to get an entry into the ""sugar code"" that confers specificity to HP capture by distinct cell populations. This project should identify new HP functions and highlight them as part of a major mode of signal transduction in the developing and mature CNS. It is expected that this will modify the way we look at many developmental, evolutionary and physiological mechanisms."
Max ERC Funding
2 499 995 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym Homo.symbiosus
Project Assessing, preserving and restoring man-microbes symbiosis
Researcher (PI) JOEL DORE
Host Institution (HI) INSTITUT NATIONAL DE LA RECHERCHE AGRONOMIQUE
Call Details Advanced Grant (AdG), LS7, ERC-2017-ADG
Summary The microbiomics revolution has favoured the recognition of the gut as a true organ and the importance of man-microbes symbiosis in health and disease. Derived from a long co-evolution the latter has been challenged by numerous environmental triggers, modern lifestyles, changes in birth modalities, nutritional transition and therapeutic attitudes. A large fraction of the human population has tentatively entered a man-microbes dysbiotic state characterized by altered interactions between microbiome and host features with auto-aggravating crosstalk signals. The result is increased incidence of incurable immune-mediated diseases of modern societies that affect a third the human population on earth today and for which current therapeutics only address symptoms alleviation, rather than considering man as a holobiont.
In this context and its resulting threat for the human species, I will carry out a project geared to open a new era of individualized preventive care and novel gut ecology-based therapeutic approaches. The project will assemble insights and contributions from theoretical to experimental ecology, quantitative and functional microbiomics, preclinical work, cohort studies and clinical trials, so as to:
• Validate the concept of critical transition and alternative stable state as it applies to a shift from man-microbes symbiosis to disease-prone man-microbes dysbiosis
• Assess the potential of diet alone to promote such a shift
• Model the symbiosis-to-dysbiosis transitions and derive predictors of tipping points
• Propose counter-measures that may allow to break vicious circles and restore a balanced, health-prone, man-microbes symbiosis by concomitantly acting upon microbiome and host features
• Validate strategies to reinforce ecological robustness and restore man-microbes symbiosis
Based on a paradigm shift, the proposed work will set the grounds for future personalized preventive nutrition and clinical management considering man as a true holobiont.
Summary
The microbiomics revolution has favoured the recognition of the gut as a true organ and the importance of man-microbes symbiosis in health and disease. Derived from a long co-evolution the latter has been challenged by numerous environmental triggers, modern lifestyles, changes in birth modalities, nutritional transition and therapeutic attitudes. A large fraction of the human population has tentatively entered a man-microbes dysbiotic state characterized by altered interactions between microbiome and host features with auto-aggravating crosstalk signals. The result is increased incidence of incurable immune-mediated diseases of modern societies that affect a third the human population on earth today and for which current therapeutics only address symptoms alleviation, rather than considering man as a holobiont.
In this context and its resulting threat for the human species, I will carry out a project geared to open a new era of individualized preventive care and novel gut ecology-based therapeutic approaches. The project will assemble insights and contributions from theoretical to experimental ecology, quantitative and functional microbiomics, preclinical work, cohort studies and clinical trials, so as to:
• Validate the concept of critical transition and alternative stable state as it applies to a shift from man-microbes symbiosis to disease-prone man-microbes dysbiosis
• Assess the potential of diet alone to promote such a shift
• Model the symbiosis-to-dysbiosis transitions and derive predictors of tipping points
• Propose counter-measures that may allow to break vicious circles and restore a balanced, health-prone, man-microbes symbiosis by concomitantly acting upon microbiome and host features
• Validate strategies to reinforce ecological robustness and restore man-microbes symbiosis
Based on a paradigm shift, the proposed work will set the grounds for future personalized preventive nutrition and clinical management considering man as a true holobiont.
Max ERC Funding
2 491 014 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym HOPE
Project Host Protective Engineering of Cancer Immunity by Targeting the Intracellular Immune Checkpoint NR2F6
Researcher (PI) Gottfried BAIER
Host Institution (HI) MEDIZINISCHE UNIVERSITAT INNSBRUCK
Call Details Advanced Grant (AdG), LS7, ERC-2017-ADG
Summary "Because of its biological complexity, cancer is still poorly understood. Chronic inflammation has been shown, both experimentally and epidemiologically, to be a predisposition to, and also an inseparable aspect of clinically prevalent cancer entities. Therefore, a detailed understanding of both tumour and immune cell functions in cancer progression is a prerequisite for more successful therapeutic startegies. My team was the first to reveal the lymphocyte-intrinsic PKC/NR2F6 axis as an essential signalling node at the crossroads between inflammation and cancer. It is the mission of this project to identify molecular signatures that influence the risk of developing tumours employing established research tools and state-of-the-art genetic, biochemical, proteomic and transcriptomic as well as large scale CRISPR/Cas9 perturbation screening-based functional genomic technologies. Defining this as yet poorly elucidated effector pathway with its profoundly relevant role would enable development of preventive and immune-therapeutic strategies against NSCLC lung cancer and potentially also against other entities. Our three-pronged approach to achieve this goal is to: (i) delineate biological and clinical properties of the immunological PKC/NR2F6 network, (ii) validate NR2F6 as an immune-oncology combination target needed to overcome limitations to ""first generation anti-PD-1 checkpoint inhibitors"" rendering T cells capable of rejecting tumours and their metastases at distal organs and (iii) exploit human combinatorial T cell therapy concepts for prevention of immune-related adverse events as well as of tumour recurrence by reducing opportunities for the tumour to develop resistance in the clinic. Insight into the functions of NR2F6 pathway and involved mechanisms is a prerequisite for understanding how the microenvironment at the tumour site either supports tumour growth and spread or prevents tumour initiation and progression, the latter by host-protective cancer immunity."
Summary
"Because of its biological complexity, cancer is still poorly understood. Chronic inflammation has been shown, both experimentally and epidemiologically, to be a predisposition to, and also an inseparable aspect of clinically prevalent cancer entities. Therefore, a detailed understanding of both tumour and immune cell functions in cancer progression is a prerequisite for more successful therapeutic startegies. My team was the first to reveal the lymphocyte-intrinsic PKC/NR2F6 axis as an essential signalling node at the crossroads between inflammation and cancer. It is the mission of this project to identify molecular signatures that influence the risk of developing tumours employing established research tools and state-of-the-art genetic, biochemical, proteomic and transcriptomic as well as large scale CRISPR/Cas9 perturbation screening-based functional genomic technologies. Defining this as yet poorly elucidated effector pathway with its profoundly relevant role would enable development of preventive and immune-therapeutic strategies against NSCLC lung cancer and potentially also against other entities. Our three-pronged approach to achieve this goal is to: (i) delineate biological and clinical properties of the immunological PKC/NR2F6 network, (ii) validate NR2F6 as an immune-oncology combination target needed to overcome limitations to ""first generation anti-PD-1 checkpoint inhibitors"" rendering T cells capable of rejecting tumours and their metastases at distal organs and (iii) exploit human combinatorial T cell therapy concepts for prevention of immune-related adverse events as well as of tumour recurrence by reducing opportunities for the tumour to develop resistance in the clinic. Insight into the functions of NR2F6 pathway and involved mechanisms is a prerequisite for understanding how the microenvironment at the tumour site either supports tumour growth and spread or prevents tumour initiation and progression, the latter by host-protective cancer immunity."
Max ERC Funding
2 484 325 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym HYGIENE
Project THE HYGIENE HYPOTHESIS : REVISITING THE CONCEPT BY INTEGRATING EPIDEMIOLOGY AND MECHANISTIC STUDIES
Researcher (PI) Jean-François Bach
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary The hygiene hypothesis postulating the paradoxical protective role of infections on immune-mediated
diseases including atopy (i.e. atopic dermatitis, rhinitis, asthma) and more recently autoimmune diseases has
been the matter of extensive investigation. The aim of the present project is to validate this hypothesis
integrating epidemiological and experimental studies.
We will review in a meta-analysis published studies of direct and indirect markers of infections and atopic
diseases, and conduct a case-control study to analyse the association between infections and atopy using
atopic dermatitis as a prototypic model. This epidemiological study will assess the occurrence of different
infections and other risk factors related to the incidence of atopic dermatitis in children under age five in
Italy (300 cases and 600 controls).
This epidemiological study will be supported by experimental approaches addressing mechanistic questions
raised by the hygiene hypothesis. Experimental models will include induction of acute and chronic
bronchoconstriction/asthma, atopic dermatitis. In addition the project will aim at devising new mouse models
of atopy. The nature of infections providing protection against allergic diseases will be investigated to
characterise the difference at the molecular level between protective and non protective pathogens or their
derivatives. The underlying immune mechanisms notably homeostasis imbalance, antigenic competition,
stimulation of regulatory immune cells and Toll-like receptor involvement will be analysed. In addition to
developing integrated in vivo models, including the use of transgenic animals, efforts will be focussed on the
study of available synthetic pathogen-derived compounds showing a protective activity to better approach
their cellular and molecular mode of action. These studies may pave the way to novel and safe therapies that
could advantageously substitute for the “protective” immune stimulation induced by major infections.
Summary
The hygiene hypothesis postulating the paradoxical protective role of infections on immune-mediated
diseases including atopy (i.e. atopic dermatitis, rhinitis, asthma) and more recently autoimmune diseases has
been the matter of extensive investigation. The aim of the present project is to validate this hypothesis
integrating epidemiological and experimental studies.
We will review in a meta-analysis published studies of direct and indirect markers of infections and atopic
diseases, and conduct a case-control study to analyse the association between infections and atopy using
atopic dermatitis as a prototypic model. This epidemiological study will assess the occurrence of different
infections and other risk factors related to the incidence of atopic dermatitis in children under age five in
Italy (300 cases and 600 controls).
This epidemiological study will be supported by experimental approaches addressing mechanistic questions
raised by the hygiene hypothesis. Experimental models will include induction of acute and chronic
bronchoconstriction/asthma, atopic dermatitis. In addition the project will aim at devising new mouse models
of atopy. The nature of infections providing protection against allergic diseases will be investigated to
characterise the difference at the molecular level between protective and non protective pathogens or their
derivatives. The underlying immune mechanisms notably homeostasis imbalance, antigenic competition,
stimulation of regulatory immune cells and Toll-like receptor involvement will be analysed. In addition to
developing integrated in vivo models, including the use of transgenic animals, efforts will be focussed on the
study of available synthetic pathogen-derived compounds showing a protective activity to better approach
their cellular and molecular mode of action. These studies may pave the way to novel and safe therapies that
could advantageously substitute for the “protective” immune stimulation induced by major infections.
Max ERC Funding
2 099 999 €
Duration
Start date: 2010-08-01, End date: 2015-07-31