Project acronym 3D-REPAIR
Project Spatial organization of DNA repair within the nucleus
Researcher (PI) Evanthia Soutoglou
Host Institution (HI) CENTRE EUROPEEN DE RECHERCHE EN BIOLOGIE ET MEDECINE
Call Details Consolidator Grant (CoG), LS2, ERC-2015-CoG
Summary Faithful repair of double stranded DNA breaks (DSBs) is essential, as they are at the origin of genome instability, chromosomal translocations and cancer. Cells repair DSBs through different pathways, which can be faithful or mutagenic, and the balance between them at a given locus must be tightly regulated to preserve genome integrity. Although, much is known about DSB repair factors, how the choice between pathways is controlled within the nuclear environment is not understood. We have shown that nuclear architecture and non-random genome organization determine the frequency of chromosomal translocations and that pathway choice is dictated by the spatial organization of DNA in the nucleus. Nevertheless, what determines which pathway is activated in response to DSBs at specific genomic locations is not understood. Furthermore, the impact of 3D-genome folding on the kinetics and efficiency of DSB repair is completely unknown.
Here we aim to understand how nuclear compartmentalization, chromatin structure and genome organization impact on the efficiency of detection, signaling and repair of DSBs. We will unravel what determines the DNA repair specificity within distinct nuclear compartments using protein tethering, promiscuous biotinylation and quantitative proteomics. We will determine how DNA repair is orchestrated at different heterochromatin structures using a CRISPR/Cas9-based system that allows, for the first time robust induction of DSBs at specific heterochromatin compartments. Finally, we will investigate the role of 3D-genome folding in the kinetics of DNA repair and pathway choice using single nucleotide resolution DSB-mapping coupled to 3D-topological maps.
This proposal has significant implications for understanding the mechanisms controlling DNA repair within the nuclear environment and will reveal the regions of the genome that are susceptible to genomic instability and help us understand why certain mutations and translocations are recurrent in cancer
Summary
Faithful repair of double stranded DNA breaks (DSBs) is essential, as they are at the origin of genome instability, chromosomal translocations and cancer. Cells repair DSBs through different pathways, which can be faithful or mutagenic, and the balance between them at a given locus must be tightly regulated to preserve genome integrity. Although, much is known about DSB repair factors, how the choice between pathways is controlled within the nuclear environment is not understood. We have shown that nuclear architecture and non-random genome organization determine the frequency of chromosomal translocations and that pathway choice is dictated by the spatial organization of DNA in the nucleus. Nevertheless, what determines which pathway is activated in response to DSBs at specific genomic locations is not understood. Furthermore, the impact of 3D-genome folding on the kinetics and efficiency of DSB repair is completely unknown.
Here we aim to understand how nuclear compartmentalization, chromatin structure and genome organization impact on the efficiency of detection, signaling and repair of DSBs. We will unravel what determines the DNA repair specificity within distinct nuclear compartments using protein tethering, promiscuous biotinylation and quantitative proteomics. We will determine how DNA repair is orchestrated at different heterochromatin structures using a CRISPR/Cas9-based system that allows, for the first time robust induction of DSBs at specific heterochromatin compartments. Finally, we will investigate the role of 3D-genome folding in the kinetics of DNA repair and pathway choice using single nucleotide resolution DSB-mapping coupled to 3D-topological maps.
This proposal has significant implications for understanding the mechanisms controlling DNA repair within the nuclear environment and will reveal the regions of the genome that are susceptible to genomic instability and help us understand why certain mutations and translocations are recurrent in cancer
Max ERC Funding
1 999 750 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym 3DEpi
Project Transgenerational epigenetic inheritance of chromatin states : the role of Polycomb and 3D chromosome architecture
Researcher (PI) Giacomo CAVALLI
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS2, ERC-2017-ADG
Summary Epigenetic inheritance entails transmission of phenotypic traits not encoded in the DNA sequence and, in the most extreme case, Transgenerational Epigenetic Inheritance (TEI) involves transmission of memory through multiple generations. Very little is known on the mechanisms governing TEI and this is the subject of the present proposal. By transiently enhancing long-range chromatin interactions, we recently established isogenic Drosophila epilines that carry stable alternative epialleles, defined by differential levels of the Polycomb-dependent H3K27me3 mark. Furthermore, we extended our paradigm to natural phenotypes. These are ideal systems to study the role of Polycomb group (PcG) proteins and other components in regulating nuclear organization and epigenetic inheritance of chromatin states. The present project conjugates genetics, epigenomics, imaging and molecular biology to reach three critical aims.
Aim 1: Analysis of the molecular mechanisms regulating Polycomb-mediated TEI. We will identify the DNA, protein and RNA components that trigger and maintain transgenerational chromatin inheritance as well as their mechanisms of action.
Aim 2: Role of 3D genome organization in the regulation of TEI. We will analyze the developmental dynamics of TEI-inducing long-range chromatin interactions, identify chromatin components mediating 3D chromatin contacts and characterize their function in the TEI process.
Aim 3: Identification of a broader role of TEI during development. TEI might reflect a normal role of PcG components in the transmission of parental chromatin onto the next embryonic generation. We will explore this possibility by establishing other TEI paradigms and by relating TEI to the normal PcG function in these systems and in normal development.
This research program will unravel the biological significance and the molecular underpinnings of TEI and lead the way towards establishing this area of research into a consolidated scientific discipline.
Summary
Epigenetic inheritance entails transmission of phenotypic traits not encoded in the DNA sequence and, in the most extreme case, Transgenerational Epigenetic Inheritance (TEI) involves transmission of memory through multiple generations. Very little is known on the mechanisms governing TEI and this is the subject of the present proposal. By transiently enhancing long-range chromatin interactions, we recently established isogenic Drosophila epilines that carry stable alternative epialleles, defined by differential levels of the Polycomb-dependent H3K27me3 mark. Furthermore, we extended our paradigm to natural phenotypes. These are ideal systems to study the role of Polycomb group (PcG) proteins and other components in regulating nuclear organization and epigenetic inheritance of chromatin states. The present project conjugates genetics, epigenomics, imaging and molecular biology to reach three critical aims.
Aim 1: Analysis of the molecular mechanisms regulating Polycomb-mediated TEI. We will identify the DNA, protein and RNA components that trigger and maintain transgenerational chromatin inheritance as well as their mechanisms of action.
Aim 2: Role of 3D genome organization in the regulation of TEI. We will analyze the developmental dynamics of TEI-inducing long-range chromatin interactions, identify chromatin components mediating 3D chromatin contacts and characterize their function in the TEI process.
Aim 3: Identification of a broader role of TEI during development. TEI might reflect a normal role of PcG components in the transmission of parental chromatin onto the next embryonic generation. We will explore this possibility by establishing other TEI paradigms and by relating TEI to the normal PcG function in these systems and in normal development.
This research program will unravel the biological significance and the molecular underpinnings of TEI and lead the way towards establishing this area of research into a consolidated scientific discipline.
Max ERC Funding
2 500 000 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym 4D-GenEx
Project Spatio-temporal Organization and Expression of the Genome
Researcher (PI) Antoine COULON
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), LS2, ERC-2017-STG
Summary This project investigates the two-way relationship between spatio-temporal genome organization and coordinated gene regulation, through an approach at the interface between physics, computer science and biology.
In the nucleus, preferred positions are observed from chromosomes to single genes, in relation to normal and pathological cellular states. Evidence indicates a complex spatio-temporal coupling between co-regulated genes: e.g. certain genes cluster spatially when responding to similar factors and transcriptional noise patterns suggest domain-wide mechanisms. Yet, no individual experiment allows probing transcriptional coordination in 4 dimensions (FISH, live locus tracking, Hi-C...). Interpreting such data also critically requires theory (stochastic processes, statistical physics…). A lack of appropriate experimental/analytical approaches is impairing our understanding of the 4D genome.
Our proposal combines cutting-edge single-molecule imaging, signal-theory data analysis and physical modeling to study how genes coordinate in space and time in a single nucleus. Our objectives are to understand (a) competition/recycling of shared resources between genes within subnuclear compartments, (b) how enhancers communicate with genes domain-wide, and (c) the role of local conformational dynamics and supercoiling in gene co-regulation. Our organizing hypothesis is that, by acting on their microenvironment, genes shape their co-expression with other genes.
Building upon my expertise, we will use dual-color MS2/PP7 RNA labeling to visualize for the first time transcription and motion of pairs of hormone-responsive genes in real time. With our innovative signal analysis tools, we will extract spatio-temporal signatures of underlying processes, which we will investigate with stochastic modeling and validate through experimental perturbations. We expect to uncover how the functional organization of the linear genome relates to its physical properties and dynamics in 4D.
Summary
This project investigates the two-way relationship between spatio-temporal genome organization and coordinated gene regulation, through an approach at the interface between physics, computer science and biology.
In the nucleus, preferred positions are observed from chromosomes to single genes, in relation to normal and pathological cellular states. Evidence indicates a complex spatio-temporal coupling between co-regulated genes: e.g. certain genes cluster spatially when responding to similar factors and transcriptional noise patterns suggest domain-wide mechanisms. Yet, no individual experiment allows probing transcriptional coordination in 4 dimensions (FISH, live locus tracking, Hi-C...). Interpreting such data also critically requires theory (stochastic processes, statistical physics…). A lack of appropriate experimental/analytical approaches is impairing our understanding of the 4D genome.
Our proposal combines cutting-edge single-molecule imaging, signal-theory data analysis and physical modeling to study how genes coordinate in space and time in a single nucleus. Our objectives are to understand (a) competition/recycling of shared resources between genes within subnuclear compartments, (b) how enhancers communicate with genes domain-wide, and (c) the role of local conformational dynamics and supercoiling in gene co-regulation. Our organizing hypothesis is that, by acting on their microenvironment, genes shape their co-expression with other genes.
Building upon my expertise, we will use dual-color MS2/PP7 RNA labeling to visualize for the first time transcription and motion of pairs of hormone-responsive genes in real time. With our innovative signal analysis tools, we will extract spatio-temporal signatures of underlying processes, which we will investigate with stochastic modeling and validate through experimental perturbations. We expect to uncover how the functional organization of the linear genome relates to its physical properties and dynamics in 4D.
Max ERC Funding
1 499 750 €
Duration
Start date: 2018-04-01, End date: 2023-03-31
Project acronym AAA
Project Adaptive Actin Architectures
Researcher (PI) Laurent Blanchoin
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS3, ERC-2016-ADG
Summary Although we have extensive knowledge of many important processes in cell biology, including information on many of the molecules involved and the physical interactions among them, we still do not understand most of the dynamical features that are the essence of living systems. This is particularly true for the actin cytoskeleton, a major component of the internal architecture of eukaryotic cells. In living cells, actin networks constantly assemble and disassemble filaments while maintaining an apparent stable structure, suggesting a perfect balance between the two processes. Such behaviors are called “dynamic steady states”. They confer upon actin networks a high degree of plasticity allowing them to adapt in response to external changes and enable cells to adjust to their environments. Despite their fundamental importance in the regulation of cell physiology, the basic mechanisms that control the coordinated dynamics of co-existing actin networks are poorly understood. In the AAA project, first, we will characterize the parameters that allow the coupling among co-existing actin networks at steady state. In vitro reconstituted systems will be used to control the actin nucleation patterns, the closed volume of the reaction chamber and the physical interaction of the networks. We hope to unravel the mechanism allowing the global coherence of a dynamic actin cytoskeleton. Second, we will use our unique capacity to perform dynamic micropatterning, to add or remove actin nucleation sites in real time, in order to investigate the ability of dynamic networks to adapt to changes and the role of coupled network dynamics in this emergent property. In this part, in vitro experiments will be complemented by the analysis of actin network remodeling in living cells. In the end, our project will provide a comprehensive understanding of how the adaptive response of the cytoskeleton derives from the complex interplay between its biochemical, structural and mechanical properties.
Summary
Although we have extensive knowledge of many important processes in cell biology, including information on many of the molecules involved and the physical interactions among them, we still do not understand most of the dynamical features that are the essence of living systems. This is particularly true for the actin cytoskeleton, a major component of the internal architecture of eukaryotic cells. In living cells, actin networks constantly assemble and disassemble filaments while maintaining an apparent stable structure, suggesting a perfect balance between the two processes. Such behaviors are called “dynamic steady states”. They confer upon actin networks a high degree of plasticity allowing them to adapt in response to external changes and enable cells to adjust to their environments. Despite their fundamental importance in the regulation of cell physiology, the basic mechanisms that control the coordinated dynamics of co-existing actin networks are poorly understood. In the AAA project, first, we will characterize the parameters that allow the coupling among co-existing actin networks at steady state. In vitro reconstituted systems will be used to control the actin nucleation patterns, the closed volume of the reaction chamber and the physical interaction of the networks. We hope to unravel the mechanism allowing the global coherence of a dynamic actin cytoskeleton. Second, we will use our unique capacity to perform dynamic micropatterning, to add or remove actin nucleation sites in real time, in order to investigate the ability of dynamic networks to adapt to changes and the role of coupled network dynamics in this emergent property. In this part, in vitro experiments will be complemented by the analysis of actin network remodeling in living cells. In the end, our project will provide a comprehensive understanding of how the adaptive response of the cytoskeleton derives from the complex interplay between its biochemical, structural and mechanical properties.
Max ERC Funding
2 349 898 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym ACAP
Project Acency Costs and Asset Pricing
Researcher (PI) Thomas Mariotti
Host Institution (HI) FONDATION JEAN-JACQUES LAFFONT,TOULOUSE SCIENCES ECONOMIQUES
Call Details Starting Grant (StG), SH1, ERC-2007-StG
Summary The main objective of this research project is to contribute at bridging the gap between the two main branches of financial theory, namely corporate finance and asset pricing. It is motivated by the conviction that these two aspects of financial activity should and can be analyzed within a unified framework. This research will borrow from these two approaches in order to construct theoretical models that allow one to analyze the design and issuance of financial securities, as well as the dynamics of their valuations. Unlike asset pricing, which takes as given the price of the fundamentals, the goal is to derive security price processes from a precise description of firm’s operations and internal frictions. Regarding the latter, and in line with traditional corporate finance theory, the analysis will emphasize the role of agency costs within the firm for the design of its securities. But the analysis will be pushed one step further by studying the impact of these agency costs on key financial variables such as stock and bond prices, leverage, book-to-market ratios, default risk, or the holding of liquidities by firms. One of the contributions of this research project is to show how these variables are interrelated when firms and investors agree upon optimal financial arrangements. The final objective is to derive a rich set of testable asset pricing implications that would eventually be brought to the data.
Summary
The main objective of this research project is to contribute at bridging the gap between the two main branches of financial theory, namely corporate finance and asset pricing. It is motivated by the conviction that these two aspects of financial activity should and can be analyzed within a unified framework. This research will borrow from these two approaches in order to construct theoretical models that allow one to analyze the design and issuance of financial securities, as well as the dynamics of their valuations. Unlike asset pricing, which takes as given the price of the fundamentals, the goal is to derive security price processes from a precise description of firm’s operations and internal frictions. Regarding the latter, and in line with traditional corporate finance theory, the analysis will emphasize the role of agency costs within the firm for the design of its securities. But the analysis will be pushed one step further by studying the impact of these agency costs on key financial variables such as stock and bond prices, leverage, book-to-market ratios, default risk, or the holding of liquidities by firms. One of the contributions of this research project is to show how these variables are interrelated when firms and investors agree upon optimal financial arrangements. The final objective is to derive a rich set of testable asset pricing implications that would eventually be brought to the data.
Max ERC Funding
1 000 000 €
Duration
Start date: 2008-11-01, End date: 2014-10-31
Project acronym ACTINIT
Project Brain-behavior forecasting: The causal determinants of spontaneous self-initiated action in the study of volition and the development of asynchronous brain-computer interfaces.
Researcher (PI) Aaron Schurger
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS5, ERC-2014-STG
Summary "How are actions initiated by the human brain when there is no external sensory cue or other immediate imperative? How do subtle ongoing interactions within the brain and between the brain, body, and sensory context influence the spontaneous initiation of action? How should we approach the problem of trying to identify the neural events that cause spontaneous voluntary action? Much is understood about how the brain decides between competing alternatives, leading to different behavioral responses. But far less is known about how the brain decides "when" to perform an action, or "whether" to perform an action in the first place, especially in a context where there is no sensory cue to act such as during foraging. This project seeks to open a new chapter in the study of spontaneous voluntary action building on a novel hypothesis recently introduced by the applicant (Schurger et al, PNAS 2012) concerning the role of ongoing neural activity in action initiation. We introduce brain-behavior forecasting, the converse of movement-locked averaging, as an approach to identifying the neurodynamic states that commit the motor system to performing an action "now", and will apply it in the context of information foraging. Spontaneous action remains a profound mystery in the brain basis of behavior, in humans and other animals, and is also central to the problem of asynchronous intention-detection in brain-computer interfaces (BCIs). A BCI must not only interpret what the user intends, but also must detect "when" the user intends to act, and not respond otherwise. This remains the biggest challenge in the development of high-performance BCIs, whether invasive or non-invasive. This project will take a systematic and collaborative approach to the study of spontaneous self-initiated action, incorporating computational modeling, neuroimaging, and machine learning techniques towards a deeper understanding of voluntary behavior and the robust asynchronous detection of decisions-to-act."
Summary
"How are actions initiated by the human brain when there is no external sensory cue or other immediate imperative? How do subtle ongoing interactions within the brain and between the brain, body, and sensory context influence the spontaneous initiation of action? How should we approach the problem of trying to identify the neural events that cause spontaneous voluntary action? Much is understood about how the brain decides between competing alternatives, leading to different behavioral responses. But far less is known about how the brain decides "when" to perform an action, or "whether" to perform an action in the first place, especially in a context where there is no sensory cue to act such as during foraging. This project seeks to open a new chapter in the study of spontaneous voluntary action building on a novel hypothesis recently introduced by the applicant (Schurger et al, PNAS 2012) concerning the role of ongoing neural activity in action initiation. We introduce brain-behavior forecasting, the converse of movement-locked averaging, as an approach to identifying the neurodynamic states that commit the motor system to performing an action "now", and will apply it in the context of information foraging. Spontaneous action remains a profound mystery in the brain basis of behavior, in humans and other animals, and is also central to the problem of asynchronous intention-detection in brain-computer interfaces (BCIs). A BCI must not only interpret what the user intends, but also must detect "when" the user intends to act, and not respond otherwise. This remains the biggest challenge in the development of high-performance BCIs, whether invasive or non-invasive. This project will take a systematic and collaborative approach to the study of spontaneous self-initiated action, incorporating computational modeling, neuroimaging, and machine learning techniques towards a deeper understanding of voluntary behavior and the robust asynchronous detection of decisions-to-act."
Max ERC Funding
1 338 130 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym ADIPOR
Project Molecular and structural pharmacology of adiponectin receptor: towards innovative treatments of obesity-related diseases.
Researcher (PI) Sebastien Jean Antoine Granier
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Consolidator Grant (CoG), LS7, ERC-2014-CoG
Summary The human kind is witnessing an escalation of obesity-related health problems such as cardiovascular diseases and type 2 diabetes. A recent groundbreaking study revealed adiponectin receptors (ADIPOR) as key targets for treating such obesity-related diseases. Indeed, the modulation of this integral membrane protein by small molecules agonists ameliorates diabetes and prolongs lifespan of genetically obese rodent model. Despite these exciting results and the importance of ADIPOR in human physiology, there is a complete lack of knowledge of ADIPOR mechanisms of action and pharmacology. This is mainly due to the challenges associated with the characterization of membrane protein structure and function. To fill this gap of knowledge and based on my extensive experience in membrane protein biology, I propose here to characterize the the proximal signaling pathways associated with ADIPOR activation as well as the molecular and structural mechanisms of ADIPOR activation. We will develop an innovative integrated strategy combining state-of-the-art molecular and structural pharmacology approaches including 1) molecular analyses of ADIPOR network of interaction using resonance energy transfer measurement in living cells and a proteomic analysis and 2) structural analyses of ADIPOR and signaling complexes using biophysics and X-ray crystallography. Our data will have a major impact on drug discovery for treating obesity-related diseases as it will enable the application of structure-based drug design and in silico screening for the molecular control of ADIPOR activity. The proposed high-risk endeavor of obtaining structural data on these atypical membrane signaling complexes is a new direction both for my career and for the field of adiponectin biology; the exceptionally high gain from these studies fully justifies the risks; the feasibility of this project is supported by my recent success in membrane protein pharmacology, biochemistry, biophysics and crystallography.
Summary
The human kind is witnessing an escalation of obesity-related health problems such as cardiovascular diseases and type 2 diabetes. A recent groundbreaking study revealed adiponectin receptors (ADIPOR) as key targets for treating such obesity-related diseases. Indeed, the modulation of this integral membrane protein by small molecules agonists ameliorates diabetes and prolongs lifespan of genetically obese rodent model. Despite these exciting results and the importance of ADIPOR in human physiology, there is a complete lack of knowledge of ADIPOR mechanisms of action and pharmacology. This is mainly due to the challenges associated with the characterization of membrane protein structure and function. To fill this gap of knowledge and based on my extensive experience in membrane protein biology, I propose here to characterize the the proximal signaling pathways associated with ADIPOR activation as well as the molecular and structural mechanisms of ADIPOR activation. We will develop an innovative integrated strategy combining state-of-the-art molecular and structural pharmacology approaches including 1) molecular analyses of ADIPOR network of interaction using resonance energy transfer measurement in living cells and a proteomic analysis and 2) structural analyses of ADIPOR and signaling complexes using biophysics and X-ray crystallography. Our data will have a major impact on drug discovery for treating obesity-related diseases as it will enable the application of structure-based drug design and in silico screening for the molecular control of ADIPOR activity. The proposed high-risk endeavor of obtaining structural data on these atypical membrane signaling complexes is a new direction both for my career and for the field of adiponectin biology; the exceptionally high gain from these studies fully justifies the risks; the feasibility of this project is supported by my recent success in membrane protein pharmacology, biochemistry, biophysics and crystallography.
Max ERC Funding
1 989 518 €
Duration
Start date: 2015-07-01, End date: 2020-06-30
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 ALS-Networks
Project Defining functional networks of genetic causes for ALS and related neurodegenerative disorders
Researcher (PI) Edor Kabashi
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Consolidator Grant (CoG), LS5, ERC-2015-CoG
Summary Brain and spinal cord diseases affect 38% of the European population and cost over 800 billion € annually; representing by far the largest health challenge. ALS is a prevalent neurological disease caused by motor neuron death with an invariably fatal outcome. I contributed to ALS research with the groundbreaking discovery of TDP-43 mutations, functionally characterized these mutations in the first vertebrate model and demonstrated a genetic interaction with another major ALS gene FUS. Emerging evidence indicates that four major causative factors in ALS, C9orf72, TDP-43, FUS & SQSTM1, genetically interact and could function in common cellular mechanisms. Here, I will develop zebrafish transgenic lines for all four genes, using state of the art genomic editing tools to combine simultaneous gene knockout and expression of the mutant alleles. Using these innovative disease models I will study the functional interactions amongst these four genes and their converging effect on key ALS pathogenic mechanisms: autophagy degradation, stress granule formation and RNA regulation. These studies will permit to pinpoint the molecular cascades that underlie ALS-related neurodegeneration. We will further expand the current ALS network by proposing and validating novel genetic interactors, which will be further screened for disease-causing variants and as pathological markers in patient samples. The power of zebrafish as a vertebrate model amenable to high-content phenotype-based screens will enable discovery of bioactive compounds that are neuroprotective in multiple animal models of disease. This project will increase the fundamental understanding of the relevance of C9orf72, TDP-43, FUS and SQSTM1 by developing animal models to characterize common pathophysiological mechanisms. Furthermore, I will uncover novel genetic, disease-related and pharmacological modifiers to extend the ALS network that will facilitate development of therapeutic strategies for neurodegenerative disorders
Summary
Brain and spinal cord diseases affect 38% of the European population and cost over 800 billion € annually; representing by far the largest health challenge. ALS is a prevalent neurological disease caused by motor neuron death with an invariably fatal outcome. I contributed to ALS research with the groundbreaking discovery of TDP-43 mutations, functionally characterized these mutations in the first vertebrate model and demonstrated a genetic interaction with another major ALS gene FUS. Emerging evidence indicates that four major causative factors in ALS, C9orf72, TDP-43, FUS & SQSTM1, genetically interact and could function in common cellular mechanisms. Here, I will develop zebrafish transgenic lines for all four genes, using state of the art genomic editing tools to combine simultaneous gene knockout and expression of the mutant alleles. Using these innovative disease models I will study the functional interactions amongst these four genes and their converging effect on key ALS pathogenic mechanisms: autophagy degradation, stress granule formation and RNA regulation. These studies will permit to pinpoint the molecular cascades that underlie ALS-related neurodegeneration. We will further expand the current ALS network by proposing and validating novel genetic interactors, which will be further screened for disease-causing variants and as pathological markers in patient samples. The power of zebrafish as a vertebrate model amenable to high-content phenotype-based screens will enable discovery of bioactive compounds that are neuroprotective in multiple animal models of disease. This project will increase the fundamental understanding of the relevance of C9orf72, TDP-43, FUS and SQSTM1 by developing animal models to characterize common pathophysiological mechanisms. Furthermore, I will uncover novel genetic, disease-related and pharmacological modifiers to extend the ALS network that will facilitate development of therapeutic strategies for neurodegenerative disorders
Max ERC Funding
2 000 000 €
Duration
Start date: 2017-04-01, End date: 2022-03-31
Project acronym AltCheM
Project In vivo functional screens to decipher mechanisms of stochastically- and mutationally-induced chemoresistance in Acute Myeloid Leukemia
Researcher (PI) Alexandre PUISSANT
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS4, ERC-2017-STG
Summary Acute Myeloid Leukemia (AML), the most common leukemia diagnosed in adults, represents the paradigm of resistance to front-line therapies in hematology. Indeed, AML is so genetically complex that only few targeted therapies are currently tested in this disease and chemotherapy remains the only standard treatment for AML since the past four decades. Despite an initial sustained remission achieved by chemotherapeutic agents, almost all patients relapse with a chemoresistant minimal residual disease (MRD). The goal of my proposal is to characterize the still poorly understood biological mechanisms underlying persistence and emergence of MRD.
MRD is the consequence of the re-expansion of leukemia-initiating cells that are intrinsically more resistant to chemotherapy. This cell fraction may be stochastically more prone to survive front-line therapy regardless of their mutational status (the stochastic model), or genetically predetermined to resist by virtue of a collection of chemoprotective mutations (the mutational model).
I have already generated in mice, by consecutive rounds of chemotherapy, a stochastic MLL-AF9-driven chemoresistance model that I examined by RNA-sequencing. I will pursue the comprehensive cell autonomous and cell non-autonomous characterization of this chemoresistant AML disease using whole-exome and ChIP-sequencing.
To establish a mutationally-induced chemoresistant mouse model, I will conduct an innovative in vivo screen using pooled mutant open reading frame and shRNA libraries in order to predict which combinations of mutations, among those already known in AML, actively promote chemoresistance.
Finally, by combining genomic profiling and in vivo shRNA screening experiments, I will decipher the molecular mechanisms and identify the functional effectors of these two modes of resistance. Ultimately, I will then be able to firmly establish the fundamental relevance of the stochastic and/or the mutational model of chemoresistance for MRD genesis.
Summary
Acute Myeloid Leukemia (AML), the most common leukemia diagnosed in adults, represents the paradigm of resistance to front-line therapies in hematology. Indeed, AML is so genetically complex that only few targeted therapies are currently tested in this disease and chemotherapy remains the only standard treatment for AML since the past four decades. Despite an initial sustained remission achieved by chemotherapeutic agents, almost all patients relapse with a chemoresistant minimal residual disease (MRD). The goal of my proposal is to characterize the still poorly understood biological mechanisms underlying persistence and emergence of MRD.
MRD is the consequence of the re-expansion of leukemia-initiating cells that are intrinsically more resistant to chemotherapy. This cell fraction may be stochastically more prone to survive front-line therapy regardless of their mutational status (the stochastic model), or genetically predetermined to resist by virtue of a collection of chemoprotective mutations (the mutational model).
I have already generated in mice, by consecutive rounds of chemotherapy, a stochastic MLL-AF9-driven chemoresistance model that I examined by RNA-sequencing. I will pursue the comprehensive cell autonomous and cell non-autonomous characterization of this chemoresistant AML disease using whole-exome and ChIP-sequencing.
To establish a mutationally-induced chemoresistant mouse model, I will conduct an innovative in vivo screen using pooled mutant open reading frame and shRNA libraries in order to predict which combinations of mutations, among those already known in AML, actively promote chemoresistance.
Finally, by combining genomic profiling and in vivo shRNA screening experiments, I will decipher the molecular mechanisms and identify the functional effectors of these two modes of resistance. Ultimately, I will then be able to firmly establish the fundamental relevance of the stochastic and/or the mutational model of chemoresistance for MRD genesis.
Max ERC Funding
1 500 000 €
Duration
Start date: 2018-03-01, End date: 2023-02-28
Project acronym altEJrepair
Project Characterisation of DNA Double-Strand Break Repair by Alternative End-Joining: Potential Targets for Cancer Therapy
Researcher (PI) Raphael CECCALDI
Host Institution (HI) INSTITUT CURIE
Call Details Starting Grant (StG), LS1, ERC-2016-STG
Summary DNA repair pathways evolved as an intricate network that senses DNA damage and resolves it in order to minimise genetic lesions and thus preventing tumour formation. Gaining in recognition the last few years, the alternative end-joining (alt-EJ) DNA repair pathway was recently shown to be up-regulated and required for cancer cell viability in the absence of homologous recombination-mediated repair (HR). Despite this integral role, the alt-EJ repair pathway remains poorly characterised in humans. As such, its molecular composition, regulation and crosstalk with HR and other repair pathways remain elusive. Additionally, the contribution of the alt-EJ pathway to tumour progression as well as the identification of a mutational signature associated with the use of alt-EJ has not yet been investigated. Moreover, the clinical relevance of developing small-molecule inhibitors targeting players in the alt-EJ pathway, such as the polymerase Pol Theta (Polθ), is of importance as current anticancer drug treatments have shown limited effectiveness in achieving cancer remission in patients with HR-deficient (HRD) tumours.
Here, we propose a novel, multidisciplinary approach that aims to characterise the players and mechanisms of action involved in the utilisation of alt-EJ in cancer. This understanding will better elucidate the changing interplay between different DNA repair pathways, thus shedding light on whether and how the use of alt-EJ contributes to the pathogenic history and survival of HRD tumours, eventually paving the way for the development of novel anticancer therapeutics.
For all the abovementioned reasons, we are convinced this project will have important implications in: 1) elucidating critical interconnections between DNA repair pathways, 2) improving the basic understanding of the composition, regulation and function of the alt-EJ pathway, and 3) facilitating the development of new synthetic lethality-based chemotherapeutics for the treatment of HRD tumours.
Summary
DNA repair pathways evolved as an intricate network that senses DNA damage and resolves it in order to minimise genetic lesions and thus preventing tumour formation. Gaining in recognition the last few years, the alternative end-joining (alt-EJ) DNA repair pathway was recently shown to be up-regulated and required for cancer cell viability in the absence of homologous recombination-mediated repair (HR). Despite this integral role, the alt-EJ repair pathway remains poorly characterised in humans. As such, its molecular composition, regulation and crosstalk with HR and other repair pathways remain elusive. Additionally, the contribution of the alt-EJ pathway to tumour progression as well as the identification of a mutational signature associated with the use of alt-EJ has not yet been investigated. Moreover, the clinical relevance of developing small-molecule inhibitors targeting players in the alt-EJ pathway, such as the polymerase Pol Theta (Polθ), is of importance as current anticancer drug treatments have shown limited effectiveness in achieving cancer remission in patients with HR-deficient (HRD) tumours.
Here, we propose a novel, multidisciplinary approach that aims to characterise the players and mechanisms of action involved in the utilisation of alt-EJ in cancer. This understanding will better elucidate the changing interplay between different DNA repair pathways, thus shedding light on whether and how the use of alt-EJ contributes to the pathogenic history and survival of HRD tumours, eventually paving the way for the development of novel anticancer therapeutics.
For all the abovementioned reasons, we are convinced this project will have important implications in: 1) elucidating critical interconnections between DNA repair pathways, 2) improving the basic understanding of the composition, regulation and function of the alt-EJ pathway, and 3) facilitating the development of new synthetic lethality-based chemotherapeutics for the treatment of HRD tumours.
Max ERC Funding
1 498 750 €
Duration
Start date: 2017-07-01, End date: 2022-06-30
Project acronym ANGI
Project Adaptive significance of Non Genetic Inheritance
Researcher (PI) Benoit François Pujol
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS8, ERC-2015-CoG
Summary Our ability to predict adaptation and the response of populations to selection is limited. Solving this issue is a fundamental challenge of evolutionary ecology with implications for applied sciences such as conservation, and agronomy. Non genetic inheritance (NGI; e.g., ecological niche transmission) is suspected to play a foremost role in adaptive evolution but such hypothesis remains untested. Using quantitative genetics in wild plant populations, experimental evolution, and epigenetics, we will assess the role of NGI in the adaptive response to selection of plant populations. The ANGI project will follow the subsequent research program: (1) Using long-term survey data, we will measure natural selection in wild populations of Antirrhinum majus within its heterogeneous array of micro-habitats. We will calculate the fitness gain provided by multiple traits and stem elongation to plants growing in bushes where they compete for light. Stem elongation is known to depend on epigenetic variation. (2) Using a statistical approach that we developed, we will estimate the quantitative genetic and non genetic heritability of traits. (3) We will identify phenotypic changes caused by fitness that are based on genetic variation and NGI and assess their respective roles in adaptive evolution. (4) In controlled conditions, we will artificially select for increased stem elongation in clonal lineages, thereby excluding DNA variation. We will quantify the non genetic response to selection and test for a quantitative epigenetic signature of selection. (5) We will build on our results to generate an inclusive theory of genetic and non genetic natural selection. ANGI builds on a confirmed expertise in selection experiments, quantitative genetics and NGI. In addition, the availability of survey data provides a solid foundation for the achievement of this project. Our ambition is to shed light on original mechanisms underlying adaptation that are an alternative to genetic selection.
Summary
Our ability to predict adaptation and the response of populations to selection is limited. Solving this issue is a fundamental challenge of evolutionary ecology with implications for applied sciences such as conservation, and agronomy. Non genetic inheritance (NGI; e.g., ecological niche transmission) is suspected to play a foremost role in adaptive evolution but such hypothesis remains untested. Using quantitative genetics in wild plant populations, experimental evolution, and epigenetics, we will assess the role of NGI in the adaptive response to selection of plant populations. The ANGI project will follow the subsequent research program: (1) Using long-term survey data, we will measure natural selection in wild populations of Antirrhinum majus within its heterogeneous array of micro-habitats. We will calculate the fitness gain provided by multiple traits and stem elongation to plants growing in bushes where they compete for light. Stem elongation is known to depend on epigenetic variation. (2) Using a statistical approach that we developed, we will estimate the quantitative genetic and non genetic heritability of traits. (3) We will identify phenotypic changes caused by fitness that are based on genetic variation and NGI and assess their respective roles in adaptive evolution. (4) In controlled conditions, we will artificially select for increased stem elongation in clonal lineages, thereby excluding DNA variation. We will quantify the non genetic response to selection and test for a quantitative epigenetic signature of selection. (5) We will build on our results to generate an inclusive theory of genetic and non genetic natural selection. ANGI builds on a confirmed expertise in selection experiments, quantitative genetics and NGI. In addition, the availability of survey data provides a solid foundation for the achievement of this project. Our ambition is to shed light on original mechanisms underlying adaptation that are an alternative to genetic selection.
Max ERC Funding
1 999 970 €
Duration
Start date: 2016-03-01, End date: 2021-02-28
Project acronym AnoPath
Project Genetics of mosquito resistance to pathogens
Researcher (PI) Kenneth Du Souchet Vernick
Host Institution (HI) INSTITUT PASTEUR
Call Details Advanced Grant (AdG), LS2, ERC-2012-ADG_20120314
Summary Malaria parasite infection in humans has been called “the strongest known force for evolutionary selection in the recent history of the human genome”, and I hypothesize that a similar statement may apply to the mosquito vector, which is the definitive host of the malaria parasite. We previously discovered efficient malaria-resistance mechanisms in natural populations of the African malaria vector, Anopheles gambiae. Aim 1 of the proposed project will implement a novel genetic mapping design to systematically survey the mosquito population for common and rare genetic variants of strong effect against the human malaria parasite, Plasmodium falciparum. A product of the mapping design will be living mosquito families carrying the resistance loci. Aim 2 will use the segregating families to functionally dissect the underlying molecular mechanisms controlled by the loci, including determination of the pathogen specificity spectra of the host-defense traits. Aim 3 targets arbovirus transmission, where Anopheles mosquitoes transmit human malaria but not arboviruses such as Dengue and Chikungunya, even though the two mosquitoes bite the same people and are exposed to the same pathogens, often in malaria-arbovirus co-infections. We will use deep-sequencing to detect processing of the arbovirus dsRNA intermediates of replication produced by the RNAi pathway of the mosquitoes. The results will reveal important new information about differences in the efficiency and quality of the RNAi response between mosquitoes, which is likely to underlie at least part of the host specificity of arbovirus transmission. The 3 Aims will make significant contributions to understanding malaria and arbovirus transmission, major global public health problems, will aid the development of a next generation of vector surveillance and control tools, and will produce a definitive description of the major genetic factors influencing host-pathogen interactions in mosquito immunity.
Summary
Malaria parasite infection in humans has been called “the strongest known force for evolutionary selection in the recent history of the human genome”, and I hypothesize that a similar statement may apply to the mosquito vector, which is the definitive host of the malaria parasite. We previously discovered efficient malaria-resistance mechanisms in natural populations of the African malaria vector, Anopheles gambiae. Aim 1 of the proposed project will implement a novel genetic mapping design to systematically survey the mosquito population for common and rare genetic variants of strong effect against the human malaria parasite, Plasmodium falciparum. A product of the mapping design will be living mosquito families carrying the resistance loci. Aim 2 will use the segregating families to functionally dissect the underlying molecular mechanisms controlled by the loci, including determination of the pathogen specificity spectra of the host-defense traits. Aim 3 targets arbovirus transmission, where Anopheles mosquitoes transmit human malaria but not arboviruses such as Dengue and Chikungunya, even though the two mosquitoes bite the same people and are exposed to the same pathogens, often in malaria-arbovirus co-infections. We will use deep-sequencing to detect processing of the arbovirus dsRNA intermediates of replication produced by the RNAi pathway of the mosquitoes. The results will reveal important new information about differences in the efficiency and quality of the RNAi response between mosquitoes, which is likely to underlie at least part of the host specificity of arbovirus transmission. The 3 Aims will make significant contributions to understanding malaria and arbovirus transmission, major global public health problems, will aid the development of a next generation of vector surveillance and control tools, and will produce a definitive description of the major genetic factors influencing host-pathogen interactions in mosquito immunity.
Max ERC Funding
2 307 800 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym ANTIViR
Project Molecular mechanisms of interferon-induced antiviral restriction and signalling
Researcher (PI) Caroline GOUJON
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS6, ERC-2017-STG
Summary Interferons (IFNs), which are signalling proteins produced by infected cells, are the first line of defence against viral infections. IFNs induce, in infected and neighbouring cells, the expression of hundreds of IFN-stimulated genes (ISGs). The ISGs in turn induce in cells a potent antiviral state, capable of preventing replication of most viruses, including Human Immunodeficiency Virus type 1 (HIV-1) and influenza A virus (FLUAV). Identifying the antiviral ISGs and understanding their mechanisms of action is therefore crucial to progress in the fight against viruses.
ISGs playing a role in the antiviral state have been identified, such as human MX1, a well-known antiviral factor able to restrict numerous viruses including FLUAV, and MX2, an HIV-1 inhibitor. Both proteins bind to viral components but their detailed mechanisms of action, as well as the consequences of restriction on the activation of the innate immune system, remain unclear. Moreover, our preliminary work shows that additional anti-HIV-1 and anti-FLUAV ISGs remain to identify.
In this context, this proposal seeks an ERC StG funding to explore 3 major aims: 1) unravelling the mechanisms of antiviral action of MX proteins, by taking advantage of their similar structure and engineered chimeric proteins, and by using functional genetic screens to identify their cofactors; 2) investigating the consequences of incoming virus recognition by MX proteins on innate immune signalling, by altering their expression in target cells and measuring the cell response in terms of gene induction and cytokine production; 3) identifying and characterizing new ISGs able to inhibit viral replication with a combination of powerful approaches, including a whole-genome CRISPR/Cas9 knock-out screen.
Overall, this proposal will provide a better understanding of the molecular mechanisms involved in the antiviral effect of IFN, and may guide future efforts to identify novel therapeutic targets against major pathogenic viruses.
Summary
Interferons (IFNs), which are signalling proteins produced by infected cells, are the first line of defence against viral infections. IFNs induce, in infected and neighbouring cells, the expression of hundreds of IFN-stimulated genes (ISGs). The ISGs in turn induce in cells a potent antiviral state, capable of preventing replication of most viruses, including Human Immunodeficiency Virus type 1 (HIV-1) and influenza A virus (FLUAV). Identifying the antiviral ISGs and understanding their mechanisms of action is therefore crucial to progress in the fight against viruses.
ISGs playing a role in the antiviral state have been identified, such as human MX1, a well-known antiviral factor able to restrict numerous viruses including FLUAV, and MX2, an HIV-1 inhibitor. Both proteins bind to viral components but their detailed mechanisms of action, as well as the consequences of restriction on the activation of the innate immune system, remain unclear. Moreover, our preliminary work shows that additional anti-HIV-1 and anti-FLUAV ISGs remain to identify.
In this context, this proposal seeks an ERC StG funding to explore 3 major aims: 1) unravelling the mechanisms of antiviral action of MX proteins, by taking advantage of their similar structure and engineered chimeric proteins, and by using functional genetic screens to identify their cofactors; 2) investigating the consequences of incoming virus recognition by MX proteins on innate immune signalling, by altering their expression in target cells and measuring the cell response in terms of gene induction and cytokine production; 3) identifying and characterizing new ISGs able to inhibit viral replication with a combination of powerful approaches, including a whole-genome CRISPR/Cas9 knock-out screen.
Overall, this proposal will provide a better understanding of the molecular mechanisms involved in the antiviral effect of IFN, and may guide future efforts to identify novel therapeutic targets against major pathogenic viruses.
Max ERC Funding
1 499 794 €
Duration
Start date: 2017-12-01, End date: 2022-11-30
Project acronym ANTIVIRALRNAI
Project RNAi-mediated viral immunity in insects
Researcher (PI) Maria-Carla Saleh
Host Institution (HI) INSTITUT PASTEUR
Call Details Starting Grant (StG), LS6, ERC-2009-StG
Summary RNA interference (RNAi) is a conserved sequence-specific, gene-silencing mechanism that is induced by double-stranded RNA (dsRNA). One of the functions of this pathway is the defense against parasitic nucleic acids: transposons and viruses. Previous results demonstrated that viral infections in Drosophila melanogaster are fought by an antiviral RNAi response and that components of the endocytic pathway are required for dsRNA entry to initiate the RNAi response. Recently we have shown that infected insect cells spread a systemic silencing signal that elicits a protective RNAi-dependent immunity throughout the organism. This suggests that the cell-autonomous RNAi response is insufficient to control a viral infection and that flies also rely on systemic immune response to fight against such infections. As a junior group leader, I will study the mechanisms that mediate the RNAi-based antiviral response in insects. By combining biochemical, cellular, molecular and genomic approaches, both in vivo and in cell culture, I will analyze the mechanisms underlying viral tropism, systemic propagation of the antiviral signal and the basis of the persistence of the antiviral state. Furthermore, I will examine whether the dsRNA-uptake pathway is conserved in mosquitoes and its relationship with viral immunity in that host. This comprehensive approach will tackle how this nucleic acid-based immunity works in insects to generate an anti-viral stage. A better understanding of the role of RNA silencing in insects during virus infection will allow the exploitation of this pathway for improvement of public health related problems such as arbovirus infection and disease.
Summary
RNA interference (RNAi) is a conserved sequence-specific, gene-silencing mechanism that is induced by double-stranded RNA (dsRNA). One of the functions of this pathway is the defense against parasitic nucleic acids: transposons and viruses. Previous results demonstrated that viral infections in Drosophila melanogaster are fought by an antiviral RNAi response and that components of the endocytic pathway are required for dsRNA entry to initiate the RNAi response. Recently we have shown that infected insect cells spread a systemic silencing signal that elicits a protective RNAi-dependent immunity throughout the organism. This suggests that the cell-autonomous RNAi response is insufficient to control a viral infection and that flies also rely on systemic immune response to fight against such infections. As a junior group leader, I will study the mechanisms that mediate the RNAi-based antiviral response in insects. By combining biochemical, cellular, molecular and genomic approaches, both in vivo and in cell culture, I will analyze the mechanisms underlying viral tropism, systemic propagation of the antiviral signal and the basis of the persistence of the antiviral state. Furthermore, I will examine whether the dsRNA-uptake pathway is conserved in mosquitoes and its relationship with viral immunity in that host. This comprehensive approach will tackle how this nucleic acid-based immunity works in insects to generate an anti-viral stage. A better understanding of the role of RNA silencing in insects during virus infection will allow the exploitation of this pathway for improvement of public health related problems such as arbovirus infection and disease.
Max ERC Funding
1 900 000 €
Duration
Start date: 2009-10-01, End date: 2014-12-31
Project acronym APPL
Project Anionic PhosPhoLipids in plant receptor kinase signaling
Researcher (PI) Yvon Jaillais
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), LS3, ERC-2013-StG
Summary "In plants, receptor kinases form the largest family of plasma membrane (PM) receptors and they are involved in virtually all aspects of the plant life, including development, immunity and reproduction. In animals, key molecules that orchestrate the recruitment of signaling proteins to membranes are anionic phospholipids (e.g. phosphatidylinositol phosphate or PIPs). Besides, recent reports in animal and yeast cells suggest the existence of PM nanodomains that are independent of cholesterol and lipid phase and rely on anionic phospholipids as well as electrostatic protein/lipid interactions. Strikingly, we know very little on the role of anionic phospholipids in plant signaling. However, our preliminary data suggest that BKI1, an inhibitory protein of the steroid receptor kinase BRI1, interacts with various PIPs in vitro and is likely targeted to the PM by electrostatic interactions with these anionic lipids. These results open the possibility that BRI1, but also other receptor kinases, might be regulated by anionic phospholipids in plants. Here, we propose to analyze the function of anionic phospholipids in BRI1 signaling, using the root epidermis as a model system. First, we will ask what are the lipids that control membrane surface charge in this tissue and recruit BR-signaling component to the PM. Second, we will probe the presence of PIP-enriched nanodomains at the plant PM using super-resolution microscopy techniques and investigate the roles of these domains in BRI1 signaling. Finally, we will analyze the function of the BKI1-related plant-specific family of anionic phospholipid effectors in plant development. In summary, using a transversal approach ranging from in vitro studies to in vivo validation and whole organism physiology, this work will unravel the interplay between anionic phospholipids and receptor signaling in plants."
Summary
"In plants, receptor kinases form the largest family of plasma membrane (PM) receptors and they are involved in virtually all aspects of the plant life, including development, immunity and reproduction. In animals, key molecules that orchestrate the recruitment of signaling proteins to membranes are anionic phospholipids (e.g. phosphatidylinositol phosphate or PIPs). Besides, recent reports in animal and yeast cells suggest the existence of PM nanodomains that are independent of cholesterol and lipid phase and rely on anionic phospholipids as well as electrostatic protein/lipid interactions. Strikingly, we know very little on the role of anionic phospholipids in plant signaling. However, our preliminary data suggest that BKI1, an inhibitory protein of the steroid receptor kinase BRI1, interacts with various PIPs in vitro and is likely targeted to the PM by electrostatic interactions with these anionic lipids. These results open the possibility that BRI1, but also other receptor kinases, might be regulated by anionic phospholipids in plants. Here, we propose to analyze the function of anionic phospholipids in BRI1 signaling, using the root epidermis as a model system. First, we will ask what are the lipids that control membrane surface charge in this tissue and recruit BR-signaling component to the PM. Second, we will probe the presence of PIP-enriched nanodomains at the plant PM using super-resolution microscopy techniques and investigate the roles of these domains in BRI1 signaling. Finally, we will analyze the function of the BKI1-related plant-specific family of anionic phospholipid effectors in plant development. In summary, using a transversal approach ranging from in vitro studies to in vivo validation and whole organism physiology, this work will unravel the interplay between anionic phospholipids and receptor signaling in plants."
Max ERC Funding
1 797 840 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym ARBODYNAMIC
Project Coupling dynamic population immunity profiles and host behaviours to arboviral spread
Researcher (PI) Henrik SALJE
Host Institution (HI) INSTITUT PASTEUR
Call Details Starting Grant (StG), LS8, ERC-2018-STG
Summary Arboviruses infect millions of people each year, however, mechanisms that drive viral emergence and maintenance remain largely unknown. A combination of host factors (e.g., human mobility), mosquito factors (e.g., abundance) and viral factors (e.g., transmissibility) interconnect to drive spread. Further, for endemic arboviruses, complex patterns of population immunity, built up over many years, appear key to the emergence of particular lineages. To disentangle the contribution of these different drivers, we need detailed data from the same pathogen system over a long time period from the same location. In addition, we need new methods, which can integrate these different data sources and allow appropriate mechanistic inferences.
In this project, I will use the most globally prevalent arbovirus, dengue virus, as a case study. I will focus on Thailand where all four dengue serotypes have circulated endemically for decades and excellent long-term data and isolates exist, to address two fundamental questions:
i) How do population-level patterns of immunity evolve over time and what is their impact on strain dynamics? I will use mechanistic models applied to historic serotype-specific case data to reconstruct the evolving immune profile of the population and explore the impact of immunity on viral diversity using sequences from archived isolates from each year over a 50-year period.
ii) How do human behaviors, vector densities interact with immunity to dictate spread? I will work with geolocated full genome sequences from across Thailand and use detailed data on how people move, their contact patterns, their immunity profiles and mosquito distributions to study competing hypotheses of how arboviruses spread. I will compare the key drivers of dengue spread with that found for outbreaks of Zika and chikungunya.
This proposal addresses fundamental questions about the mechanisms that drive arboviral emergence and spread that will be relevant across disease systems.
Summary
Arboviruses infect millions of people each year, however, mechanisms that drive viral emergence and maintenance remain largely unknown. A combination of host factors (e.g., human mobility), mosquito factors (e.g., abundance) and viral factors (e.g., transmissibility) interconnect to drive spread. Further, for endemic arboviruses, complex patterns of population immunity, built up over many years, appear key to the emergence of particular lineages. To disentangle the contribution of these different drivers, we need detailed data from the same pathogen system over a long time period from the same location. In addition, we need new methods, which can integrate these different data sources and allow appropriate mechanistic inferences.
In this project, I will use the most globally prevalent arbovirus, dengue virus, as a case study. I will focus on Thailand where all four dengue serotypes have circulated endemically for decades and excellent long-term data and isolates exist, to address two fundamental questions:
i) How do population-level patterns of immunity evolve over time and what is their impact on strain dynamics? I will use mechanistic models applied to historic serotype-specific case data to reconstruct the evolving immune profile of the population and explore the impact of immunity on viral diversity using sequences from archived isolates from each year over a 50-year period.
ii) How do human behaviors, vector densities interact with immunity to dictate spread? I will work with geolocated full genome sequences from across Thailand and use detailed data on how people move, their contact patterns, their immunity profiles and mosquito distributions to study competing hypotheses of how arboviruses spread. I will compare the key drivers of dengue spread with that found for outbreaks of Zika and chikungunya.
This proposal addresses fundamental questions about the mechanisms that drive arboviral emergence and spread that will be relevant across disease systems.
Max ERC Funding
1 499 896 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym ARTTOUCH
Project Generating artificial touch: from the contribution of single tactile afferents to the encoding of complex percepts, and their implications for clinical innovation
Researcher (PI) Rochelle ACKERLEY
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS5, ERC-2017-COG
Summary Somatosensation encompass a wide range of processes, from feeling touch to temperature, as well as experiencing pleasure and pain. When afferent inputs are degraded or removed, such as in neuropathies or amputation, exploring the world becomes extremely difficult. Chronic pain is a major health issue that greatly diminishes quality of life and is one of the most disabling and costly conditions in Europe. The loss of a body part is common due to accidents, tumours, or peripheral diseases, and it has instantaneous effects on somatosensory functioning. Treating such disorders entails detailed knowledge about how somatosensory signals are encoded. Understanding these processes will enable the restoration of healthy function, such as providing real-time, naturalistic feedback in prostheses. To date, no prosthesis currently provides long-term sensory feedback, yet accomplishing this will lead to great quality of life improvements. The present proposal aims to uncover how basic tactile processes are encoded and represented centrally, as well as how more complex somatosensation is generated (e.g. wetness, pleasantness). Novel investigations will be conducted in humans to probe these mechanisms, including peripheral in vivo recording (microneurography) and neural stimulation, combined with advanced brain imaging and behavioural experiments. Preliminary work has shown the feasibility of the approach, where it is possible to visualise the activation of single mechanoreceptive afferents in the human brain. The multi-disciplinary approach unites detailed, high-resolution, functional investigations with actual sensations generated. The results will elucidate how basic and complex somatosensory processes are encoded, providing insights into the recovery of such signals. The knowledge gained aims to provide pain-free, efficient diagnostic capabilities for detecting and quantifying a range of somatosensory disorders, as well as identifying new potential therapeutic targets.
Summary
Somatosensation encompass a wide range of processes, from feeling touch to temperature, as well as experiencing pleasure and pain. When afferent inputs are degraded or removed, such as in neuropathies or amputation, exploring the world becomes extremely difficult. Chronic pain is a major health issue that greatly diminishes quality of life and is one of the most disabling and costly conditions in Europe. The loss of a body part is common due to accidents, tumours, or peripheral diseases, and it has instantaneous effects on somatosensory functioning. Treating such disorders entails detailed knowledge about how somatosensory signals are encoded. Understanding these processes will enable the restoration of healthy function, such as providing real-time, naturalistic feedback in prostheses. To date, no prosthesis currently provides long-term sensory feedback, yet accomplishing this will lead to great quality of life improvements. The present proposal aims to uncover how basic tactile processes are encoded and represented centrally, as well as how more complex somatosensation is generated (e.g. wetness, pleasantness). Novel investigations will be conducted in humans to probe these mechanisms, including peripheral in vivo recording (microneurography) and neural stimulation, combined with advanced brain imaging and behavioural experiments. Preliminary work has shown the feasibility of the approach, where it is possible to visualise the activation of single mechanoreceptive afferents in the human brain. The multi-disciplinary approach unites detailed, high-resolution, functional investigations with actual sensations generated. The results will elucidate how basic and complex somatosensory processes are encoded, providing insights into the recovery of such signals. The knowledge gained aims to provide pain-free, efficient diagnostic capabilities for detecting and quantifying a range of somatosensory disorders, as well as identifying new potential therapeutic targets.
Max ERC Funding
1 223 639 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym AstroWireSyn
Project Wiring synaptic circuits with astroglial connexins: mechanisms, dynamics and impact for critical period plasticity
Researcher (PI) Nathalie Rouach
Host Institution (HI) COLLEGE DE FRANCE
Call Details Consolidator Grant (CoG), LS5, ERC-2015-CoG
Summary Brain information processing is commonly thought to be a neuronal performance. However recent data point to a key role of astrocytes in brain development, activity and pathology. Indeed astrocytes are now viewed as crucial elements of the brain circuitry that control synapse formation, maturation, activity and elimination. How do astrocytes exert such control is matter of intense research, as they are now known to participate in critical developmental periods as well as in psychiatric disorders involving synapse alterations. Thus unraveling how astrocytes control synaptic circuit formation and maturation is crucial, not only for our understanding of brain development, but also for identifying novel therapeutic targets.
We recently found that connexin 30 (Cx30), an astroglial gap junction subunit expressed postnatally, tunes synaptic activity via an unprecedented non-channel function setting the proximity of glial processes to synaptic clefts, essential for synaptic glutamate clearance efficacy. Our work not only reveals Cx30 as a key determinant of glial synapse coverage, but also extends the classical model of neuroglial interactions in which astrocytes are generally considered as extrasynaptic elements indirectly regulating neurotransmission. Yet the molecular mechanisms involved in such control, its dynamic regulation by activity and impact in a native developmental context are unknown. We will now address these important questions, focusing on the involvement of this novel astroglial function in wiring developing synaptic circuits.
Thus using a multidisciplinary approach we will investigate:
1) the molecular and cellular mechanisms underlying Cx30 regulation of synaptic function
2) the activity-dependent dynamics of Cx30 function at synapses
3) a role for Cx30 in wiring synaptic circuits during critical developmental periods
This ambitious project will provide essential knowledge on the molecular mechanisms underlying astroglial control of synaptic circuits.
Summary
Brain information processing is commonly thought to be a neuronal performance. However recent data point to a key role of astrocytes in brain development, activity and pathology. Indeed astrocytes are now viewed as crucial elements of the brain circuitry that control synapse formation, maturation, activity and elimination. How do astrocytes exert such control is matter of intense research, as they are now known to participate in critical developmental periods as well as in psychiatric disorders involving synapse alterations. Thus unraveling how astrocytes control synaptic circuit formation and maturation is crucial, not only for our understanding of brain development, but also for identifying novel therapeutic targets.
We recently found that connexin 30 (Cx30), an astroglial gap junction subunit expressed postnatally, tunes synaptic activity via an unprecedented non-channel function setting the proximity of glial processes to synaptic clefts, essential for synaptic glutamate clearance efficacy. Our work not only reveals Cx30 as a key determinant of glial synapse coverage, but also extends the classical model of neuroglial interactions in which astrocytes are generally considered as extrasynaptic elements indirectly regulating neurotransmission. Yet the molecular mechanisms involved in such control, its dynamic regulation by activity and impact in a native developmental context are unknown. We will now address these important questions, focusing on the involvement of this novel astroglial function in wiring developing synaptic circuits.
Thus using a multidisciplinary approach we will investigate:
1) the molecular and cellular mechanisms underlying Cx30 regulation of synaptic function
2) the activity-dependent dynamics of Cx30 function at synapses
3) a role for Cx30 in wiring synaptic circuits during critical developmental periods
This ambitious project will provide essential knowledge on the molecular mechanisms underlying astroglial control of synaptic circuits.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym Autophagy in vitro
Project Reconstituting Autophagosome Biogenesis in vitro
Researcher (PI) Thomas Wollert
Host Institution (HI) INSTITUT PASTEUR
Call Details Starting Grant (StG), LS1, ERC-2014-STG
Summary Autophagy is a catabolic pathway that delivers cytoplasmic material to lysosomes for degradation. Under vegetative conditions, the pathway serves as quality control system, specifically targeting damaged or superfluous organelles and protein-aggregates. Cytotoxic stresses and starvation, however, induces the formation of larger autophagosomes that capture cargo unselectively. Autophagosomes are being generated from a cup-shaped precursor membrane, the isolation membrane, which expands to engulf cytoplasmic components. Sealing of this structure gives rise to the double-membrane surrounded autophagosomes. Two interconnected ubiquitin (Ub)-like conjugation systems coordinate the expansion of autophagosomes by conjugating the autophagy related (Atg)-protein Atg8 to the isolation membrane. In an effort to unravel the function of Atg8, we reconstituted the system on model membranes in vitro and found that Atg8 forms together with the Atg12–Atg5-Atg16 complex a membrane scaffold which is required for productive autophagy in yeast. Humans possess seven Atg8-homologs and two mutually exclusive Atg16-variants. Here, we propose to investigate the function of the human Ub-like conjugation system using a fully reconstituted in vitro system. The spatiotemporal organization of recombinant fluorescent-labeled proteins with synthetic model membranes will be investigated using confocal and TIRF-microscopy. Structural information will be obtained by atomic force and electron microscopy. Mechanistic insights, obtained from the in vitro work, will be tested in vivo in cultured human cells. We belief that revealing 1) the function of the human Ub-like conjugation system in autophagy, 2) the functional differences of Atg8-homologs and the two Atg16-variants Atg16L1 and TECPR1 and 3) how Atg16L1 coordinates non-canonical autophagy will provide essential insights into the pathophysiology of cancer, neurodegenerative, and autoimmune diseases.
Summary
Autophagy is a catabolic pathway that delivers cytoplasmic material to lysosomes for degradation. Under vegetative conditions, the pathway serves as quality control system, specifically targeting damaged or superfluous organelles and protein-aggregates. Cytotoxic stresses and starvation, however, induces the formation of larger autophagosomes that capture cargo unselectively. Autophagosomes are being generated from a cup-shaped precursor membrane, the isolation membrane, which expands to engulf cytoplasmic components. Sealing of this structure gives rise to the double-membrane surrounded autophagosomes. Two interconnected ubiquitin (Ub)-like conjugation systems coordinate the expansion of autophagosomes by conjugating the autophagy related (Atg)-protein Atg8 to the isolation membrane. In an effort to unravel the function of Atg8, we reconstituted the system on model membranes in vitro and found that Atg8 forms together with the Atg12–Atg5-Atg16 complex a membrane scaffold which is required for productive autophagy in yeast. Humans possess seven Atg8-homologs and two mutually exclusive Atg16-variants. Here, we propose to investigate the function of the human Ub-like conjugation system using a fully reconstituted in vitro system. The spatiotemporal organization of recombinant fluorescent-labeled proteins with synthetic model membranes will be investigated using confocal and TIRF-microscopy. Structural information will be obtained by atomic force and electron microscopy. Mechanistic insights, obtained from the in vitro work, will be tested in vivo in cultured human cells. We belief that revealing 1) the function of the human Ub-like conjugation system in autophagy, 2) the functional differences of Atg8-homologs and the two Atg16-variants Atg16L1 and TECPR1 and 3) how Atg16L1 coordinates non-canonical autophagy will provide essential insights into the pathophysiology of cancer, neurodegenerative, and autoimmune diseases.
Max ERC Funding
1 499 726 €
Duration
Start date: 2015-04-01, End date: 2020-03-31
Project acronym B-response
Project Memory and innate-like B-cell subsets: deciphering a multi-layered B-cell response in mice and humans
Researcher (PI) Claude-Agnes REYNAUD
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS6, ERC-2015-AdG
Summary B cells are the main actors of successful vaccines, and their protective capacity relies on several subsets with innate-like and memory properties that fulfill different effector functions. In the present project, we wish to develop approaches in both mice and humans, to confront the similarities and the differences of their B cell responses.
The three aims proposed are:
1) To study the different B cell subsets and TFH cells engaged in a memory response through the use of a new mouse reporter line allowing their irreversible labeling (inducible Cre recombinase under the control of the Bcl6 gene): this will be performed in different conditions of TH1 vs. TH2 polarization, as well as during a chronic viral infection, in which virus-specific antibodies have been shown to be required to control the disease (in collaboration with D. Pinschewer, Basel)
2) To study whether the lifelong persistence of B cell memory, as occurs for memory B cells against smallpox that we can obtain at high purity from aged donor's spleens, corresponds to a specific transcriptional program at the miRNA, lncRNA or mRNA level, as well as a specific cell homeostasis
3) To discriminate the specific effector function of human marginal zone and IgM memory B cells in, respectively, T-independent and T-dependent responses, as well as their specific differentiation/diversification pathway.
The general goal is to delineate the regulatory pathways leading to the activation and persistence of the different B cell subsets, allowing for a better understanding of the conditions leading to their pathological or beneficial mobilization.
Summary
B cells are the main actors of successful vaccines, and their protective capacity relies on several subsets with innate-like and memory properties that fulfill different effector functions. In the present project, we wish to develop approaches in both mice and humans, to confront the similarities and the differences of their B cell responses.
The three aims proposed are:
1) To study the different B cell subsets and TFH cells engaged in a memory response through the use of a new mouse reporter line allowing their irreversible labeling (inducible Cre recombinase under the control of the Bcl6 gene): this will be performed in different conditions of TH1 vs. TH2 polarization, as well as during a chronic viral infection, in which virus-specific antibodies have been shown to be required to control the disease (in collaboration with D. Pinschewer, Basel)
2) To study whether the lifelong persistence of B cell memory, as occurs for memory B cells against smallpox that we can obtain at high purity from aged donor's spleens, corresponds to a specific transcriptional program at the miRNA, lncRNA or mRNA level, as well as a specific cell homeostasis
3) To discriminate the specific effector function of human marginal zone and IgM memory B cells in, respectively, T-independent and T-dependent responses, as well as their specific differentiation/diversification pathway.
The general goal is to delineate the regulatory pathways leading to the activation and persistence of the different B cell subsets, allowing for a better understanding of the conditions leading to their pathological or beneficial mobilization.
Max ERC Funding
2 098 750 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym BacCellEpi
Project Bacterial, cellular and epigenetic factors that control enteropathogenicity
Researcher (PI) Pascale Cossart
Host Institution (HI) INSTITUT PASTEUR
Call Details Advanced Grant (AdG), LS6, ERC-2014-ADG
Summary Understanding the establishment and persistence of bacterial infections in the gut requires integrating an ensemble of factors including bacterial and host components and the presence of other microorganisms. We will capitalize on 25 years of studies on the bacterium Listeria monocytogenes used as a model, to focus on three objectives which will significantly increase our knowledge of the bacterium, of the cell biology of infection and of the epigenetic reprogramming upon infection.
Our aims are:
- at the bacterial level : to describe for the first time, the proteomic landscape of a bacterium during switch from saprophytism to virulence. We will use a proteogenomic approach together with ribosome profiling, to analyze the translation of the whole transcriptome after bacterial growth in several conditions, including in vivo, in order to barcode all the proteins which play a role in infection. This will open the way to assess the role of 1) small proteins; 2) internal translation initiation sites ; 3) the coupling of transcription and translation.
- at the host cell level : To decipher the molecular mechanisms underlying the dynamics and role in infection of host intracellular organelles, starting with mitochondria.
- At the host epigenetic level : To explore how the microbe reprograms host transcription and how tolerance to a commensal such as Akkermansia muciniphila differs from responsiveness to a pathogen insult, at the level of histones and mRNA modifications by studying 1) chromatin remodeling, in particular histones modifications during infection ; 2) modifications of the epitranscriptome during Listeria infection and colonization with Akkermansia ; 3) whether there is an epigenetic memory of infection and colonization.
This ambitious multidisciplinary project will not only generate new concepts in infection biology but also will unravel fundamental mechanisms in microbiology, cell biology, and epigenetics opening new avenues for further research.
Summary
Understanding the establishment and persistence of bacterial infections in the gut requires integrating an ensemble of factors including bacterial and host components and the presence of other microorganisms. We will capitalize on 25 years of studies on the bacterium Listeria monocytogenes used as a model, to focus on three objectives which will significantly increase our knowledge of the bacterium, of the cell biology of infection and of the epigenetic reprogramming upon infection.
Our aims are:
- at the bacterial level : to describe for the first time, the proteomic landscape of a bacterium during switch from saprophytism to virulence. We will use a proteogenomic approach together with ribosome profiling, to analyze the translation of the whole transcriptome after bacterial growth in several conditions, including in vivo, in order to barcode all the proteins which play a role in infection. This will open the way to assess the role of 1) small proteins; 2) internal translation initiation sites ; 3) the coupling of transcription and translation.
- at the host cell level : To decipher the molecular mechanisms underlying the dynamics and role in infection of host intracellular organelles, starting with mitochondria.
- At the host epigenetic level : To explore how the microbe reprograms host transcription and how tolerance to a commensal such as Akkermansia muciniphila differs from responsiveness to a pathogen insult, at the level of histones and mRNA modifications by studying 1) chromatin remodeling, in particular histones modifications during infection ; 2) modifications of the epitranscriptome during Listeria infection and colonization with Akkermansia ; 3) whether there is an epigenetic memory of infection and colonization.
This ambitious multidisciplinary project will not only generate new concepts in infection biology but also will unravel fundamental mechanisms in microbiology, cell biology, and epigenetics opening new avenues for further research.
Max ERC Funding
1 147 500 €
Duration
Start date: 2015-10-01, End date: 2019-09-30
Project acronym BACEMO
Project Bacterial Cell Morphogenesis
Researcher (PI) Rut Carballido Lopez
Host Institution (HI) INSTITUT NATIONAL DE LA RECHERCHE AGRONOMIQUE
Call Details Starting Grant (StG), LS3, ERC-2012-StG_20111109
Summary In bacteria, the though external cell wall and the intracellular actin-like (MreB) cytoskeleton are major determinants of cell shape. The biosynthetic pathways and chemical composition of the cell wall, a three dimensional polymer network that is one of the most prominent targets for antibiotics, are well understood. However, despite decades of study, little is known about the complex cell wall ultrastructure and the molecular mechanisms that control cell wall morphogenesis in time and space. In rod-shaped bacteria, MreB homologues assemble into dynamic structures thought to control shape by serving as organizers for the movement and assembly of macromolecular machineries that effect sidewall elongation. However, the mechanistic details used by the MreB cytoskeleton to fulfil this role remain to be elucidated. Furthermore, development of high-resolution microscopy techniques has led to new breakthroughs this year, published by our lab and others, which are shaking the model developed over the last decade and re-questioning the MreB “actin cytoskeleton” designation.
The aim of this project is to combine powerful genetic, biochemical, genomic and systems biology approaches available in the model bacterium Bacillus subtilis with modern high-resolution light microscopic techniques to study the dynamics and mechanistic details of the MreB cytoskeleton and of CW assembly. Parameters measured by the different approaches will be combined to quantitatively describe the features of bacterial cell morphogenesis.
Summary
In bacteria, the though external cell wall and the intracellular actin-like (MreB) cytoskeleton are major determinants of cell shape. The biosynthetic pathways and chemical composition of the cell wall, a three dimensional polymer network that is one of the most prominent targets for antibiotics, are well understood. However, despite decades of study, little is known about the complex cell wall ultrastructure and the molecular mechanisms that control cell wall morphogenesis in time and space. In rod-shaped bacteria, MreB homologues assemble into dynamic structures thought to control shape by serving as organizers for the movement and assembly of macromolecular machineries that effect sidewall elongation. However, the mechanistic details used by the MreB cytoskeleton to fulfil this role remain to be elucidated. Furthermore, development of high-resolution microscopy techniques has led to new breakthroughs this year, published by our lab and others, which are shaking the model developed over the last decade and re-questioning the MreB “actin cytoskeleton” designation.
The aim of this project is to combine powerful genetic, biochemical, genomic and systems biology approaches available in the model bacterium Bacillus subtilis with modern high-resolution light microscopic techniques to study the dynamics and mechanistic details of the MreB cytoskeleton and of CW assembly. Parameters measured by the different approaches will be combined to quantitatively describe the features of bacterial cell morphogenesis.
Max ERC Funding
1 650 050 €
Duration
Start date: 2013-02-01, End date: 2019-01-31
Project acronym BACTIN
Project Shaping the bacterial cell wall: the actin-like cytoskeleton, from single molecules to morphogenesis and antimicrobials
Researcher (PI) Rut CARBALLIDO LOPEZ
Host Institution (HI) INSTITUT NATIONAL DE LA RECHERCHE AGRONOMIQUE
Call Details Consolidator Grant (CoG), LS3, ERC-2017-COG
Summary One of the ultimate goals in cell biology is to understand how cells determine their shape. In bacteria, the cell wall and the actin-like (MreB) cytoskeleton are major determinants of cell shape. As a hallmark of microbial life, the external cell wall is the most conspicuous macromolecule expanding in concert with cell growth and one of the most prominent targets for antibiotics. Despite decades of study, the mechanism of cell wall morphogenesis remains poorly understood. In rod-shaped bacteria, actin-like MreB proteins assemble into disconnected membrane-associated structures (patches) that move processively around the cell periphery and are thought to control shape by spatiotemporally organizing macromolecular machineries that effect sidewall elongation. However, the ultrastructure of MreB assemblies and the mechanistic details underlying their morphogenetic function remain to be elucidated.
The aim of this project is to combine ground-breaking light microscopy and spectroscopy techniques with cutting-edge genetic, biochemical and systems biology approaches available in the model rod-shaped bacterium Bacillus subtilis to elucidate how MreB and cell wall biosynthetic enzymes collectively act to build a cell. Within this context, new features of MreB assemblies will be determined in vivo and in vitro, and a “toolbox” of approaches to determine the modes of action of antibiotics targeting cell wall processes will be developed. Parameters measured by the different approaches will be used to refine a mathematical model aiming to quantitatively describe the features of bacterial cell wall growth. The long-term goals of BActin are to understand general principles of bacterial cell morphogenesis and to provide mechanistic templates and new reporters for the screening of novel antibiotics.
Summary
One of the ultimate goals in cell biology is to understand how cells determine their shape. In bacteria, the cell wall and the actin-like (MreB) cytoskeleton are major determinants of cell shape. As a hallmark of microbial life, the external cell wall is the most conspicuous macromolecule expanding in concert with cell growth and one of the most prominent targets for antibiotics. Despite decades of study, the mechanism of cell wall morphogenesis remains poorly understood. In rod-shaped bacteria, actin-like MreB proteins assemble into disconnected membrane-associated structures (patches) that move processively around the cell periphery and are thought to control shape by spatiotemporally organizing macromolecular machineries that effect sidewall elongation. However, the ultrastructure of MreB assemblies and the mechanistic details underlying their morphogenetic function remain to be elucidated.
The aim of this project is to combine ground-breaking light microscopy and spectroscopy techniques with cutting-edge genetic, biochemical and systems biology approaches available in the model rod-shaped bacterium Bacillus subtilis to elucidate how MreB and cell wall biosynthetic enzymes collectively act to build a cell. Within this context, new features of MreB assemblies will be determined in vivo and in vitro, and a “toolbox” of approaches to determine the modes of action of antibiotics targeting cell wall processes will be developed. Parameters measured by the different approaches will be used to refine a mathematical model aiming to quantitatively describe the features of bacterial cell wall growth. The long-term goals of BActin are to understand general principles of bacterial cell morphogenesis and to provide mechanistic templates and new reporters for the screening of novel antibiotics.
Max ERC Funding
1 902 195 €
Duration
Start date: 2019-02-01, End date: 2024-01-31
Project acronym BactRNA
Project Bacterial small RNAs networks unravelling novel features of transcription and translation
Researcher (PI) Maude Audrey Guillier
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS2, ERC-2018-COG
Summary Regulation of gene expression plays a key role in the ability of bacteria to rapidly adapt to changing environments and to colonize extremely diverse habitats. The relatively recent discovery of a plethora of small regulatory RNAs and the beginning of their characterization has unravelled new aspects of bacterial gene expression. First, the expression of many bacterial genes responds to a complex network of both transcriptional and post-transcriptional regulators. However, the properties of the resulting regulatory circuits on the dynamics of gene expression and in the bacterial adaptive response have been poorly addressed so far. In a first part of this project, we will tackle this question by characterizing the circuits that are formed between two widespread classes of bacterial regulators, the sRNAs and the two-component systems, which act at the post-transcriptional and the transcriptional level, respectively. The study of sRNAs also led to major breakthroughs regarding the basic mechanisms of gene expression. In particular, we recently showed that repressor sRNAs can target activating stem-loop structures located within the coding region of mRNAs that promote translation initiation, in striking contrast with the previously recognized inhibitory role of mRNA structures in translation. The second objective of this project is thus to draw an unprecedented map of non-canonical translation initiation events and their regulation by sRNAs.
Overall, this project will greatly improve our understanding of how bacteria can so rapidly and successfully adapt to many different environments, and in the long term, provide clues towards the development of anti-bacterial strategies.
Summary
Regulation of gene expression plays a key role in the ability of bacteria to rapidly adapt to changing environments and to colonize extremely diverse habitats. The relatively recent discovery of a plethora of small regulatory RNAs and the beginning of their characterization has unravelled new aspects of bacterial gene expression. First, the expression of many bacterial genes responds to a complex network of both transcriptional and post-transcriptional regulators. However, the properties of the resulting regulatory circuits on the dynamics of gene expression and in the bacterial adaptive response have been poorly addressed so far. In a first part of this project, we will tackle this question by characterizing the circuits that are formed between two widespread classes of bacterial regulators, the sRNAs and the two-component systems, which act at the post-transcriptional and the transcriptional level, respectively. The study of sRNAs also led to major breakthroughs regarding the basic mechanisms of gene expression. In particular, we recently showed that repressor sRNAs can target activating stem-loop structures located within the coding region of mRNAs that promote translation initiation, in striking contrast with the previously recognized inhibitory role of mRNA structures in translation. The second objective of this project is thus to draw an unprecedented map of non-canonical translation initiation events and their regulation by sRNAs.
Overall, this project will greatly improve our understanding of how bacteria can so rapidly and successfully adapt to many different environments, and in the long term, provide clues towards the development of anti-bacterial strategies.
Max ERC Funding
1 999 754 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym BASILIC
Project Decoding at systems-level the crosstalk between the T cell antigen receptor, the CD28 costimulator and the PD-1 coinhibitor under physiological and pathological conditions.
Researcher (PI) Bernard MALISSEN
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS6, ERC-2017-ADG
Summary Although the T cell antigen receptor (TCR) occupies a central place in T cell physiology, it does not work in isolation and the signals it triggers are tuned by receptors that convey positive (costimulators) and negative (coinhibitors) informations. We lack a satisfying comprehension of the way T cells integrate such multiple inputs to make informed decisions. The proteomics-based methodology we developed around the TCR places us in a favorable situation to decode at systems-level the crosstalk between the TCR, the CD28 costimulator and the PD-1 coinhibitor signaling pathways. The novelty of our approach stems from (1) its use of primary T cells, (2) its capacity to probe the architecture and dynamics of signalosomes resulting from T cell-antigen presenting cell encounters, (3) the attention we pay to the stoichiometry of the studied signalosomes, a key parameter largely ignored in previous studies, and (4) its multidisciplinary nature straddling molecular and organismal scales.
Our specific aims are:
Aim 1. To understand how the TCR and CD28 signaling pathways cooperate to achieve optimal T cell responses.
Aim 2. To determine whether CD28 is the sole target of the PD-1 coinhibitor.
Aim 3. To determine how under inflammatory conditions CD28 functions can be superseded by those of OX40, a costimulator of the TNFR superfamily.
Aim 4. To unveil how malfunctions of LAT, a key signaling hub used by the TCR, disrupt the TCR-CD28 crosstalk and result in unique pathogenic T cells that by becoming ‘autistic’ to TCR signals and addicted to CD28 signals lead to severe immunopathologies.
We think that combining genetic, epigenomics, proteomics, and computational approaches creates ideal experimental conditions to understand at system-levels how TCR, costimulatory, coinhibitory and inflammatory signals are integrated during T cell clonal expansion. Although of fundamental nature, our project should help understanding the harmful role PD-1 plays during anti-tumoral responses.
Summary
Although the T cell antigen receptor (TCR) occupies a central place in T cell physiology, it does not work in isolation and the signals it triggers are tuned by receptors that convey positive (costimulators) and negative (coinhibitors) informations. We lack a satisfying comprehension of the way T cells integrate such multiple inputs to make informed decisions. The proteomics-based methodology we developed around the TCR places us in a favorable situation to decode at systems-level the crosstalk between the TCR, the CD28 costimulator and the PD-1 coinhibitor signaling pathways. The novelty of our approach stems from (1) its use of primary T cells, (2) its capacity to probe the architecture and dynamics of signalosomes resulting from T cell-antigen presenting cell encounters, (3) the attention we pay to the stoichiometry of the studied signalosomes, a key parameter largely ignored in previous studies, and (4) its multidisciplinary nature straddling molecular and organismal scales.
Our specific aims are:
Aim 1. To understand how the TCR and CD28 signaling pathways cooperate to achieve optimal T cell responses.
Aim 2. To determine whether CD28 is the sole target of the PD-1 coinhibitor.
Aim 3. To determine how under inflammatory conditions CD28 functions can be superseded by those of OX40, a costimulator of the TNFR superfamily.
Aim 4. To unveil how malfunctions of LAT, a key signaling hub used by the TCR, disrupt the TCR-CD28 crosstalk and result in unique pathogenic T cells that by becoming ‘autistic’ to TCR signals and addicted to CD28 signals lead to severe immunopathologies.
We think that combining genetic, epigenomics, proteomics, and computational approaches creates ideal experimental conditions to understand at system-levels how TCR, costimulatory, coinhibitory and inflammatory signals are integrated during T cell clonal expansion. Although of fundamental nature, our project should help understanding the harmful role PD-1 plays during anti-tumoral responses.
Max ERC Funding
2 000 000 €
Duration
Start date: 2018-08-01, End date: 2022-07-31
Project acronym BayCellS
Project A Bayesian Framework for Cellular Structural Biology
Researcher (PI) Michael Nilges
Host Institution (HI) INSTITUT PASTEUR
Call Details Advanced Grant (AdG), LS1, ERC-2011-ADG_20110310
Summary The functioning of a single cell or organism is governed by the laws of chemistry and physics. The bridge from biology to chemistry and physics is provided by structural biology: to understand the functioning of a cell, it is necessary to know the atomic structure of macromolecular assemblies, which may contain hundreds of components. To characterise the structures of the increasingly large and often flexible complexes, high resolution structure determination (as was possible for example for the ribosome) will likely stay the exception, and multiple sources of structural data at multiple resolutions are employed. Integrating these data into one consistent picture poses particular difficulties, since data are much more sparse than in high resolution methods, and the data sets from heterogeneous sources are of highly different and unknown quality and may be mutually inconsistent, and that data are in general averaged over large ensembles and long times. Molecular modelling, a crucial element of any structure determination, plays an even more important role in these multi-scale and multi-technique approaches, not only to obtain structures from the data, but also to evaluate their reliability. This proposal is to develop a consistent framework for this highly complex data integration problem, principally based on Bayesian probability theory. Appropriate models for the major types data types used in hybrid approaches will be developed, as well as representations to include structural knowledge for the components of the complexes, at multiple scales. The new methods will be applied to a series of problems with increasing complexity, going from the determination of protein complexes with high resolution information, over low resolution structures based on protein-protein interaction data such as the nuclear pore, to the genome organisation in the nucleus.
Summary
The functioning of a single cell or organism is governed by the laws of chemistry and physics. The bridge from biology to chemistry and physics is provided by structural biology: to understand the functioning of a cell, it is necessary to know the atomic structure of macromolecular assemblies, which may contain hundreds of components. To characterise the structures of the increasingly large and often flexible complexes, high resolution structure determination (as was possible for example for the ribosome) will likely stay the exception, and multiple sources of structural data at multiple resolutions are employed. Integrating these data into one consistent picture poses particular difficulties, since data are much more sparse than in high resolution methods, and the data sets from heterogeneous sources are of highly different and unknown quality and may be mutually inconsistent, and that data are in general averaged over large ensembles and long times. Molecular modelling, a crucial element of any structure determination, plays an even more important role in these multi-scale and multi-technique approaches, not only to obtain structures from the data, but also to evaluate their reliability. This proposal is to develop a consistent framework for this highly complex data integration problem, principally based on Bayesian probability theory. Appropriate models for the major types data types used in hybrid approaches will be developed, as well as representations to include structural knowledge for the components of the complexes, at multiple scales. The new methods will be applied to a series of problems with increasing complexity, going from the determination of protein complexes with high resolution information, over low resolution structures based on protein-protein interaction data such as the nuclear pore, to the genome organisation in the nucleus.
Max ERC Funding
2 130 212 €
Duration
Start date: 2012-05-01, End date: 2017-04-30
Project acronym BEHAVIORAL THEORY
Project Behavioral Theory and Economic Applications
Researcher (PI) Botond Koszegi
Host Institution (HI) KOZEP-EUROPAI EGYETEM
Call Details Starting Grant (StG), SH1, ERC-2012-StG_20111124
Summary "This proposal outlines projects to develop robust and portable theories studying the impact of psychological phenomena in economic settings. The proposed work falls in three broad research agendas.
My first main agenda is to formally model and economically apply a simple observation: that when people make decisions, they do not focus equally on all attributes of their available options, and overweight the attributes they focus on. I will build a set of portable models of focusing in attribute-based choice and risky choice based on the idea that a person focuses more on attributes in which her options differ more. I will also use the framework to develop novel, focus-based, theories of intertemporal choice and social preferences, as well as analyze the implications of focusing for product design, principal-agent relationships, and other economic questions.
My second main agenda is to explore some implications for market outcomes, welfare, and policy of the possibility that consumers misperceive certain aspects of products. I will investigate the circumstances that facilitate the profitable deception of consumers; firms' incentives for ""innovating"" deceptive products, including novel financial products aimed at exploiting investors; how firms' ability to distinguish naive and sophisticated consumers affects the consequences of deception; whether learning on the part of consumers will help them to avoid making mistakes; and how regulators and other observers can detect consumer mistakes from market data.
Two further projects apply the model of reference-dependent utility I have developed in earlier work to understand the pricing and advertising behavior of firms. I will also aim to disseminate some of my work, along with other cutting-edge research in psychology and economics, in a Journal of Economic Literature survey on ""Behavioral Contract Theory."""
Summary
"This proposal outlines projects to develop robust and portable theories studying the impact of psychological phenomena in economic settings. The proposed work falls in three broad research agendas.
My first main agenda is to formally model and economically apply a simple observation: that when people make decisions, they do not focus equally on all attributes of their available options, and overweight the attributes they focus on. I will build a set of portable models of focusing in attribute-based choice and risky choice based on the idea that a person focuses more on attributes in which her options differ more. I will also use the framework to develop novel, focus-based, theories of intertemporal choice and social preferences, as well as analyze the implications of focusing for product design, principal-agent relationships, and other economic questions.
My second main agenda is to explore some implications for market outcomes, welfare, and policy of the possibility that consumers misperceive certain aspects of products. I will investigate the circumstances that facilitate the profitable deception of consumers; firms' incentives for ""innovating"" deceptive products, including novel financial products aimed at exploiting investors; how firms' ability to distinguish naive and sophisticated consumers affects the consequences of deception; whether learning on the part of consumers will help them to avoid making mistakes; and how regulators and other observers can detect consumer mistakes from market data.
Two further projects apply the model of reference-dependent utility I have developed in earlier work to understand the pricing and advertising behavior of firms. I will also aim to disseminate some of my work, along with other cutting-edge research in psychology and economics, in a Journal of Economic Literature survey on ""Behavioral Contract Theory."""
Max ERC Funding
1 275 448 €
Duration
Start date: 2012-11-01, End date: 2018-10-31
Project acronym BetaRegeneration
Project Induction of Insulin-producing beta-cells Regeneration in vivo
Researcher (PI) Patrick Collombat
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary Diabetes has become one of the most widespread metabolic disorders with epidemic dimensions affecting almost 6% of the world’s population. Despite modern treatments, the life expectancy of patients with Type 1 diabetes remains reduced as compared to healthy subjects. There is therefore a need for alternative therapies. Towards this aim, using the mouse, we recently demonstrated that the in vivo forced expression of a single factor in pancreatic alpha-cells is sufficient to induce a continuous regeneration of alpha-cells and their subsequent conversion into beta-like cells, such converted cells being capable of reversing the consequences of chemically-induced diabetes in vivo (Collombat et al. Cell, 2009).
The PI and his team therefore propose to further decipher the mechanisms involved in this alpha-cell-mediated beta-cell regeneration process and determine whether this approach may be applied to adult animals and whether it would efficiently reverse Type 1 diabetes. Furthermore, a major effort will be made to verify whether our findings could be translated to human. Specifically, we will use a tri-partite approach to address the following issues: (1) Can the in vivo alpha-cell-mediated beta-cell regeneration be induced in adults mice? What would be the genetic determinants involved? (2) Can alpha-cell-mediated beta-cell regeneration reverse diabetes in the NOD Type 1 diabetes mouse model? (3) Can adult human alpha-cells be converted into beta-like cells?
Together, these ambitious objectives will most certainly allow us to gain new insight into the mechanisms defining the identity and the reprogramming capabilities of mouse and human endocrine cells and may thereby open new avenues for the treatment of diabetes. Similarly, the determination of the molecular triggers implicated in the beta-cell regeneration observed in our diabetic mice may lead to exciting new findings, including the identification of “drugable” targets of importance for human diabetic patients.
Summary
Diabetes has become one of the most widespread metabolic disorders with epidemic dimensions affecting almost 6% of the world’s population. Despite modern treatments, the life expectancy of patients with Type 1 diabetes remains reduced as compared to healthy subjects. There is therefore a need for alternative therapies. Towards this aim, using the mouse, we recently demonstrated that the in vivo forced expression of a single factor in pancreatic alpha-cells is sufficient to induce a continuous regeneration of alpha-cells and their subsequent conversion into beta-like cells, such converted cells being capable of reversing the consequences of chemically-induced diabetes in vivo (Collombat et al. Cell, 2009).
The PI and his team therefore propose to further decipher the mechanisms involved in this alpha-cell-mediated beta-cell regeneration process and determine whether this approach may be applied to adult animals and whether it would efficiently reverse Type 1 diabetes. Furthermore, a major effort will be made to verify whether our findings could be translated to human. Specifically, we will use a tri-partite approach to address the following issues: (1) Can the in vivo alpha-cell-mediated beta-cell regeneration be induced in adults mice? What would be the genetic determinants involved? (2) Can alpha-cell-mediated beta-cell regeneration reverse diabetes in the NOD Type 1 diabetes mouse model? (3) Can adult human alpha-cells be converted into beta-like cells?
Together, these ambitious objectives will most certainly allow us to gain new insight into the mechanisms defining the identity and the reprogramming capabilities of mouse and human endocrine cells and may thereby open new avenues for the treatment of diabetes. Similarly, the determination of the molecular triggers implicated in the beta-cell regeneration observed in our diabetic mice may lead to exciting new findings, including the identification of “drugable” targets of importance for human diabetic patients.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym BinD
Project Mitotic Bookmarking, Stem Cells and early Development
Researcher (PI) Pablo Navarro Gil
Host Institution (HI) INSTITUT PASTEUR
Call Details Consolidator Grant (CoG), LS3, ERC-2017-COG
Summary The goal of this proposal is to deliver a new theoretical framework to understand how transcription factors (TFs) sustain cell identity during developmental processes. Recognised as key drivers of cell fate acquisition, TFs are currently not considered to directly contribute to the mitotic inheritance of chromatin states. Instead, these are passively propagated through cell division by a variety of epigenetic marks. Recent discoveries, including by our lab, challenge this view: developmental TFs may impact the propagation of regulatory information from mother to daughter cells through a process known as mitotic bookmarking. This hypothesis, largely overlooked by mainstream epigenetic research during the last two decades, will be investigated in embryo-derived stem cells and during early mouse development. Indeed, these immature cell identities are largely independent from canonical epigenetic repression; hence, current models cannot account for their properties. We will comprehensively identify mitotic bookmarking factors in stem cells and early embryos, establish their function in stem cell self-renewal, cell fate acquisition and dissect how they contribute to chromatin regulation in mitosis. This will allow us to study the relationships between bookmarking factors and other mechanisms of epigenetic inheritance. To achieve this, unique techniques to modulate protein activity and histone modifications specifically in mitotic cells will be established. Thus, a mechanistic understanding of how mitosis influences gene regulation and of how mitotic bookmarking contributes to the propagation of immature cell identities will be delivered. Based on robust preliminary data, we anticipate the discovery of new functions for TFs in several genetic and epigenetic processes. This knowledge should have a wide impact on chromatin biology and cell fate studies as well as in other fields studying processes dominated by TFs and cell proliferation.
Summary
The goal of this proposal is to deliver a new theoretical framework to understand how transcription factors (TFs) sustain cell identity during developmental processes. Recognised as key drivers of cell fate acquisition, TFs are currently not considered to directly contribute to the mitotic inheritance of chromatin states. Instead, these are passively propagated through cell division by a variety of epigenetic marks. Recent discoveries, including by our lab, challenge this view: developmental TFs may impact the propagation of regulatory information from mother to daughter cells through a process known as mitotic bookmarking. This hypothesis, largely overlooked by mainstream epigenetic research during the last two decades, will be investigated in embryo-derived stem cells and during early mouse development. Indeed, these immature cell identities are largely independent from canonical epigenetic repression; hence, current models cannot account for their properties. We will comprehensively identify mitotic bookmarking factors in stem cells and early embryos, establish their function in stem cell self-renewal, cell fate acquisition and dissect how they contribute to chromatin regulation in mitosis. This will allow us to study the relationships between bookmarking factors and other mechanisms of epigenetic inheritance. To achieve this, unique techniques to modulate protein activity and histone modifications specifically in mitotic cells will be established. Thus, a mechanistic understanding of how mitosis influences gene regulation and of how mitotic bookmarking contributes to the propagation of immature cell identities will be delivered. Based on robust preliminary data, we anticipate the discovery of new functions for TFs in several genetic and epigenetic processes. This knowledge should have a wide impact on chromatin biology and cell fate studies as well as in other fields studying processes dominated by TFs and cell proliferation.
Max ERC Funding
1 900 844 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym BioMatrix
Project Structural Biology of Exopolysaccharide Secretion in Bacterial Biofilms
Researcher (PI) Petya Violinova KRASTEVA
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), LS1, ERC-2017-STG
Summary Bacterial biofilm formation is a paramount developmental process in both Gram-positive and Gram-negative species and in many pathogens has been associated with processes of horizontal gene transfer, antibiotic resistance development and pathogen persistence. Bacterial biofilms are collaborative sessile macrocolonies embedded in complex extracellular matrix that secures both mechanical resistance and a medium for intercellular exchange.
Biogenesis platforms for the secretion of biofilm matrix components - many of which controlled directly or indirectly by the intracellular second messenger c-di-GMP - are important determinants for biofilm formation and bacterial disease, and therefore present compelling targets for the development of novel therapeutics. During my Ph.D. and post-doctoral work I studied the structure and function of c-di-GMP-sensing protein factors controling extracellular matrix production by DNA-binding at the transcription initiation level or by inside-out signalling mechanisms at the cell envelope, as well as membrane exporters involved directly in downstream matrix component secretion.
Here, I propose to apply my expertise in microbiology, protein science and structural biology to study the structure and function of exopolysaccharide secretion systems in Gram-negative species. Using Pseudomonas aeruginosa, Vibrio spp. and Escherichia coli as model organisms, my team will aim to reveal the global architecture and individual building components of several expolysaccharide-producing protein megacomplexes. We will combine X-ray crystallography, biophysical and biochemical assays, electron microscopy and in cellulo functional studies to provide a comprehensive view of extracellular matrix production that spans the different resolution levels and presents molecular blueprints for the development of novel anti-infectives. Over the last year I have laid the foundation of these studies and have demonstrated the overall feasibility of the project.
Summary
Bacterial biofilm formation is a paramount developmental process in both Gram-positive and Gram-negative species and in many pathogens has been associated with processes of horizontal gene transfer, antibiotic resistance development and pathogen persistence. Bacterial biofilms are collaborative sessile macrocolonies embedded in complex extracellular matrix that secures both mechanical resistance and a medium for intercellular exchange.
Biogenesis platforms for the secretion of biofilm matrix components - many of which controlled directly or indirectly by the intracellular second messenger c-di-GMP - are important determinants for biofilm formation and bacterial disease, and therefore present compelling targets for the development of novel therapeutics. During my Ph.D. and post-doctoral work I studied the structure and function of c-di-GMP-sensing protein factors controling extracellular matrix production by DNA-binding at the transcription initiation level or by inside-out signalling mechanisms at the cell envelope, as well as membrane exporters involved directly in downstream matrix component secretion.
Here, I propose to apply my expertise in microbiology, protein science and structural biology to study the structure and function of exopolysaccharide secretion systems in Gram-negative species. Using Pseudomonas aeruginosa, Vibrio spp. and Escherichia coli as model organisms, my team will aim to reveal the global architecture and individual building components of several expolysaccharide-producing protein megacomplexes. We will combine X-ray crystallography, biophysical and biochemical assays, electron microscopy and in cellulo functional studies to provide a comprehensive view of extracellular matrix production that spans the different resolution levels and presents molecular blueprints for the development of novel anti-infectives. Over the last year I have laid the foundation of these studies and have demonstrated the overall feasibility of the project.
Max ERC Funding
1 499 901 €
Duration
Start date: 2018-08-01, End date: 2023-07-31
Project acronym BIOMECAMORPH
Project The Biomechanics of Epithelial Cell and Tissue Morphogenesis
Researcher (PI) Thomas Marie Michel Lecuit
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS3, ERC-2012-ADG_20120314
Summary Tissue morphogenesis is a complex process that emerges from spatially controlled patterns of cell shape changes. Dedicated genetic programmes regulate cell behaviours, exemplified in animals by the specification of apical constriction in invaginating epithelial tissues, or the orientation of cell intercalation during tissue extension. This genetic control is constrained by physical properties of cells that dictate how they can modify their shape. A major challenge is to understand how biochemical pathways control subcellular mechanics in epithelia, such as how forces are produced by interactions between actin filaments and myosin motors, and how these forces are transmitted at cell junctions. The major objective of our project is to investigate the fundamental principles of epithelial mechanics and to understand how intercellular signals and mechanical coupling between cells coordinate individual behaviours at the tissue level.
We will study early Drosophila embryogenesis and combine quantitative cell biological studies of cell dynamics, biophysical characterization of cell mechanics and genetic control of cell signalling to answer the following questions: i) how are forces generated, in particular what underlies deformation and stabilization of cell shape by actomyosin networks, and pulsatile contractility; ii) how are forces transmitted at junctions, what are the feedback interactions between tension generation and transmission; iii) how are individual cell mechanics orchestrated at the tissue level to yield collective tissue morphogenesis?
We expect to encapsulate the information-based, cell biological and physical descriptions of morphogenesis in a single, coherent framework. The project should impact more broadly on morphogenesis in other organisms and shed light on the mechanisms underlying robustness and plasticity in epithelia.
Summary
Tissue morphogenesis is a complex process that emerges from spatially controlled patterns of cell shape changes. Dedicated genetic programmes regulate cell behaviours, exemplified in animals by the specification of apical constriction in invaginating epithelial tissues, or the orientation of cell intercalation during tissue extension. This genetic control is constrained by physical properties of cells that dictate how they can modify their shape. A major challenge is to understand how biochemical pathways control subcellular mechanics in epithelia, such as how forces are produced by interactions between actin filaments and myosin motors, and how these forces are transmitted at cell junctions. The major objective of our project is to investigate the fundamental principles of epithelial mechanics and to understand how intercellular signals and mechanical coupling between cells coordinate individual behaviours at the tissue level.
We will study early Drosophila embryogenesis and combine quantitative cell biological studies of cell dynamics, biophysical characterization of cell mechanics and genetic control of cell signalling to answer the following questions: i) how are forces generated, in particular what underlies deformation and stabilization of cell shape by actomyosin networks, and pulsatile contractility; ii) how are forces transmitted at junctions, what are the feedback interactions between tension generation and transmission; iii) how are individual cell mechanics orchestrated at the tissue level to yield collective tissue morphogenesis?
We expect to encapsulate the information-based, cell biological and physical descriptions of morphogenesis in a single, coherent framework. The project should impact more broadly on morphogenesis in other organisms and shed light on the mechanisms underlying robustness and plasticity in epithelia.
Max ERC Funding
2 473 313 €
Duration
Start date: 2013-05-01, End date: 2018-04-30
Project acronym BIOSTASES
Project BIOdiversity, STAbility and sustainability in Spatial Ecological and social-ecological Systems
Researcher (PI) Michel Loreau
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS8, ERC-2014-ADG
Summary Biodiversity loss is one of the greatest environmental challenges of our time. There is mounting evidence that biodiversity increases the stability of ecosystem functions and services, suggesting that it may be critical to the sustainability of ecosystems and human societies in the face of environmental changes. Classical ecological theory, however, has focused on measures of stability that cannot explain and predict these stabilizing effects, especially in spatial systems.
The goal of BIOSTASES is to develop a coherent body of new theory on the stability of ecosystems and coupled social–ecological systems and its relationships with biodiversity at multiple spatial scales that can better inform empirical research. BIOSTASES will reach this goal through four complementary objectives. First, it will propose a mathematical framework focused on temporal variability as an empirically relevant measure of stability, and use this framework to build robust early warning signals for critical transitions. Second, it will use dynamical metacommunity models to explore a wide range of novel questions related to ecosystem stability and diversity–stability relationships across scales. Third, it will study the stability of complex meta-ecosystems to provide new perspectives on the stability of food webs and on synergies and trade-offs between multiple ecosystem services across space. Fourth, it will develop novel theory to study the long-term dynamics and sustainability of coupled social–ecological systems.
BIOSTASES proposes an ambitious innovative research programme that will provide new perspectives on the stability and sustainability of ecological and coupled social–ecological systems in the face of environmental changes. It will contribute to bridging the gaps between theoretical and empirical ecology and between ecology and social sciences, and to developing new approaches in biodiversity conservation, landscape management, and sustainable development.
Summary
Biodiversity loss is one of the greatest environmental challenges of our time. There is mounting evidence that biodiversity increases the stability of ecosystem functions and services, suggesting that it may be critical to the sustainability of ecosystems and human societies in the face of environmental changes. Classical ecological theory, however, has focused on measures of stability that cannot explain and predict these stabilizing effects, especially in spatial systems.
The goal of BIOSTASES is to develop a coherent body of new theory on the stability of ecosystems and coupled social–ecological systems and its relationships with biodiversity at multiple spatial scales that can better inform empirical research. BIOSTASES will reach this goal through four complementary objectives. First, it will propose a mathematical framework focused on temporal variability as an empirically relevant measure of stability, and use this framework to build robust early warning signals for critical transitions. Second, it will use dynamical metacommunity models to explore a wide range of novel questions related to ecosystem stability and diversity–stability relationships across scales. Third, it will study the stability of complex meta-ecosystems to provide new perspectives on the stability of food webs and on synergies and trade-offs between multiple ecosystem services across space. Fourth, it will develop novel theory to study the long-term dynamics and sustainability of coupled social–ecological systems.
BIOSTASES proposes an ambitious innovative research programme that will provide new perspectives on the stability and sustainability of ecological and coupled social–ecological systems in the face of environmental changes. It will contribute to bridging the gaps between theoretical and empirical ecology and between ecology and social sciences, and to developing new approaches in biodiversity conservation, landscape management, and sustainable development.
Max ERC Funding
2 092 644 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym BIRTOACTION
Project From birth to action: regulation of gene expression through transcription complex biogenesis
Researcher (PI) Laszlo Tora
Host Institution (HI) CENTRE EUROPEEN DE RECHERCHE EN BIOLOGIE ET MEDECINE
Call Details Advanced Grant (AdG), LS1, ERC-2013-ADG
Summary "Transcriptional regulation of protein coding genes in eukaryotic cells requires a complex interplay of sequence-specific DNA-binding factors, co-activators, general transcription factors (GTFs), RNA polymerase II and the epigenetic status of target sequences. Nuclear transcription complexes function as large multiprotein assemblies and are often composed of functional modules. The regulated decision-making that exists in cells governing the assembly and the allocation of factors to different transcription complexes to regulate distinct gene expression pathways is not yet understood. To tackle this fundamental question, we will systematically analyse the regulated biogenesis of transcription complexes from their sites of translation in the cytoplasm, through their assembly intermediates and nuclear import, to their site of action in the nucleus. The project will have four main Aims to decipher the biogenesis of transcription complexes:
I) Investigate their co-translation-driven assembly
II) Determine their cytoplasmic intermediates and factors required for their assembly pathways
III) Uncover their nuclear import
IV) Understand at the single molecule level their nuclear assembly, dynamics and action at target genes
To carry out these aims we propose a combination of multidisciplinary and cutting edge approaches, out of which some of them will be high-risk taking, while others will utilize methods routinely run by the group. The project builds on several complementary expertise and knowledge either already existing in the group or that will be implemented during the project. At the end of the proposed project we will obtain novel results extensively describing the different steps of the regulatory mechanisms that control the assembly and the consequent gene regulatory function of transcription complexes. Thus, we anticipate that the results of our research will have a major impact on the field and will lead to a new paradigm for contemporary metazoan transcription."
Summary
"Transcriptional regulation of protein coding genes in eukaryotic cells requires a complex interplay of sequence-specific DNA-binding factors, co-activators, general transcription factors (GTFs), RNA polymerase II and the epigenetic status of target sequences. Nuclear transcription complexes function as large multiprotein assemblies and are often composed of functional modules. The regulated decision-making that exists in cells governing the assembly and the allocation of factors to different transcription complexes to regulate distinct gene expression pathways is not yet understood. To tackle this fundamental question, we will systematically analyse the regulated biogenesis of transcription complexes from their sites of translation in the cytoplasm, through their assembly intermediates and nuclear import, to their site of action in the nucleus. The project will have four main Aims to decipher the biogenesis of transcription complexes:
I) Investigate their co-translation-driven assembly
II) Determine their cytoplasmic intermediates and factors required for their assembly pathways
III) Uncover their nuclear import
IV) Understand at the single molecule level their nuclear assembly, dynamics and action at target genes
To carry out these aims we propose a combination of multidisciplinary and cutting edge approaches, out of which some of them will be high-risk taking, while others will utilize methods routinely run by the group. The project builds on several complementary expertise and knowledge either already existing in the group or that will be implemented during the project. At the end of the proposed project we will obtain novel results extensively describing the different steps of the regulatory mechanisms that control the assembly and the consequent gene regulatory function of transcription complexes. Thus, we anticipate that the results of our research will have a major impact on the field and will lead to a new paradigm for contemporary metazoan transcription."
Max ERC Funding
2 500 000 €
Duration
Start date: 2014-01-01, End date: 2018-12-31
Project acronym BODYBUILT
Project Building The Vertebrate Body
Researcher (PI) Olivier Pourquie
Host Institution (HI) CENTRE EUROPEEN DE RECHERCHE EN BIOLOGIE ET MEDECINE
Call Details Advanced Grant (AdG), LS3, ERC-2009-AdG
Summary My lab is interested in the development of the tissue that gives rise to vertebrae and skeletal muscles called the paraxial mesoderm. A striking feature of this tissue is its segmental organization and we have made major contributions to the understanding of the molecular control of the segmentation process. We identified a molecular oscillator associated to the rhythmic production of somites and proposed a model for vertebrate segmentation based on the integration of a rhythmic signaling pulse gated spatially by a system of traveling FGF and Wnt signaling gradients. We are also studying the differentiation of paraxial mesoderm precursors into the muscle, cartilage and dermis lineages. Our work identified the Wnt, FGF and Notch pathways as playing a prominent role in the patterning and differentiation of paraxial mesoderm. In this application, we largely focus on the molecular control of paraxial mesoderm development. Using microarray and high throughput sequencing-based approaches and bioinformatics, we will characterize the transcriptional network acting downstream of Wnt, FGF and Notch in the presomitic mesoderm (PSM). We will also use genetic and pharmacological approaches utilizing real-time imaging reporters to characterize the pacemaker of the segmentation clock in vivo, and also in vitro using differentiated embryonic stem cells. We further propose to characterize in detail a novel RA-dependent pathway that we identified and which controls the somite left-right symmetry. Our work is expected to have a strong impact in the field of congenital spine anomalies, currently an understudied biomedical problem, and will be of utility in elucidating the etiology and eventual prevention of these disorders. This work is also expected to further our understanding of the Notch, Wnt, FGF and RA signalling pathways which are involved in segmentation and in the establishment of the vertebrate body plan, and which play important roles in a wide array of human diseases.
Summary
My lab is interested in the development of the tissue that gives rise to vertebrae and skeletal muscles called the paraxial mesoderm. A striking feature of this tissue is its segmental organization and we have made major contributions to the understanding of the molecular control of the segmentation process. We identified a molecular oscillator associated to the rhythmic production of somites and proposed a model for vertebrate segmentation based on the integration of a rhythmic signaling pulse gated spatially by a system of traveling FGF and Wnt signaling gradients. We are also studying the differentiation of paraxial mesoderm precursors into the muscle, cartilage and dermis lineages. Our work identified the Wnt, FGF and Notch pathways as playing a prominent role in the patterning and differentiation of paraxial mesoderm. In this application, we largely focus on the molecular control of paraxial mesoderm development. Using microarray and high throughput sequencing-based approaches and bioinformatics, we will characterize the transcriptional network acting downstream of Wnt, FGF and Notch in the presomitic mesoderm (PSM). We will also use genetic and pharmacological approaches utilizing real-time imaging reporters to characterize the pacemaker of the segmentation clock in vivo, and also in vitro using differentiated embryonic stem cells. We further propose to characterize in detail a novel RA-dependent pathway that we identified and which controls the somite left-right symmetry. Our work is expected to have a strong impact in the field of congenital spine anomalies, currently an understudied biomedical problem, and will be of utility in elucidating the etiology and eventual prevention of these disorders. This work is also expected to further our understanding of the Notch, Wnt, FGF and RA signalling pathways which are involved in segmentation and in the establishment of the vertebrate body plan, and which play important roles in a wide array of human diseases.
Max ERC Funding
2 500 000 €
Duration
Start date: 2010-04-01, End date: 2015-03-31
Project acronym Brain3.0
Project Invasive cognitive brain computer interfaces to enhance and restore attention: proof of concept and underlying cortical mechanisms.
Researcher (PI) Suliann Benhamed-Daghighi-Ardekani
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS5, ERC-2015-CoG
Summary The present project focuses on a barely scratched aspect of invasive cognitive brain-computer interfaces (cBCIs), i.e. closed-loop invasive cBCIs to augment and restore attentional functions. Its aim is to achieve an efficient enhanced cognition protocol both in the healthy brain and in the damaged brain and to study the local and global plasticity mechanisms underlying these effects. The project relies on the unique methodological combination of multi-electrode multisite intracortical recordings and functional magnetic resonance imaging, in association with reversible cortical lesions and intracortical microstimulations, in an experimental model allowing to approach the attentional human function and its dysfunctions to the best. Our goal is to achieve:
1. A closed-loop invasive cBCI for augmented attention, by providing the subjects with a feedback on their cortical spatial and feature attention information content as estimated from real-time population decoding procedures, using reinforcement learning, to have them improve this cognitive content, and as a result, improve their overt attentional behavioural performance.
2. A closed-loop invasive cBCI for restored attention, by inducing a controlled attentional loss thanks to reversible cortical lesions targeted to key functionally-identified cortical regions and using the closed-loop cBCI to restore attentional performance.
3. An invasive cBCI for stimulated attentional functions. We will identify the neuronal population changes leading to a voluntary enhancement of attentional functions as quantified in aim 1 and inject these changes, using complex patterns of microstimulations, mimicking spikes, to enhance or restore attention, in the absence of any active control by the subjects.
This project will contribute to the development of novel therapeutical applications to restore acute or chronic severe attentional deficits and to provide an in depth understanding of the neural bases underlying closed-loop cBCIs.
Summary
The present project focuses on a barely scratched aspect of invasive cognitive brain-computer interfaces (cBCIs), i.e. closed-loop invasive cBCIs to augment and restore attentional functions. Its aim is to achieve an efficient enhanced cognition protocol both in the healthy brain and in the damaged brain and to study the local and global plasticity mechanisms underlying these effects. The project relies on the unique methodological combination of multi-electrode multisite intracortical recordings and functional magnetic resonance imaging, in association with reversible cortical lesions and intracortical microstimulations, in an experimental model allowing to approach the attentional human function and its dysfunctions to the best. Our goal is to achieve:
1. A closed-loop invasive cBCI for augmented attention, by providing the subjects with a feedback on their cortical spatial and feature attention information content as estimated from real-time population decoding procedures, using reinforcement learning, to have them improve this cognitive content, and as a result, improve their overt attentional behavioural performance.
2. A closed-loop invasive cBCI for restored attention, by inducing a controlled attentional loss thanks to reversible cortical lesions targeted to key functionally-identified cortical regions and using the closed-loop cBCI to restore attentional performance.
3. An invasive cBCI for stimulated attentional functions. We will identify the neuronal population changes leading to a voluntary enhancement of attentional functions as quantified in aim 1 and inject these changes, using complex patterns of microstimulations, mimicking spikes, to enhance or restore attention, in the absence of any active control by the subjects.
This project will contribute to the development of novel therapeutical applications to restore acute or chronic severe attentional deficits and to provide an in depth understanding of the neural bases underlying closed-loop cBCIs.
Max ERC Funding
1 997 748 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym BRAINCANNABINOIDS
Project Understanding the molecular blueprint and functional complexity of the endocannabinoid metabolome in the brain
Researcher (PI) István Katona
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Starting Grant (StG), LS5, ERC-2009-StG
Summary We and others have recently delineated the molecular architecture of a new feedback pathway in brain synapses, which operates as a synaptic circuit breaker. This pathway is supposed to use a group of lipid messengers as retrograde synaptic signals, the so-called endocannabinoids. Although heterogeneous in their chemical structures, these molecules along with the psychoactive compound in cannabis are thought to target the same effector in the brain, the CB1 receptor. However, the molecular catalog of these bioactive lipids and their metabolic enzymes has been expanding rapidly by recent advances in lipidomics and proteomics raising the possibility that these lipids may also serve novel, yet unidentified physiological functions. Thus, the overall aim of our research program is to define the molecular and anatomical organization of these endocannabinoid-mediated pathways and to determine their functional significance. In the present proposal, we will focus on understanding how these novel pathways regulate synaptic and extrasynaptic signaling in hippocampal neurons. Using combination of lipidomic, genetic and high-resolution anatomical approaches, we will identify distinct chemical species of endocannabinoids and will show how their metabolic enzymes are segregated into different subcellular compartments in cell type- and synapse-specific manner. Subsequently, we will use genetically encoded gain-of-function, loss-of-function and reporter constructs in imaging experiments and electrophysiological recordings to gain insights into the diverse tasks that these new pathways serve in synaptic transmission and extrasynaptic signal processing. Our proposed experiments will reveal fundamental principles of intercellular and intracellular endocannabinoid signaling in the brain.
Summary
We and others have recently delineated the molecular architecture of a new feedback pathway in brain synapses, which operates as a synaptic circuit breaker. This pathway is supposed to use a group of lipid messengers as retrograde synaptic signals, the so-called endocannabinoids. Although heterogeneous in their chemical structures, these molecules along with the psychoactive compound in cannabis are thought to target the same effector in the brain, the CB1 receptor. However, the molecular catalog of these bioactive lipids and their metabolic enzymes has been expanding rapidly by recent advances in lipidomics and proteomics raising the possibility that these lipids may also serve novel, yet unidentified physiological functions. Thus, the overall aim of our research program is to define the molecular and anatomical organization of these endocannabinoid-mediated pathways and to determine their functional significance. In the present proposal, we will focus on understanding how these novel pathways regulate synaptic and extrasynaptic signaling in hippocampal neurons. Using combination of lipidomic, genetic and high-resolution anatomical approaches, we will identify distinct chemical species of endocannabinoids and will show how their metabolic enzymes are segregated into different subcellular compartments in cell type- and synapse-specific manner. Subsequently, we will use genetically encoded gain-of-function, loss-of-function and reporter constructs in imaging experiments and electrophysiological recordings to gain insights into the diverse tasks that these new pathways serve in synaptic transmission and extrasynaptic signal processing. Our proposed experiments will reveal fundamental principles of intercellular and intracellular endocannabinoid signaling in the brain.
Max ERC Funding
1 638 000 €
Duration
Start date: 2009-11-01, End date: 2014-10-31
Project acronym BrainDyn
Project Tracking information flow in the brain: A unified and general framework for dynamic communication in brain networks
Researcher (PI) Mathilde BONNEFOND
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS5, ERC-2016-STG
Summary The brain is composed of a set of areas specialized in specific computations whose outputs need to be transferred to other specialized areas for cognition to emerge. To account for context-dependent behaviors, the information has to be flexibly routed through the fixed anatomy of the brain. The aim of my proposal is to test a general framework for flexible communication between brain areas based on nested oscillations which I recently developed. The general idea is that internally-driven slow oscillations (<20Hz) either set-up or prevent the communication between brain areas. Stimulus-driven gamma oscillations (>30Hz), nested in the slow oscillations, can then be directed to task-relevant areas of the network. I plan to use a multimodal, multi-scale and transversal (human and monkey) approach in experiments manipulating visual processing, attention and memory to test core predictions of my framework. The theoretical approach and the methodological development used in my project will provide the basis for future fundamental and clinical research.
Summary
The brain is composed of a set of areas specialized in specific computations whose outputs need to be transferred to other specialized areas for cognition to emerge. To account for context-dependent behaviors, the information has to be flexibly routed through the fixed anatomy of the brain. The aim of my proposal is to test a general framework for flexible communication between brain areas based on nested oscillations which I recently developed. The general idea is that internally-driven slow oscillations (<20Hz) either set-up or prevent the communication between brain areas. Stimulus-driven gamma oscillations (>30Hz), nested in the slow oscillations, can then be directed to task-relevant areas of the network. I plan to use a multimodal, multi-scale and transversal (human and monkey) approach in experiments manipulating visual processing, attention and memory to test core predictions of my framework. The theoretical approach and the methodological development used in my project will provide the basis for future fundamental and clinical research.
Max ERC Funding
1 333 718 €
Duration
Start date: 2017-02-01, End date: 2022-01-31
Project acronym BRAINSTRUCT
Project Building up a brain: understanding how neural stem cell fate and regulation controls nervous tissue architecture
Researcher (PI) Jean Livet
Host Institution (HI) SORBONNE UNIVERSITE
Call Details Consolidator Grant (CoG), LS5, ERC-2014-CoG
Summary The brain is an extraordinary complex assembly of neuronal and glial cells that underpins cognitive functions. How adequate numbers of these cells are generated by neural stem cells in embryonic and early postnatal development and how they distribute and interconnect within brain tissue is still debated. In particular, the potentialities of individual neural stem cells, their potential heterogeneity and the mechanisms regulating their function are still poorly characterized in situ; likewise, the clonal architecture of mature brain tissue and its influence on neural circuitry are only partially explored. Deciphering these aspects is essential to link neural circuit development, structure and function, and to understand the aetiology of neurodevelopmental disorders.
We have recently established transgenic strategies to simultaneously track the lineage of multiple individual neural stem cells in the intact developing brain and experimentally perturb their development. We will use these approaches in combination with recent large-volume imaging methods for high-throughput analysis of individual neural and glial clones in the mouse cortex. This will allow us to assay neural progenitor potentialities and equivalence, characterize developmental changes occurring in the neurogenic niche, describe the clonal organization of the mature cortex and study its link with neural connectivity. To decipher intrinsic and extrinsic mechanisms regulating neural progenitor activity and understand how they produce appropriate numbers of cells, we will assay the outcome of functional perturbations targeting key steps of neural development, introduced in precursors or in their local environment. These experiments will reveal how neural stem cell output might be regulated by cell interactions and intercellular signals. This multidisciplinary project will set the basis for quantitative analysis of brain development with single-cell resolution in normal and pathological conditions.
Summary
The brain is an extraordinary complex assembly of neuronal and glial cells that underpins cognitive functions. How adequate numbers of these cells are generated by neural stem cells in embryonic and early postnatal development and how they distribute and interconnect within brain tissue is still debated. In particular, the potentialities of individual neural stem cells, their potential heterogeneity and the mechanisms regulating their function are still poorly characterized in situ; likewise, the clonal architecture of mature brain tissue and its influence on neural circuitry are only partially explored. Deciphering these aspects is essential to link neural circuit development, structure and function, and to understand the aetiology of neurodevelopmental disorders.
We have recently established transgenic strategies to simultaneously track the lineage of multiple individual neural stem cells in the intact developing brain and experimentally perturb their development. We will use these approaches in combination with recent large-volume imaging methods for high-throughput analysis of individual neural and glial clones in the mouse cortex. This will allow us to assay neural progenitor potentialities and equivalence, characterize developmental changes occurring in the neurogenic niche, describe the clonal organization of the mature cortex and study its link with neural connectivity. To decipher intrinsic and extrinsic mechanisms regulating neural progenitor activity and understand how they produce appropriate numbers of cells, we will assay the outcome of functional perturbations targeting key steps of neural development, introduced in precursors or in their local environment. These experiments will reveal how neural stem cell output might be regulated by cell interactions and intercellular signals. This multidisciplinary project will set the basis for quantitative analysis of brain development with single-cell resolution in normal and pathological conditions.
Max ERC Funding
1 929 713 €
Duration
Start date: 2015-07-01, End date: 2020-06-30
Project acronym BreakingBarriers
Project Targeting endothelial barriers to combat disease
Researcher (PI) Anne Eichmann
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS4, ERC-2018-ADG
Summary Tissue homeostasis requires coordinated barrier function in blood and lymphatic vessels. Opening of junctions between endothelial cells (ECs) lining blood vessels leads to tissue fluid accumulation that is drained by lymphatic vessels. A pathological increase in blood vessel permeability or lack or malfunction of lymphatic vessels leads to edema and associated defects in macromolecule and immune cell clearance. Unbalanced barrier function between blood and lymphatic vessels contributes to neurodegeneration, chronic inflammation, and cardiovascular disease. In this proposal, we seek to gain mechanistic understanding into coordination of barrier function between blood and lymphatic vessels, how this process is altered in disease models and how it can be manipulated for therapeutic purposes. We will focus on two critical barriers with diametrically opposing functions, the blood-brain barrier (BBB) and the lymphatic capillary barrier (LCB). ECs of the BBB form very tight junctions that restrict paracellular access to the brain. In contrast, open junctions of the LCB ensure uptake of extravasated fluid, macromolecules and immune cells, as well as lipid in the gut. We have identified novel effectors of BBB and LCB junctions and will determine their role in adult homeostasis and in disease models. Mouse genetic gain and loss of function approaches in combination with histological, ultrastructural, functional and molecular analysis will determine mechanisms underlying formation of tissue specific EC barriers. Deliverables include in vivo validated targets that could be used for i) opening the BBB on demand for drug delivery into the brain, and ii) to lower plasma lipid uptake via interfering with the LCB, with implications for prevention of obesity, cardiovascular disease and inflammation. These pioneering studies promise to open up new opportunities for research and treatment of neurovascular and cardiovascular disease.
Summary
Tissue homeostasis requires coordinated barrier function in blood and lymphatic vessels. Opening of junctions between endothelial cells (ECs) lining blood vessels leads to tissue fluid accumulation that is drained by lymphatic vessels. A pathological increase in blood vessel permeability or lack or malfunction of lymphatic vessels leads to edema and associated defects in macromolecule and immune cell clearance. Unbalanced barrier function between blood and lymphatic vessels contributes to neurodegeneration, chronic inflammation, and cardiovascular disease. In this proposal, we seek to gain mechanistic understanding into coordination of barrier function between blood and lymphatic vessels, how this process is altered in disease models and how it can be manipulated for therapeutic purposes. We will focus on two critical barriers with diametrically opposing functions, the blood-brain barrier (BBB) and the lymphatic capillary barrier (LCB). ECs of the BBB form very tight junctions that restrict paracellular access to the brain. In contrast, open junctions of the LCB ensure uptake of extravasated fluid, macromolecules and immune cells, as well as lipid in the gut. We have identified novel effectors of BBB and LCB junctions and will determine their role in adult homeostasis and in disease models. Mouse genetic gain and loss of function approaches in combination with histological, ultrastructural, functional and molecular analysis will determine mechanisms underlying formation of tissue specific EC barriers. Deliverables include in vivo validated targets that could be used for i) opening the BBB on demand for drug delivery into the brain, and ii) to lower plasma lipid uptake via interfering with the LCB, with implications for prevention of obesity, cardiovascular disease and inflammation. These pioneering studies promise to open up new opportunities for research and treatment of neurovascular and cardiovascular disease.
Max ERC Funding
2 499 969 €
Duration
Start date: 2019-07-01, End date: 2024-06-30
Project acronym BRIDGING
Project The function of membrane tethering in plant intercellular communication
Researcher (PI) Emmanuelle Maria Françoise Bayer
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS3, ERC-2017-COG
Summary Intercellular communication is critical for multicellularity. It coordinates the activities within individual cells to support the function of an organism as a whole. Plants have developed remarkable cellular machines -the Plasmodesmata (PD) pores- which interconnect every single cell within the plant body, establishing direct membrane and cytoplasmic continuity, a situation unique to plants. PD are indispensable for plant life. They control the flux of molecules between cells and are decisive for development, environmental adaptation and defence signalling. However, how PD integrate signalling to coordinate responses at a multicellular level remains unclear.
A striking feature of PD organisation, setting them apart from animal cell junctions, is a strand of endoplasmic reticulum (ER) running through the pore, tethered extremely tight (~10nm) to the plasma membrane (PM) by unidentified “spokes”. To date, the function of ER-PM contacts at PD remains a complete enigma. We don’t know how and why the two organelles come together at PD cellular junctions.
I recently proposed that ER-PM tethering is in fact central to PD function. In this project I will investigate the question of how integrated cellular responses benefit from organelle cross-talk at PD. The project integrates proteomic/bioinformatic approaches, biophysical/modelling methods and ultra-high resolution 3D imaging into molecular cell biology of plant cell-to-cell communication and will, for the first time, directly address the mechanism and function of ER-PM contacts at PD. We will pursue three complementary objectives to attain our goal: 1) Identify the mechanisms of PD membrane-tethering at the molecular level 2) Elucidate the dynamics and 3D architecture of ER-PM contact sites at PD 3) Uncover the function of ER-PM apposition for plant intercellular communication. Overall, the project will pioneer a radically new perspective on PD-mediated cell-to-cell communication, a fundamental aspect of plant biology
Summary
Intercellular communication is critical for multicellularity. It coordinates the activities within individual cells to support the function of an organism as a whole. Plants have developed remarkable cellular machines -the Plasmodesmata (PD) pores- which interconnect every single cell within the plant body, establishing direct membrane and cytoplasmic continuity, a situation unique to plants. PD are indispensable for plant life. They control the flux of molecules between cells and are decisive for development, environmental adaptation and defence signalling. However, how PD integrate signalling to coordinate responses at a multicellular level remains unclear.
A striking feature of PD organisation, setting them apart from animal cell junctions, is a strand of endoplasmic reticulum (ER) running through the pore, tethered extremely tight (~10nm) to the plasma membrane (PM) by unidentified “spokes”. To date, the function of ER-PM contacts at PD remains a complete enigma. We don’t know how and why the two organelles come together at PD cellular junctions.
I recently proposed that ER-PM tethering is in fact central to PD function. In this project I will investigate the question of how integrated cellular responses benefit from organelle cross-talk at PD. The project integrates proteomic/bioinformatic approaches, biophysical/modelling methods and ultra-high resolution 3D imaging into molecular cell biology of plant cell-to-cell communication and will, for the first time, directly address the mechanism and function of ER-PM contacts at PD. We will pursue three complementary objectives to attain our goal: 1) Identify the mechanisms of PD membrane-tethering at the molecular level 2) Elucidate the dynamics and 3D architecture of ER-PM contact sites at PD 3) Uncover the function of ER-PM apposition for plant intercellular communication. Overall, the project will pioneer a radically new perspective on PD-mediated cell-to-cell communication, a fundamental aspect of plant biology
Max ERC Funding
1 999 840 €
Duration
Start date: 2018-06-01, End date: 2023-05-31
Project acronym BUNDLEFORCE
Project Unravelling the Mechanosensitivity of Actin Bundles in Filopodia
Researcher (PI) Antoine Guillaume Jegou
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), LS1, ERC-2015-STG
Summary Eukaryotic cells constantly convert signals between biochemical energy and mechanical work to timely accomplish many key functions such as migration, division or development. Filopodia are essential finger-like structures that emerge at the cell front to orient the cell in response to its chemical and mechanical environment. Yet, the molecular interactions that make the filopodia mechanosensitive are not known. To tackle this challenge we propose unique biophysical in vitro and in vivo experiments of increasing complexity. Here we will focus on how the underlying actin filament bundle regulates filopodium growth and retraction cycles at the micrometer and seconds scales. These parallel actin filaments are mainly elongated at their barbed-end by formins and cross-linked by bundling proteins such as fascins.
We aim to:
1) Elucidate how formin and fascin functions are regulated by mechanics at the single filament level. We will investigate how formin partners and competitors present in filopodia affect formin processivity; how fascin affinity for the side of filaments is modified by filament tension and formin presence at the barbed-end.
2) Reconstitute filopodium-like actin bundles in vitro to understand how actin bundle size and fate are regulated down to the molecular scale. Using a unique experimental setup that combines microfluidics and optical tweezers, we will uncover for the first time actin bundles mechanosensitive capabilities, both in tension and compression.
3) Decipher in vivo the mechanics of actin bundles in filopodia, using fascins and formins with integrated fluorescent tension sensors.
This framework spanning from in vitro single filament to in vivo meso-scale actin networks will bring unprecedented insights into the role of actin bundles in filopodia mechanosensitivity.
Summary
Eukaryotic cells constantly convert signals between biochemical energy and mechanical work to timely accomplish many key functions such as migration, division or development. Filopodia are essential finger-like structures that emerge at the cell front to orient the cell in response to its chemical and mechanical environment. Yet, the molecular interactions that make the filopodia mechanosensitive are not known. To tackle this challenge we propose unique biophysical in vitro and in vivo experiments of increasing complexity. Here we will focus on how the underlying actin filament bundle regulates filopodium growth and retraction cycles at the micrometer and seconds scales. These parallel actin filaments are mainly elongated at their barbed-end by formins and cross-linked by bundling proteins such as fascins.
We aim to:
1) Elucidate how formin and fascin functions are regulated by mechanics at the single filament level. We will investigate how formin partners and competitors present in filopodia affect formin processivity; how fascin affinity for the side of filaments is modified by filament tension and formin presence at the barbed-end.
2) Reconstitute filopodium-like actin bundles in vitro to understand how actin bundle size and fate are regulated down to the molecular scale. Using a unique experimental setup that combines microfluidics and optical tweezers, we will uncover for the first time actin bundles mechanosensitive capabilities, both in tension and compression.
3) Decipher in vivo the mechanics of actin bundles in filopodia, using fascins and formins with integrated fluorescent tension sensors.
This framework spanning from in vitro single filament to in vivo meso-scale actin networks will bring unprecedented insights into the role of actin bundles in filopodia mechanosensitivity.
Max ERC Funding
1 499 190 €
Duration
Start date: 2016-03-01, End date: 2021-02-28
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 CANALOHMICS
Project Biophysical networks underlying the robustness of neuronal excitability
Researcher (PI) Jean-Marc Goaillard
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Consolidator Grant (CoG), LS5, ERC-2013-CoG
Summary The mammalian nervous system is in some respect surprisingly robust to perturbations, as suggested by the virtually complete recovery of brain function after strokes or the pre-clinical asymptomatic phase of Parkinson’s disease. Ultimately though, cognitive and behavioral robustness relies on the ability of single neurons to cope with perturbations, and in particular to maintain a constant and reliable transfer of information.
So far, the main facet of robustness that has been studied at the neuronal level is homeostatic plasticity of electrical activity, which refers to the ability of neurons to stabilize their activity level in response to external perturbations. But neurons are also able to maintain their function when one of the major ion channels underlying their activity is deleted or mutated: the number of ion channel subtypes expressed by most excitable cells by far exceeds the minimal number of components necessary to achieve function, offering great potential for compensation when one of the channel’s function is altered. How ion channels are dynamically co-regulated to maintain the appropriate pattern of activity has yet to be determined.
In the current project, we will develop a systems-level approach to robustness of neuronal activity based on the combination of electrophysiology, microfluidic single-cell qPCR and computational modeling. We propose to i) characterize the electrical phenotype of dopaminergic neurons following different types of perturbations (ion channel KO, chronic pharmacological treatment), ii) measure the quantitatives changes in ion channel transcriptome (40 voltage-dependent ion channels) associated with these perturbations and iii) determine the mathematical relationships between quantitative changes in ion channel expression and electrical phenotype. Although focused on dopaminergic neurons, this project will provide a general framework that could be applied to any type of excitable cell to decipher its code of robustness.
Summary
The mammalian nervous system is in some respect surprisingly robust to perturbations, as suggested by the virtually complete recovery of brain function after strokes or the pre-clinical asymptomatic phase of Parkinson’s disease. Ultimately though, cognitive and behavioral robustness relies on the ability of single neurons to cope with perturbations, and in particular to maintain a constant and reliable transfer of information.
So far, the main facet of robustness that has been studied at the neuronal level is homeostatic plasticity of electrical activity, which refers to the ability of neurons to stabilize their activity level in response to external perturbations. But neurons are also able to maintain their function when one of the major ion channels underlying their activity is deleted or mutated: the number of ion channel subtypes expressed by most excitable cells by far exceeds the minimal number of components necessary to achieve function, offering great potential for compensation when one of the channel’s function is altered. How ion channels are dynamically co-regulated to maintain the appropriate pattern of activity has yet to be determined.
In the current project, we will develop a systems-level approach to robustness of neuronal activity based on the combination of electrophysiology, microfluidic single-cell qPCR and computational modeling. We propose to i) characterize the electrical phenotype of dopaminergic neurons following different types of perturbations (ion channel KO, chronic pharmacological treatment), ii) measure the quantitatives changes in ion channel transcriptome (40 voltage-dependent ion channels) associated with these perturbations and iii) determine the mathematical relationships between quantitative changes in ion channel expression and electrical phenotype. Although focused on dopaminergic neurons, this project will provide a general framework that could be applied to any type of excitable cell to decipher its code of robustness.
Max ERC Funding
1 972 797 €
Duration
Start date: 2014-05-01, End date: 2019-04-30
Project acronym CD4DNASP
Project Cell intrinsic control of CD4 T cell differentiation by cytosolic DNA sensing pathways
Researcher (PI) Lionel Jerome Apetoh
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS6, ERC-2015-STG
Summary This proposal aims to investigate the role of cytosolic DNA sensing pathways in CD4 T cell differentiation.
Cellular host defense to pathogens relies on the detection of pathogen-associated molecular patterns including deoxyribonucleic acid (DNA), which can be recognized by host myeloid cells through Toll-like receptor (TLR) 9 binding. Recent evidence however suggests that innate immune cells can also perceive cytoplasmic DNA from infectious or autologous origin through cytosolic DNA sensors triggering TLR9-independent signaling. Activation of cytosolic DNA sensor-dependent signaling pathways has been clearly shown to trigger innate immune responses to microbial and host DNA, but the contribution of cytosolic DNA sensors to the differentiation of CD4 T cells, an essential event for shaping adaptive immune responses, has not been documented. This proposal aims to fill this current knowledge gap.
We aim to decipher the molecular series of transcriptional events triggered by DNA in CD4 T cells that ultimately result in altered T cell differentiation. This aim will be addressed by combining in vitro and in vivo approaches such as advanced gene expression analysis of CD4 T cells and use of transgenic and gene-deficient mice. Structure activity relationship and biophysical studies will also be performed to unravel novel immunomodulators able to affect CD4 T cell differentiation.
Summary
This proposal aims to investigate the role of cytosolic DNA sensing pathways in CD4 T cell differentiation.
Cellular host defense to pathogens relies on the detection of pathogen-associated molecular patterns including deoxyribonucleic acid (DNA), which can be recognized by host myeloid cells through Toll-like receptor (TLR) 9 binding. Recent evidence however suggests that innate immune cells can also perceive cytoplasmic DNA from infectious or autologous origin through cytosolic DNA sensors triggering TLR9-independent signaling. Activation of cytosolic DNA sensor-dependent signaling pathways has been clearly shown to trigger innate immune responses to microbial and host DNA, but the contribution of cytosolic DNA sensors to the differentiation of CD4 T cells, an essential event for shaping adaptive immune responses, has not been documented. This proposal aims to fill this current knowledge gap.
We aim to decipher the molecular series of transcriptional events triggered by DNA in CD4 T cells that ultimately result in altered T cell differentiation. This aim will be addressed by combining in vitro and in vivo approaches such as advanced gene expression analysis of CD4 T cells and use of transgenic and gene-deficient mice. Structure activity relationship and biophysical studies will also be performed to unravel novel immunomodulators able to affect CD4 T cell differentiation.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-08-01, End date: 2021-07-31
Project acronym CD8 T CELLS
Project Development and differentiation of CD8 T lymphocytes
Researcher (PI) Benedita Rocha
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS6, ERC-2008-AdG
Summary CD8 T lymphocytes have a fundamental role in ensuring the control of different types of intracellular pathogens including bacteria, parasites and most viruses. This control may fail due to several reasons. The current aggressive anti-cancer therapies (or rarely certain congenital immune deficiencies) induce CD8 depletion. After bone-marrow transplantation, long time periods are required to ensure T cell reconstitution particularly in the adult. This long lag-time is due to the long-time periods required for hematopoietic precursors to generate T lymphocytes and to a thymus insufficiency in the adult. However, even when CD8 T cells are present CD8 immune responses are not always adequate. Certain chronic infections, as HIV, induce CD8 dysfunction and it is yet unclear how to generate efficient CD8 memory responses conferring adequate protection. To address these questions this project aims 1) To find strategies ensuring the rapid reconstitution of the peripheral and the gut CD8 T cell compartments a) by studying the mechanisms involved HSC division and T cell commitment; b) by isolating and characterizing progenitors we previously described that are T cell committed and able of an accelerated CD8 reconstitution c) by developing new strategies that may allow stable thymus transplantation and continuous thymus T cell generation. 2) To determine the mechanics associated to efficient CD8 memory generation a) by evaluating cellular modifications that ensure the efficient division and the remarkable accumulation and survival of CD8 T cells during the adequate immune responses as compared to inefficient responses b) by studying CD8 differentiation into effector and memory cells in both conditions. These studies will use original experiment mouse models we develop in the laboratory, that allow to address each of these aims. Besides state of the art methods, they will also apply unique very advanced approaches we introduced and are the sole laboratory to perform.
Summary
CD8 T lymphocytes have a fundamental role in ensuring the control of different types of intracellular pathogens including bacteria, parasites and most viruses. This control may fail due to several reasons. The current aggressive anti-cancer therapies (or rarely certain congenital immune deficiencies) induce CD8 depletion. After bone-marrow transplantation, long time periods are required to ensure T cell reconstitution particularly in the adult. This long lag-time is due to the long-time periods required for hematopoietic precursors to generate T lymphocytes and to a thymus insufficiency in the adult. However, even when CD8 T cells are present CD8 immune responses are not always adequate. Certain chronic infections, as HIV, induce CD8 dysfunction and it is yet unclear how to generate efficient CD8 memory responses conferring adequate protection. To address these questions this project aims 1) To find strategies ensuring the rapid reconstitution of the peripheral and the gut CD8 T cell compartments a) by studying the mechanisms involved HSC division and T cell commitment; b) by isolating and characterizing progenitors we previously described that are T cell committed and able of an accelerated CD8 reconstitution c) by developing new strategies that may allow stable thymus transplantation and continuous thymus T cell generation. 2) To determine the mechanics associated to efficient CD8 memory generation a) by evaluating cellular modifications that ensure the efficient division and the remarkable accumulation and survival of CD8 T cells during the adequate immune responses as compared to inefficient responses b) by studying CD8 differentiation into effector and memory cells in both conditions. These studies will use original experiment mouse models we develop in the laboratory, that allow to address each of these aims. Besides state of the art methods, they will also apply unique very advanced approaches we introduced and are the sole laboratory to perform.
Max ERC Funding
1 969 644 €
Duration
Start date: 2009-02-01, End date: 2014-05-31
Project acronym CDNF
Project Compartmentalization and dynamics of Nuclear functions
Researcher (PI) Angela Taddei
Host Institution (HI) INSTITUT CURIE
Call Details Starting Grant (StG), LS2, ERC-2007-StG
Summary The eukaryotic genome is packaged into large-scale chromatin structures that occupy distinct domains in the nucleus and this organization is now seen as a key contributor to genome functions. Two key functions of the genome can take advantage of nuclear organization: regulated gene expression and the propagation of a stable genome. To understand these fundamental processes, we have chosen to use yeast as a model system that allows genetics, molecular biology and advanced live microscopy approaches to be combined. Budding yeast have been very powerful to demonstrate that gene position can play an active role in regulating gene expression. Distinct subcompartments dedicated to either gene silencing or activation of specific genes are positioned at the nuclear periphery. To gain insight into the mechanisms underlying this sub-compartmentalization, we will address three complementary issues: - What are the mechanisms involved in the establishment and maintenance of silent nuclear compartments? - How and why are some activated genes recruited to the nuclear periphery? - What are the relationships between repressive and activating nuclear compartments? Concerning the maintenance of genome integrity, recent advances in yeast highlight the importance of nuclear architecture. However, how nuclear organization influences the formation and processing of DNA lesions remain poorly understood. We will focus on two main questions: - How and where in the nucleus are double strand breaks recognized, processed, and repaired? - Where do breaks or gaps resulting from replicative stress at 'fragile sites' arise in the nucleus and how does nuclear organization influence their stability? We hope to gain a better understanding of the mechanisms presiding nuclear organization and its importance for genome functions. These mechanisms are likely to be conserved and will be subsequently tested in higher eukaryotic cells.
Summary
The eukaryotic genome is packaged into large-scale chromatin structures that occupy distinct domains in the nucleus and this organization is now seen as a key contributor to genome functions. Two key functions of the genome can take advantage of nuclear organization: regulated gene expression and the propagation of a stable genome. To understand these fundamental processes, we have chosen to use yeast as a model system that allows genetics, molecular biology and advanced live microscopy approaches to be combined. Budding yeast have been very powerful to demonstrate that gene position can play an active role in regulating gene expression. Distinct subcompartments dedicated to either gene silencing or activation of specific genes are positioned at the nuclear periphery. To gain insight into the mechanisms underlying this sub-compartmentalization, we will address three complementary issues: - What are the mechanisms involved in the establishment and maintenance of silent nuclear compartments? - How and why are some activated genes recruited to the nuclear periphery? - What are the relationships between repressive and activating nuclear compartments? Concerning the maintenance of genome integrity, recent advances in yeast highlight the importance of nuclear architecture. However, how nuclear organization influences the formation and processing of DNA lesions remain poorly understood. We will focus on two main questions: - How and where in the nucleus are double strand breaks recognized, processed, and repaired? - Where do breaks or gaps resulting from replicative stress at 'fragile sites' arise in the nucleus and how does nuclear organization influence their stability? We hope to gain a better understanding of the mechanisms presiding nuclear organization and its importance for genome functions. These mechanisms are likely to be conserved and will be subsequently tested in higher eukaryotic cells.
Max ERC Funding
1 000 000 €
Duration
Start date: 2008-09-01, End date: 2014-05-31
Project acronym Celcelfus
Project Cell-Cell fusion in fertilization and developmental biology: a structural biology approach
Researcher (PI) Félix A. Rey
Host Institution (HI) INSTITUT PASTEUR
Call Details Advanced Grant (AdG), LS1, ERC-2013-ADG
Summary My group has made seminal contributions in the past toward understanding the mechanism of membrane fusion used by enveloped viruses to infect a cell. This aim of this ERC grant proposal is to achieve similar breakthroughs in understanding fusion between cells, both during fertilization and organogenesis. This proposal is based in recent important results not yet published.
We have determined the crystal structure of the C. elegans protein EFF-1, a member of the “fusion family” (FF). EFF-1 is responsible for a cell-cell fusion event during skin formation in the nematode. Strikingly, the crystal structure shows that EFF-1 is homologous to the “Class II” viral protein fusogens, thus indicating that they have diverged from a common ancestor. The observed homology could not be identified by other means because the proteins have diverged to the point where no remnants of sequence similarity are left, yet the tertiary and quaternary organization is the same. However, the homotypic fusion mechanism of EFF-1 is clearly different to that of viral fusion proteins.
This proposal intends to build on the momentum generated by this exciting discovery, in an attempt to cast light into the fusion mechanism of FF proteins. We will reconstitute them in artificial liposomes and will also follow them within cells with the use of light microscopy. We will also focus in determining the crystal structure of the monomeric pre-fusion form of EFF-1,and of the intact trans-membrane post fusion trimer. In parallel, we want to make use the experience accumulated over the years in crystallizing viral glycoproteins, to apply it to the conserved family of HAP2/GSC1 proteins involved in fusion of gametes during fertilization. These proteins exhibit a similar pattern of secondary structure elements in the ectodomain as class II proteins, but only a crystallographic analysis can identify a possible structural homology and provide the basis to understand the molecular mechanisms of cell-cell fusion.
Summary
My group has made seminal contributions in the past toward understanding the mechanism of membrane fusion used by enveloped viruses to infect a cell. This aim of this ERC grant proposal is to achieve similar breakthroughs in understanding fusion between cells, both during fertilization and organogenesis. This proposal is based in recent important results not yet published.
We have determined the crystal structure of the C. elegans protein EFF-1, a member of the “fusion family” (FF). EFF-1 is responsible for a cell-cell fusion event during skin formation in the nematode. Strikingly, the crystal structure shows that EFF-1 is homologous to the “Class II” viral protein fusogens, thus indicating that they have diverged from a common ancestor. The observed homology could not be identified by other means because the proteins have diverged to the point where no remnants of sequence similarity are left, yet the tertiary and quaternary organization is the same. However, the homotypic fusion mechanism of EFF-1 is clearly different to that of viral fusion proteins.
This proposal intends to build on the momentum generated by this exciting discovery, in an attempt to cast light into the fusion mechanism of FF proteins. We will reconstitute them in artificial liposomes and will also follow them within cells with the use of light microscopy. We will also focus in determining the crystal structure of the monomeric pre-fusion form of EFF-1,and of the intact trans-membrane post fusion trimer. In parallel, we want to make use the experience accumulated over the years in crystallizing viral glycoproteins, to apply it to the conserved family of HAP2/GSC1 proteins involved in fusion of gametes during fertilization. These proteins exhibit a similar pattern of secondary structure elements in the ectodomain as class II proteins, but only a crystallographic analysis can identify a possible structural homology and provide the basis to understand the molecular mechanisms of cell-cell fusion.
Max ERC Funding
2 478 800 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym CENEVO
Project A new paradigm for centromere biology:Evolution and mechanism of CenH3-independent chromosome segregation in holocentric insects
Researcher (PI) Ines DRINNENBERG
Host Institution (HI) INSTITUT CURIE
Call Details Starting Grant (StG), LS2, ERC-2017-STG
Summary Faithful chromosome segregation in all eukaryotes relies on centromeres, the chromosomal sites that recruit kinetochore proteins and mediate spindle attachment during cell division. Fundamental to centromere function is a histone H3 variant, CenH3, that initiates kinetochore assembly on centromeric DNA. CenH3 is conserved throughout most eukaryotes; its deletion is lethal in all organisms tested. These findings established the paradigm that CenH3 is an absolute requirement for centromere function. My recent findings undermined this paradigm of CenH3 essentiality. I showed that CenH3 was lost independently in four lineages of insects. These losses are concomitant with dramatic changes in their centromeric architecture, in which each lineage independently transitioned from monocentromeres (where microtubules attach to a single chromosomal region) to holocentromeres (where microtubules attach along the entire length of the chromosome). Here, I aim to characterize this unique CenH3-deficient chromosome segregation pathway. Using proteomic and genomic approaches in lepidopteran cell lines, I will determine the mechanism of CenH3-independent kinetochore assembly that led to the establishment of their holocentric architecture. Using comparative genomic approaches, I will determine whether this kinetochore assembly pathway has recurrently evolved over the course of 400 million years of evolution and its impact on the chromosome segregation machinery.
My discovery of CenH3 loss in holocentric insects establishes a new class of centromeres. My research will reveal how CenH3 that is essential in most other eukaryotes, could have become dispensable in holocentric insects. Since the evolution of this CenH3-independent chromosome segregation pathway is associated with the independent rises of holocentric architectures, my research will also provide the first insights into the transition from a monocentromere to a holocentromere.
Summary
Faithful chromosome segregation in all eukaryotes relies on centromeres, the chromosomal sites that recruit kinetochore proteins and mediate spindle attachment during cell division. Fundamental to centromere function is a histone H3 variant, CenH3, that initiates kinetochore assembly on centromeric DNA. CenH3 is conserved throughout most eukaryotes; its deletion is lethal in all organisms tested. These findings established the paradigm that CenH3 is an absolute requirement for centromere function. My recent findings undermined this paradigm of CenH3 essentiality. I showed that CenH3 was lost independently in four lineages of insects. These losses are concomitant with dramatic changes in their centromeric architecture, in which each lineage independently transitioned from monocentromeres (where microtubules attach to a single chromosomal region) to holocentromeres (where microtubules attach along the entire length of the chromosome). Here, I aim to characterize this unique CenH3-deficient chromosome segregation pathway. Using proteomic and genomic approaches in lepidopteran cell lines, I will determine the mechanism of CenH3-independent kinetochore assembly that led to the establishment of their holocentric architecture. Using comparative genomic approaches, I will determine whether this kinetochore assembly pathway has recurrently evolved over the course of 400 million years of evolution and its impact on the chromosome segregation machinery.
My discovery of CenH3 loss in holocentric insects establishes a new class of centromeres. My research will reveal how CenH3 that is essential in most other eukaryotes, could have become dispensable in holocentric insects. Since the evolution of this CenH3-independent chromosome segregation pathway is associated with the independent rises of holocentric architectures, my research will also provide the first insights into the transition from a monocentromere to a holocentromere.
Max ERC Funding
1 497 500 €
Duration
Start date: 2018-04-01, End date: 2023-03-31
Project acronym CENTROSTEMCANCER
Project Investigating the link between centrosomes, stem cells and cancer
Researcher (PI) Renata Homem De Gouveia Xavier De Basto
Host Institution (HI) INSTITUT CURIE
Call Details Starting Grant (StG), LS3, ERC-2009-StG
Summary Centrosomes are cytoplasmic organelles found in most animal cells with important roles in polarity establishment and maintenance. Theodor Boveri s pioneering work first suggested that extra-centrosomes could contribute to genetic instability and consequently to tumourigenesis. Although many human tumours do exhibit centrosome amplification (extra centrosomes) or centrosome abnormalities, the exact contribution of centrosomes to tumour initiation in vertebrate organisms remains to be determined. I have recently showed that Drosophila flies carrying extra-centrosomes, following the over-expression of the centriole replication kinase Sak, did not exhibit chromosome segregation errors and were able to maintain a stable diploid genome over many generations. Surprisingly, however, neural stem cells fail frequently to align the mitotic spindle with their polarity axis during asymmetric division. Moreover, I have found that centrosome amplification is permissive to tumour formation in flies. So far, however, we do not know the molecular mechanisms that allow transformation when extra centrosomes are present and elucidating these mechanisms is the aim of the work presented in this proposal. Here, I describe a series of complementary approaches that will help us to decipher the link between centrosomes, stem cells and tumour biology. In addition, I wish to pursue the original observations made in Drosophila and investigate the consequences of centrosome amplification in mammals.
Summary
Centrosomes are cytoplasmic organelles found in most animal cells with important roles in polarity establishment and maintenance. Theodor Boveri s pioneering work first suggested that extra-centrosomes could contribute to genetic instability and consequently to tumourigenesis. Although many human tumours do exhibit centrosome amplification (extra centrosomes) or centrosome abnormalities, the exact contribution of centrosomes to tumour initiation in vertebrate organisms remains to be determined. I have recently showed that Drosophila flies carrying extra-centrosomes, following the over-expression of the centriole replication kinase Sak, did not exhibit chromosome segregation errors and were able to maintain a stable diploid genome over many generations. Surprisingly, however, neural stem cells fail frequently to align the mitotic spindle with their polarity axis during asymmetric division. Moreover, I have found that centrosome amplification is permissive to tumour formation in flies. So far, however, we do not know the molecular mechanisms that allow transformation when extra centrosomes are present and elucidating these mechanisms is the aim of the work presented in this proposal. Here, I describe a series of complementary approaches that will help us to decipher the link between centrosomes, stem cells and tumour biology. In addition, I wish to pursue the original observations made in Drosophila and investigate the consequences of centrosome amplification in mammals.
Max ERC Funding
1 550 000 €
Duration
Start date: 2010-01-01, End date: 2015-06-30
Project acronym CEPODRO
Project Cell polarization in Drosophila
Researcher (PI) Yohanns Bellaiche
Host Institution (HI) INSTITUT CURIE
Call Details Starting Grant (StG), LS1, ERC-2007-StG
Summary Cell polarity is fundamental to many aspects of cell and developmental biology and it is implicated in differentiation, proliferation and morphogenesis in both unicellular and multi-cellular organisms. We study the mechanisms that regulate cell polarity during both asymmetric cell division and epithelial cell polarization in Drosophila. To understand these fundamental processes, we are currently using two complementary approaches. Firstly, we are coupling genetic tools to state of the art time-lapse microscopy to genetically dissect the mechanisms of cortical cell polarization and mitotic spindle orientation. Secondly, we are introducing two innovative inter-disciplinary methodologies into the fields of cell and developmental biology: 1) single molecule imaging during asymmetric cell division, to unravel the mechanism of polarized protein distribution within the cell; 2) multi-scale tensor analysis of epithelial tissues to describe and understand how epithelial tissues grow, acquire and maintain their shape and organization during development. Using both conventional and innovative methodologies, our goals over the next four years are to better understand how molecules and protein complexes move and are activated at different locations within the cell and how cell polarization impacts on cell identities and on epithelial tissue growth and morphogenesis. Since the mechanisms underlying cell polarization are conserved throughout evolution, the proposed experiments will improve our understanding of these processes not only in Drosophila, but in all animals.
Summary
Cell polarity is fundamental to many aspects of cell and developmental biology and it is implicated in differentiation, proliferation and morphogenesis in both unicellular and multi-cellular organisms. We study the mechanisms that regulate cell polarity during both asymmetric cell division and epithelial cell polarization in Drosophila. To understand these fundamental processes, we are currently using two complementary approaches. Firstly, we are coupling genetic tools to state of the art time-lapse microscopy to genetically dissect the mechanisms of cortical cell polarization and mitotic spindle orientation. Secondly, we are introducing two innovative inter-disciplinary methodologies into the fields of cell and developmental biology: 1) single molecule imaging during asymmetric cell division, to unravel the mechanism of polarized protein distribution within the cell; 2) multi-scale tensor analysis of epithelial tissues to describe and understand how epithelial tissues grow, acquire and maintain their shape and organization during development. Using both conventional and innovative methodologies, our goals over the next four years are to better understand how molecules and protein complexes move and are activated at different locations within the cell and how cell polarization impacts on cell identities and on epithelial tissue growth and morphogenesis. Since the mechanisms underlying cell polarization are conserved throughout evolution, the proposed experiments will improve our understanding of these processes not only in Drosophila, but in all animals.
Max ERC Funding
1 159 000 €
Duration
Start date: 2008-09-01, End date: 2013-08-31
Project acronym CEVAL
Project Clonal evolution in acute leukemia: from molecular and functional profiling towards therapeutic intervention
Researcher (PI) Jean Soulier
Host Institution (HI) UNIVERSITE PARIS DIDEROT - PARIS 7
Call Details Starting Grant (StG), LS7, ERC-2012-StG_20111109
Summary "Clonal evolution represents a driving force for cancer cells and a major challenge for therapy. There is a long-standing knowledge that clonal diversification and selection allows cancer cells to pass through distinct steps of transformation. More recently has emerged the concept that ""more malignant"" leukemia subclones can persist after treatment leading to relapse. My research is focused on two diseases which represent privileged models to study clonal evolution: Fanconi anemia (FA), which is a genetic condition predisposing to acute leukemia, and T-cell acute lymphoblastic leukemia (T-ALL), an aggressive leukemia which frequently relapses. The goal of this project is to decipher the crucial molecular and cellular events that drive gain of malignancy and to design new strategies to follow up and treat the patients. Specifically, we aim to:
1. Identify the key pathways involved in leukemia progression at two major steps: from pre-leukemia to overt leukemia (in FA), and from primary diagnosis to relapse (in T-ALL). This aim will use high-throughput molecular profiling and functional characterization of longitudinal samples from patients.
2. Model leukemia progression in vivo to functionally validate these pathways. We have developed read-outs based on gene silencing in human primary cells from patients followed by leukemia monitoring in immunodeficient mice.
3. Define new strategies to prevent the transition towards acute leukemia in pre-leukemic states in FA patients. We will develop new markers for transformation that should help in monitoring therapeutic intervention.
4. Test drugs in pre-clinical models to target critical pathways of relapsed ALL. We will use xenografted T-ALL which recapitulates leukemia progression.
Completion of this innovative transversal project should markedly improve the knowledge on tumor progression and lead to new strategies to prevent, early detect and/or treat relapse, with the final objective to cure more patients."
Summary
"Clonal evolution represents a driving force for cancer cells and a major challenge for therapy. There is a long-standing knowledge that clonal diversification and selection allows cancer cells to pass through distinct steps of transformation. More recently has emerged the concept that ""more malignant"" leukemia subclones can persist after treatment leading to relapse. My research is focused on two diseases which represent privileged models to study clonal evolution: Fanconi anemia (FA), which is a genetic condition predisposing to acute leukemia, and T-cell acute lymphoblastic leukemia (T-ALL), an aggressive leukemia which frequently relapses. The goal of this project is to decipher the crucial molecular and cellular events that drive gain of malignancy and to design new strategies to follow up and treat the patients. Specifically, we aim to:
1. Identify the key pathways involved in leukemia progression at two major steps: from pre-leukemia to overt leukemia (in FA), and from primary diagnosis to relapse (in T-ALL). This aim will use high-throughput molecular profiling and functional characterization of longitudinal samples from patients.
2. Model leukemia progression in vivo to functionally validate these pathways. We have developed read-outs based on gene silencing in human primary cells from patients followed by leukemia monitoring in immunodeficient mice.
3. Define new strategies to prevent the transition towards acute leukemia in pre-leukemic states in FA patients. We will develop new markers for transformation that should help in monitoring therapeutic intervention.
4. Test drugs in pre-clinical models to target critical pathways of relapsed ALL. We will use xenografted T-ALL which recapitulates leukemia progression.
Completion of this innovative transversal project should markedly improve the knowledge on tumor progression and lead to new strategies to prevent, early detect and/or treat relapse, with the final objective to cure more patients."
Max ERC Funding
1 497 028 €
Duration
Start date: 2013-04-01, End date: 2018-03-31
Project acronym Chap4Resp
Project Catching in action a novel bacterial chaperone for respiratory complexes
Researcher (PI) Irina Gutsche
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS1, ERC-2014-CoG
Summary Cellular respiration provides energy to power essential processes of life. Respiratory complexes are macromolecular batteries coupling electron flow through a wire of metal clusters and cofactors with proton transfer across the inner membrane of mitochondria and bacteria. Waste products of these cellular factories are reactive oxygen species causing ageing and diseases. Assembly and maturation mechanisms of respiratory complexes remain enigmatic because of their membrane location, multisubunit composition and cofactor insertion. E. coli Complex I, one of the largest membrane proteins, composed of 14 conserved subunits with 9 Fe/S clusters and a flavin, is a minimal model for its 45-subunit human homologue. When proton pumping by respiratory complexes is affected, bacteria become resistant to antibiotics requiring proton gradient for uptake. Based on the latest genetic data, we realize that the huge E. coli macromolecular cage, the structure of which we recently solved by cryo-electron microscopy (cryoEM), in conjunction with a novel protein cofactor, is a specific chaperone for Fe/S cluster biogenesis and assembly of respiratory complexes. This integrated multidisciplinary project combines cryoEM and other structural, biophysical and spectroscopic techniques, to uncover the functional mechanism of this emerging chaperone. The structural plasticity of the chaperone fuelled by ATP hydrolysis, and its interaction with Fe/S cluster biogenesis systems and the main respiratory complexes as a function of stresses, will be scrutinized to gain quasiatomic insights into the way the chaperone operates on its substrates. A novel technology for synergetic in situ investigation of protein complexes in the bacterial cytoplasm by optical imaging, state-of-the-art cryogenic correlative light and electron microscopy, and subtomogram analysis, will be developed and used to obtain snapshots of the chaperone-substrate interactions in the cellular context.
Summary
Cellular respiration provides energy to power essential processes of life. Respiratory complexes are macromolecular batteries coupling electron flow through a wire of metal clusters and cofactors with proton transfer across the inner membrane of mitochondria and bacteria. Waste products of these cellular factories are reactive oxygen species causing ageing and diseases. Assembly and maturation mechanisms of respiratory complexes remain enigmatic because of their membrane location, multisubunit composition and cofactor insertion. E. coli Complex I, one of the largest membrane proteins, composed of 14 conserved subunits with 9 Fe/S clusters and a flavin, is a minimal model for its 45-subunit human homologue. When proton pumping by respiratory complexes is affected, bacteria become resistant to antibiotics requiring proton gradient for uptake. Based on the latest genetic data, we realize that the huge E. coli macromolecular cage, the structure of which we recently solved by cryo-electron microscopy (cryoEM), in conjunction with a novel protein cofactor, is a specific chaperone for Fe/S cluster biogenesis and assembly of respiratory complexes. This integrated multidisciplinary project combines cryoEM and other structural, biophysical and spectroscopic techniques, to uncover the functional mechanism of this emerging chaperone. The structural plasticity of the chaperone fuelled by ATP hydrolysis, and its interaction with Fe/S cluster biogenesis systems and the main respiratory complexes as a function of stresses, will be scrutinized to gain quasiatomic insights into the way the chaperone operates on its substrates. A novel technology for synergetic in situ investigation of protein complexes in the bacterial cytoplasm by optical imaging, state-of-the-art cryogenic correlative light and electron microscopy, and subtomogram analysis, will be developed and used to obtain snapshots of the chaperone-substrate interactions in the cellular context.
Max ERC Funding
1 999 956 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym ChloroMito
Project Chloroplast and Mitochondria interactions for microalgal acclimation
Researcher (PI) Giovanni Finazzi
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS8, ERC-2018-ADG
Summary Photosynthesis emerged as an energy-harvesting process at least 3.5 billion years ago, first in anoxygenic bacteria and then in oxygen-producing organisms, which led to the evolution of complex life forms with oxygen-based metabolisms (e.g. humans). Oxygenic photosynthesis produces ATP and NADPH, and the correct balance between these energy-rich molecules allows assimilation of CO2 into organic matter. Although the mechanisms of ATP/NADPH synthesis are well understood, less is known about how CO2 assimilation was optimised. This process was essential to the successful phototrophic colonisation of land (by Plantae) and the oceans (by phytoplankton). Plants optimised CO2 assimilation using chloroplast-localised ATP-generating processes to control the ATP/NADPH ratio, but the strategies developed by phytoplankton are poorly understood. However, diatoms—ecologically successful ocean organisms—are known to control this ratio by exchanging energy between plastids and mitochondria. Is this mechanism a paradigm for optimisation of photosynthesis in the ocean? The ChloroMito project aims to first decipher the mechanism(s) behind plastid-mitochondria interactions. Thanks to a novel combination of whole-cell approaches, including (opto)genetics, cellular tomography and single-cell spectroscopy, we will identify the nature of the exchanges occurring in diatoms and assess their contribution to dynamic responses to environmental stimuli (light, temperature, nutrients). We will then assess conservation of this mechanism in ecologically relevant phytoplankton taxa, test its role in supporting different lifestyles (autotrophy, mixotrophy, photosymbiosis) encountered in the ocean, and track transitions between these different lifestyles as part of an unprecedented effort to visualise ocean dynamics. Overall, the ChloroMito project will alter our understanding of ocean photosynthesis, challenging textbook concepts which are often inferred from plant-based concepts
Summary
Photosynthesis emerged as an energy-harvesting process at least 3.5 billion years ago, first in anoxygenic bacteria and then in oxygen-producing organisms, which led to the evolution of complex life forms with oxygen-based metabolisms (e.g. humans). Oxygenic photosynthesis produces ATP and NADPH, and the correct balance between these energy-rich molecules allows assimilation of CO2 into organic matter. Although the mechanisms of ATP/NADPH synthesis are well understood, less is known about how CO2 assimilation was optimised. This process was essential to the successful phototrophic colonisation of land (by Plantae) and the oceans (by phytoplankton). Plants optimised CO2 assimilation using chloroplast-localised ATP-generating processes to control the ATP/NADPH ratio, but the strategies developed by phytoplankton are poorly understood. However, diatoms—ecologically successful ocean organisms—are known to control this ratio by exchanging energy between plastids and mitochondria. Is this mechanism a paradigm for optimisation of photosynthesis in the ocean? The ChloroMito project aims to first decipher the mechanism(s) behind plastid-mitochondria interactions. Thanks to a novel combination of whole-cell approaches, including (opto)genetics, cellular tomography and single-cell spectroscopy, we will identify the nature of the exchanges occurring in diatoms and assess their contribution to dynamic responses to environmental stimuli (light, temperature, nutrients). We will then assess conservation of this mechanism in ecologically relevant phytoplankton taxa, test its role in supporting different lifestyles (autotrophy, mixotrophy, photosymbiosis) encountered in the ocean, and track transitions between these different lifestyles as part of an unprecedented effort to visualise ocean dynamics. Overall, the ChloroMito project will alter our understanding of ocean photosynthesis, challenging textbook concepts which are often inferred from plant-based concepts
Max ERC Funding
2 498 207 €
Duration
Start date: 2020-01-01, End date: 2024-12-31
Project acronym CholAminCo
Project Synergy and antagonism of cholinergic and dopaminergic systems in associative learning
Researcher (PI) Balazs Gyoergy HANGYA
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Starting Grant (StG), LS5, ERC-2016-STG
Summary Neuromodulators such as acetylcholine and dopamine are able to rapidly reprogram neuronal information processing and dynamically change brain states. Degeneration or dysfunction of cholinergic and dopaminergic neurons can lead to neuropsychiatric conditions like schizophrenia and addiction or cognitive diseases such as Alzheimer’s. Neuromodulatory systems control overlapping cognitive processes and often have similar modes of action; therefore it is important to reveal cooperation and competition between different systems to understand their unique contributions to cognitive functions like learning, memory and attention. This is only possible by direct comparison, which necessitates monitoring multiple neuromodulatory systems under identical experimental conditions. Moreover, simultaneous recording of different neuromodulatory cell types goes beyond phenomenological description of similarities and differences by revealing the underlying correlation structure at the level of action potential timing. However, such data allowing direct comparison of neuromodulatory actions are still sparse. As a first step to bridge this gap, I propose to elucidate the unique versus complementary roles of two “classical” neuromodulatory systems, the cholinergic and dopaminergic projection system implicated in various cognitive functions including associative learning and plasticity. First, we will record optogenetically identified cholinergic and dopaminergic neurons simultaneously using chronic extracellular recording in mice undergoing classical and operant conditioning. Second, we will determine the postsynaptic impact of cholinergic and dopaminergic neurons by manipulating them both separately and simultaneously while recording consequential changes in cortical neuronal activity and learning behaviour. These experiments will reveal how major neuromodulatory systems interact to mediate similar or different aspects of the same cognitive functions.
Summary
Neuromodulators such as acetylcholine and dopamine are able to rapidly reprogram neuronal information processing and dynamically change brain states. Degeneration or dysfunction of cholinergic and dopaminergic neurons can lead to neuropsychiatric conditions like schizophrenia and addiction or cognitive diseases such as Alzheimer’s. Neuromodulatory systems control overlapping cognitive processes and often have similar modes of action; therefore it is important to reveal cooperation and competition between different systems to understand their unique contributions to cognitive functions like learning, memory and attention. This is only possible by direct comparison, which necessitates monitoring multiple neuromodulatory systems under identical experimental conditions. Moreover, simultaneous recording of different neuromodulatory cell types goes beyond phenomenological description of similarities and differences by revealing the underlying correlation structure at the level of action potential timing. However, such data allowing direct comparison of neuromodulatory actions are still sparse. As a first step to bridge this gap, I propose to elucidate the unique versus complementary roles of two “classical” neuromodulatory systems, the cholinergic and dopaminergic projection system implicated in various cognitive functions including associative learning and plasticity. First, we will record optogenetically identified cholinergic and dopaminergic neurons simultaneously using chronic extracellular recording in mice undergoing classical and operant conditioning. Second, we will determine the postsynaptic impact of cholinergic and dopaminergic neurons by manipulating them both separately and simultaneously while recording consequential changes in cortical neuronal activity and learning behaviour. These experiments will reveal how major neuromodulatory systems interact to mediate similar or different aspects of the same cognitive functions.
Max ERC Funding
1 499 463 €
Duration
Start date: 2017-05-01, End date: 2022-04-30
Project acronym ChromADICT
Project Chromatin Adaptations through Interactions of Chaperones in Time
Researcher (PI) Genevieve ALMOUZNI
Host Institution (HI) INSTITUT CURIE
Call Details Advanced Grant (AdG), LS1, ERC-2015-AdG
Summary A central question in chromatin biology is how to organize the genome and mark specific regions with histone variants. Understanding how to establish and maintain, but also change chromatin states is a fundamental challenge. Histone chaperones, escort factors that regulate the supply, loading, and degradation of histone variants, are key in their placement at specific chromatin landmarks and bridge organization from nucleosomes to higher order structures. A series of studies have underlined chaperone-variant partner selectivity in multicellular organisms, yet recently, dosage imbalances in natural and pathological contexts highlight plasticity in these interactions. Considering known changes in histone dosage during development, one should evaluate chaperone function not as fixed modules, but as a dynamic circuitry that adapts to cellular needs during the cell cycle, replication and repair, differentiation, development and pathology.
Here we propose to decipher the mechanisms enabling adaptability to natural and experimentally induced changes in the dosage of histone chaperones and variants over time. To follow new and old proteins, and control dosage, we will engineer cellular and animal models and exploit quantitative readout methods using mass spectrometry, imaging, and single-cell approaches. We will evaluate with an unprecedented level of detail the impact on i) soluble histone complexes and ii) specific chromatin landmarks (centromere, telomeres, heterochromatin and regulatory elements) and their crosstalk. We will apply this to determine the impact of these parameters during distinct developmental transitions, such as ES cell differentiation and T cell commitment in mice.
We aim to define general principles for variants in nuclear organization and dynamic changes during the cell cycle/repair and in differentiation and unravel locus specific-roles of chaperones as architects and bricklayers of the genome, in designing and building specific nuclear domains.
Summary
A central question in chromatin biology is how to organize the genome and mark specific regions with histone variants. Understanding how to establish and maintain, but also change chromatin states is a fundamental challenge. Histone chaperones, escort factors that regulate the supply, loading, and degradation of histone variants, are key in their placement at specific chromatin landmarks and bridge organization from nucleosomes to higher order structures. A series of studies have underlined chaperone-variant partner selectivity in multicellular organisms, yet recently, dosage imbalances in natural and pathological contexts highlight plasticity in these interactions. Considering known changes in histone dosage during development, one should evaluate chaperone function not as fixed modules, but as a dynamic circuitry that adapts to cellular needs during the cell cycle, replication and repair, differentiation, development and pathology.
Here we propose to decipher the mechanisms enabling adaptability to natural and experimentally induced changes in the dosage of histone chaperones and variants over time. To follow new and old proteins, and control dosage, we will engineer cellular and animal models and exploit quantitative readout methods using mass spectrometry, imaging, and single-cell approaches. We will evaluate with an unprecedented level of detail the impact on i) soluble histone complexes and ii) specific chromatin landmarks (centromere, telomeres, heterochromatin and regulatory elements) and their crosstalk. We will apply this to determine the impact of these parameters during distinct developmental transitions, such as ES cell differentiation and T cell commitment in mice.
We aim to define general principles for variants in nuclear organization and dynamic changes during the cell cycle/repair and in differentiation and unravel locus specific-roles of chaperones as architects and bricklayers of the genome, in designing and building specific nuclear domains.
Max ERC Funding
2 499 697 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym CHROMONUMBER
Project Chromosome number variations in vivo: probing mechanisms of genesis and elimination
Researcher (PI) Renata BASTO
Host Institution (HI) INSTITUT CURIE
Call Details Consolidator Grant (CoG), LS3, ERC-2016-COG
Summary How variations in whole chromosome number impact organism homeostasis remains an open question. Variations to the normal euploid genome content are frequently found in healthy animals and are thought to contribute with phenotypic variability in adverse situations. Yet they are also at the basis of several human diseases, including neuro-developmental disorders and cancer. Our preliminary data shows that physiological aneuploidy can be identified in certain cells during development. Moreover, we have observed that when induced through mutations, non-euploid cells are effectively eliminated from the cycling population. A quantitative view of the frequency of non-euploid karyotypes and the mechanisms underlying their genesis is lacking in the literature. Further, the tissue specific responses at play to eliminate non-euploid cells, when induced through mutations are not understood. The objectives of this proposal are to quantitatively assess the occurrence of physiological chromosome number variations gaining insight into mechanisms involved in generating it. Additionally, we will identify the tissue-specific pathways involved in maintaining organism homeostasis through the elimination of non-euploid cells. We will use a novel genetic approach to monitor individual chromosome loss at the level of the entire organism, combine it with quantitative methods and state-of-the art-microscopy, and focus on two model organisms - Drosophila and mouse - during development and adulthood. We predict that the findings resulting from this proposal will significantly impact the fields of cell, developmental and animal physiology, generating novel concepts that will bridge the existing gaps in the field, and expand our understanding of the links between karyotype variations, animal development and disease establishment.
Summary
How variations in whole chromosome number impact organism homeostasis remains an open question. Variations to the normal euploid genome content are frequently found in healthy animals and are thought to contribute with phenotypic variability in adverse situations. Yet they are also at the basis of several human diseases, including neuro-developmental disorders and cancer. Our preliminary data shows that physiological aneuploidy can be identified in certain cells during development. Moreover, we have observed that when induced through mutations, non-euploid cells are effectively eliminated from the cycling population. A quantitative view of the frequency of non-euploid karyotypes and the mechanisms underlying their genesis is lacking in the literature. Further, the tissue specific responses at play to eliminate non-euploid cells, when induced through mutations are not understood. The objectives of this proposal are to quantitatively assess the occurrence of physiological chromosome number variations gaining insight into mechanisms involved in generating it. Additionally, we will identify the tissue-specific pathways involved in maintaining organism homeostasis through the elimination of non-euploid cells. We will use a novel genetic approach to monitor individual chromosome loss at the level of the entire organism, combine it with quantitative methods and state-of-the art-microscopy, and focus on two model organisms - Drosophila and mouse - during development and adulthood. We predict that the findings resulting from this proposal will significantly impact the fields of cell, developmental and animal physiology, generating novel concepts that will bridge the existing gaps in the field, and expand our understanding of the links between karyotype variations, animal development and disease establishment.
Max ERC Funding
2 000 000 €
Duration
Start date: 2017-07-01, End date: 2022-06-30
Project acronym CHROMTOPOLOGY
Project Understanding and manipulating the dynamics of chromosome topologies in transcriptional control
Researcher (PI) Thomas, Ivor Sexton
Host Institution (HI) CENTRE EUROPEEN DE RECHERCHE EN BIOLOGIE ET MEDECINE
Call Details Starting Grant (StG), LS2, ERC-2015-STG
Summary Transcriptional regulation of genes in eukaryotic cells requires a complex and highly regulated interplay of chromatin environment, epigenetic status of target sequences and several different transcription factors. Eukaryotic genomes are tightly packaged within nuclei, yet must be accessible for transcription, replication and repair. A striking correlation exists between chromatin topology and underlying gene activity. According to the textbook view, chromatin loops bring genes into direct contact with distal regulatory elements, such as enhancers. Moreover, we and others have shown that genomes are organized into discretely folded megabase-sized regions, denoted as topologically associated domains (TADs), which seem to correlate well with transcription activity and histone modifications. However, it is unknown whether chromosome folding is a cause or consequence of underlying gene function.
To better understand the role of genome organization in transcription regulation, I will address the following questions:
(i) How are chromatin configurations altered during transcriptional changes accompanying development?
(ii) What are the real-time kinetics and cell-to-cell variabilities of chromatin interactions and TAD architectures?
(iii) Can chromatin loops be engineered de novo, and do they influence gene expression?
(iv) What genetic elements and trans-acting factors are required to organize TADs?
To address these fundamental questions, I will use a combination of novel technologies and approaches, such as Hi-C, CRISPR knock-ins, ANCHOR tagging of DNA loci, high- and super-resolution single-cell imaging, genome-wide screens and optogenetics, in order to both study and engineer chromatin architectures.
These studies will give groundbreaking insight into if and how chromatin topology regulates transcription. Thus, I anticipate that the results of this project will have a major impact on the field and will lead to a new paradigm for metazoan transcription control.
Summary
Transcriptional regulation of genes in eukaryotic cells requires a complex and highly regulated interplay of chromatin environment, epigenetic status of target sequences and several different transcription factors. Eukaryotic genomes are tightly packaged within nuclei, yet must be accessible for transcription, replication and repair. A striking correlation exists between chromatin topology and underlying gene activity. According to the textbook view, chromatin loops bring genes into direct contact with distal regulatory elements, such as enhancers. Moreover, we and others have shown that genomes are organized into discretely folded megabase-sized regions, denoted as topologically associated domains (TADs), which seem to correlate well with transcription activity and histone modifications. However, it is unknown whether chromosome folding is a cause or consequence of underlying gene function.
To better understand the role of genome organization in transcription regulation, I will address the following questions:
(i) How are chromatin configurations altered during transcriptional changes accompanying development?
(ii) What are the real-time kinetics and cell-to-cell variabilities of chromatin interactions and TAD architectures?
(iii) Can chromatin loops be engineered de novo, and do they influence gene expression?
(iv) What genetic elements and trans-acting factors are required to organize TADs?
To address these fundamental questions, I will use a combination of novel technologies and approaches, such as Hi-C, CRISPR knock-ins, ANCHOR tagging of DNA loci, high- and super-resolution single-cell imaging, genome-wide screens and optogenetics, in order to both study and engineer chromatin architectures.
These studies will give groundbreaking insight into if and how chromatin topology regulates transcription. Thus, I anticipate that the results of this project will have a major impact on the field and will lead to a new paradigm for metazoan transcription control.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
Project acronym CoBABATI
Project Cofactor Binding Antibodies – Basic Aspects and Therapeutic Innovations
Researcher (PI) Jordan Dimitrov
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS7, ERC-2015-STG
Summary The immune repertoire of healthy individuals contains a fraction of antibodies (Abs) that are able to bind with high affinity various endogenous or exogenous low molecular weight compounds, including cofactors essential for the aerobic life, such as riboflavin, heme and ATP. Despite identification of cofactor-binding Abs as a constituent of normal immune repertoires, their fundamental characteristics and have not been systematically investigated. Thus, we do not know the origin, prevalence and physiopathological significance of cofactor-binding Abs. Moreover, the molecular mechanisms of interaction of cofactors with Abs are ill defined. Different proteins use cofactors to extend the chemistry intrinsic to the amino acid sequence of their polypeptide chain(s). Thus, one can speculate that the alliance of Abs with low molecular weight compounds results in the emergence of untypical properties of Abs and offers a strategy for designing a new generation of therapeutic Abs. Moreover, cofactor-binding Abs may be used for delivery of cytotoxic compounds to particular sites in the body, or for scavenging pro-inflammatory compounds. The principal goal of the present proposal is to gain a basic understanding on the fraction of cofactor-binding Abs in immune repertoires and to use this knowledge for the rational design of novel classes of therapeutic Abs. In this project, we will address the following questions: 1) understand the origin and prevalence of cofactor-binding Abs in immune repertoires; 2) characterize the molecular mechanisms of interaction of cofactors with Abs; 3) Understand the physiopathological roles of cofactor-binding Abs, and 4) use cofactor binding for the development of novel types of therapeutic Abs. A comprehensive understanding of various aspects of cofactor-binding Abs should lead to advances in fundamental understanding and in the development of innovative therapeutic and diagnostic tools.
Summary
The immune repertoire of healthy individuals contains a fraction of antibodies (Abs) that are able to bind with high affinity various endogenous or exogenous low molecular weight compounds, including cofactors essential for the aerobic life, such as riboflavin, heme and ATP. Despite identification of cofactor-binding Abs as a constituent of normal immune repertoires, their fundamental characteristics and have not been systematically investigated. Thus, we do not know the origin, prevalence and physiopathological significance of cofactor-binding Abs. Moreover, the molecular mechanisms of interaction of cofactors with Abs are ill defined. Different proteins use cofactors to extend the chemistry intrinsic to the amino acid sequence of their polypeptide chain(s). Thus, one can speculate that the alliance of Abs with low molecular weight compounds results in the emergence of untypical properties of Abs and offers a strategy for designing a new generation of therapeutic Abs. Moreover, cofactor-binding Abs may be used for delivery of cytotoxic compounds to particular sites in the body, or for scavenging pro-inflammatory compounds. The principal goal of the present proposal is to gain a basic understanding on the fraction of cofactor-binding Abs in immune repertoires and to use this knowledge for the rational design of novel classes of therapeutic Abs. In this project, we will address the following questions: 1) understand the origin and prevalence of cofactor-binding Abs in immune repertoires; 2) characterize the molecular mechanisms of interaction of cofactors with Abs; 3) Understand the physiopathological roles of cofactor-binding Abs, and 4) use cofactor binding for the development of novel types of therapeutic Abs. A comprehensive understanding of various aspects of cofactor-binding Abs should lead to advances in fundamental understanding and in the development of innovative therapeutic and diagnostic tools.
Max ERC Funding
1 255 000 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym CODOVIREVOL
Project Evolution of viral codon usage preferences:manipulation of translation accuracy and evasion of immune response
Researcher (PI) Ignacio González Bravo
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS8, ERC-2014-CoG
Summary Fidelity during information transfer is essential for life, but it pays to be unfaithful if it provides an evolutionary advantage. The immune system continuously generates diversity to put up with recurrent pathogen challenges, and many viruses, in its turn, have evolved mechanisms to generate diversity to evade immune restrictions, even at the cost of enduring high mutation rates.
Synonymous codons are not used at random and are not translated with similar efficiency. A large proportion of viruses infecting humans, especially those causing chronic infections, display a poor adaptation to the codon usage preferences of their host. This observation is a paradox, as viral genes completely depend upon the cellular translation machinery for protein synthesis. The poor match between codon usage preferences of virus and host negatively affects speed and accuracy of viral protein translation. We propose here that maladaptation of codon usage preferences in human viruses may have an adaptive value as it decreases translational fidelity, results in the synthesis of an ill-defined population of viral proteins and provides a way to escape immune surveillance.
We will address the fitness effects of codon usage bias at the molecular and cellular levels, and later at the organism level in a rabbit model of papillomavirus infection. We will apply experimental evolution to analyse genotypic changes by means of next generation sequencing and will monitor phenotypic changes through real-time cell monitoring techniques, comparative proteomics, and anatomopathological analysis of virus-induced lesions.
Our results will help solve the evolutionary puzzle of codon usage bias, and will have implications for the development of therapeutic vaccines to guide the immune response towards the identification and targeting of the main protein species, avoiding the chemical noise generated by protein mistranslation.
Summary
Fidelity during information transfer is essential for life, but it pays to be unfaithful if it provides an evolutionary advantage. The immune system continuously generates diversity to put up with recurrent pathogen challenges, and many viruses, in its turn, have evolved mechanisms to generate diversity to evade immune restrictions, even at the cost of enduring high mutation rates.
Synonymous codons are not used at random and are not translated with similar efficiency. A large proportion of viruses infecting humans, especially those causing chronic infections, display a poor adaptation to the codon usage preferences of their host. This observation is a paradox, as viral genes completely depend upon the cellular translation machinery for protein synthesis. The poor match between codon usage preferences of virus and host negatively affects speed and accuracy of viral protein translation. We propose here that maladaptation of codon usage preferences in human viruses may have an adaptive value as it decreases translational fidelity, results in the synthesis of an ill-defined population of viral proteins and provides a way to escape immune surveillance.
We will address the fitness effects of codon usage bias at the molecular and cellular levels, and later at the organism level in a rabbit model of papillomavirus infection. We will apply experimental evolution to analyse genotypic changes by means of next generation sequencing and will monitor phenotypic changes through real-time cell monitoring techniques, comparative proteomics, and anatomopathological analysis of virus-induced lesions.
Our results will help solve the evolutionary puzzle of codon usage bias, and will have implications for the development of therapeutic vaccines to guide the immune response towards the identification and targeting of the main protein species, avoiding the chemical noise generated by protein mistranslation.
Max ERC Funding
1 997 100 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym COGNITION
Project Cognition and Decision-Making: Laws, Norms and Contracts
Researcher (PI) Jean Tirole
Host Institution (HI) FONDATION JEAN-JACQUES LAFFONT,TOULOUSE SCIENCES ECONOMIQUES
Call Details Advanced Grant (AdG), SH1, ERC-2009-AdG
Summary The application's unifying theme is cognition. Any decision reflects the information that comes to the decision-maker's awareness at the moment of making the decision. In turn, this information is the stochastic outcome of a sequence of more or less conscious choices and of awareness manipulation by third parties. The three parts of this application all are concerned with two factors of limited awareness (cognitive costs and motivated beliefs) and with the application of imperfect cognition to economics. The various projects can be subsumed into three themes, each with different subprojects: 1. Self-serving beliefs, laws, norms and taboos (expressive function of the law, taboos, dignity and contracts). 2. Cognition, markets, and contracts (mechanism design under costly cognition, directing attention in markets and politics). 3. Cognition and individual decision-making (foundations of some non-standard preferences). The methodology for this research will be that of formal economic modeling and welfare analysis, enriched with important insights from psychology and sociology. It will also include experimental (laboratory) investigations. The output will first take the form of a series of articles in economics journals, as well as, for the research described in Part 1, a book to disseminate the research to broader, multidisciplinary and non-specialized audiences.
Summary
The application's unifying theme is cognition. Any decision reflects the information that comes to the decision-maker's awareness at the moment of making the decision. In turn, this information is the stochastic outcome of a sequence of more or less conscious choices and of awareness manipulation by third parties. The three parts of this application all are concerned with two factors of limited awareness (cognitive costs and motivated beliefs) and with the application of imperfect cognition to economics. The various projects can be subsumed into three themes, each with different subprojects: 1. Self-serving beliefs, laws, norms and taboos (expressive function of the law, taboos, dignity and contracts). 2. Cognition, markets, and contracts (mechanism design under costly cognition, directing attention in markets and politics). 3. Cognition and individual decision-making (foundations of some non-standard preferences). The methodology for this research will be that of formal economic modeling and welfare analysis, enriched with important insights from psychology and sociology. It will also include experimental (laboratory) investigations. The output will first take the form of a series of articles in economics journals, as well as, for the research described in Part 1, a book to disseminate the research to broader, multidisciplinary and non-specialized audiences.
Max ERC Funding
1 910 400 €
Duration
Start date: 2010-04-01, End date: 2016-03-31
Project acronym COLOUR PATTERN
Project Morphogenesis and Molecular Regulation of Colour Patterning in Birds
Researcher (PI) Marie Celine Manceau
Host Institution (HI) COLLEGE DE FRANCE
Call Details Starting Grant (StG), LS3, ERC-2014-STG
Summary Animals display a tremendous diversity of patterns ‒from the colourful designs that adorn their body to repeated segmented appendages. Natural patterns result from the formation of discrete domains within developing tissues through the integration of positional cues by cells that consequently adopt specific fates and produce spatial heterogeneity. How can such developmental processes underlie the apparent complexity and diversity of natural patterns? We propose to address this long-standing question with an innovative experimental design: we will make use of natural variation as a powerful tool to facilitate the identification of patterning molecules and morphogenetic events. We will study colour pattern, a crucial adaptive trait that varies extensively in nature, from large colour domains to periodic designs. In amniotes, colour pattern is formed by spatial differences in the distribution of pigment cells and integumentary appendages. While the pigmentation system has been well characterized, the mechanisms governing the formation of compartments in the skin of wild animals have remained unclear, largely because laboratory models do not display ecologically-relevant colour patterns. We will use a combination of forward genetics, developmental biology, modelling, and imaging to study natural variation in the large colour domains of Estrildid finches and the periodic stripes of Galliform birds. For both phenotypes, we will characterize the organization of the embryonic skin and the mode of patterning (i.e., instructional patterning via external cues vs locally-occurring self-organization) underlying their formation, and identify the molecular factors and developmental processes contributing to their variation. Results from these studies will elucidate the biochemical events and tissue rearrangements orchestrating colour patterning in development and shed light on how these processes shape natural variation in this trait‒ and more generally, in natural patterns.
Summary
Animals display a tremendous diversity of patterns ‒from the colourful designs that adorn their body to repeated segmented appendages. Natural patterns result from the formation of discrete domains within developing tissues through the integration of positional cues by cells that consequently adopt specific fates and produce spatial heterogeneity. How can such developmental processes underlie the apparent complexity and diversity of natural patterns? We propose to address this long-standing question with an innovative experimental design: we will make use of natural variation as a powerful tool to facilitate the identification of patterning molecules and morphogenetic events. We will study colour pattern, a crucial adaptive trait that varies extensively in nature, from large colour domains to periodic designs. In amniotes, colour pattern is formed by spatial differences in the distribution of pigment cells and integumentary appendages. While the pigmentation system has been well characterized, the mechanisms governing the formation of compartments in the skin of wild animals have remained unclear, largely because laboratory models do not display ecologically-relevant colour patterns. We will use a combination of forward genetics, developmental biology, modelling, and imaging to study natural variation in the large colour domains of Estrildid finches and the periodic stripes of Galliform birds. For both phenotypes, we will characterize the organization of the embryonic skin and the mode of patterning (i.e., instructional patterning via external cues vs locally-occurring self-organization) underlying their formation, and identify the molecular factors and developmental processes contributing to their variation. Results from these studies will elucidate the biochemical events and tissue rearrangements orchestrating colour patterning in development and shed light on how these processes shape natural variation in this trait‒ and more generally, in natural patterns.
Max ERC Funding
1 483 144 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym COMPUCELL
Project Autonomous Cellular Computers for Diagnosis
Researcher (PI) JEROME, Cédric Bonnet
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS7, ERC-2014-STG
Summary Early diagnostics based on multiple biomarkers is key in numerous diseases, yet current technologies for multiplexed detection are complicated and expensive. Living cells detect and process various environmental signals in parallel and can self-replicate, presenting an attractive platform for scalable and affordable autonomous diagnostic devices. In this project, I will apply my expertise in synthetic biology, the rational engineering of biological systems, to build cell-based biosensors for multiplexed diagnosis using the non-pathogenic bacterium Bacillus subtilis.
In a first research line, I will conceive a scalable detection machinery by engineering chimeric receptors detecting extracellular biomarkers via sensing domains derived from antibodies. In a second research line, I will implement bio-molecular computing systems operating within and across bacterial cells to perform multiplexed biomarkers analysis. I will deploy in B. subtilis biomolecular logic gates and will engineer specific cell-cell communication systems to perform distributed multicellular computation in a bacterial consortia.
My project is highly interdisciplinary and is at the cross-roads of genetic engineering, structural biology, biophysics, modeling, and clinics. On foundational point of view, I will make several breakthrough contributions to synthetic biology: (i) Advancing engineering frameworks for the Gram-positive model, B. subtilis. (ii) Pushing the limits of custom-ligand detection by engineered cells (iii) Exploring the frontiers of man-made biological computers. On an applied point of view, I plan to deliver a first prototype for the urinary diagnostic of diabetic nephropathy, a major complication of diabetes. Because of the modular design principles applied, my sensing platform will be reusable to diagnose other pathologies as well as for applications requiring custom-detection and bio-molecular computation like targeted therapy, drug delivery, or environmental monitoring.
Summary
Early diagnostics based on multiple biomarkers is key in numerous diseases, yet current technologies for multiplexed detection are complicated and expensive. Living cells detect and process various environmental signals in parallel and can self-replicate, presenting an attractive platform for scalable and affordable autonomous diagnostic devices. In this project, I will apply my expertise in synthetic biology, the rational engineering of biological systems, to build cell-based biosensors for multiplexed diagnosis using the non-pathogenic bacterium Bacillus subtilis.
In a first research line, I will conceive a scalable detection machinery by engineering chimeric receptors detecting extracellular biomarkers via sensing domains derived from antibodies. In a second research line, I will implement bio-molecular computing systems operating within and across bacterial cells to perform multiplexed biomarkers analysis. I will deploy in B. subtilis biomolecular logic gates and will engineer specific cell-cell communication systems to perform distributed multicellular computation in a bacterial consortia.
My project is highly interdisciplinary and is at the cross-roads of genetic engineering, structural biology, biophysics, modeling, and clinics. On foundational point of view, I will make several breakthrough contributions to synthetic biology: (i) Advancing engineering frameworks for the Gram-positive model, B. subtilis. (ii) Pushing the limits of custom-ligand detection by engineered cells (iii) Exploring the frontiers of man-made biological computers. On an applied point of view, I plan to deliver a first prototype for the urinary diagnostic of diabetic nephropathy, a major complication of diabetes. Because of the modular design principles applied, my sensing platform will be reusable to diagnose other pathologies as well as for applications requiring custom-detection and bio-molecular computation like targeted therapy, drug delivery, or environmental monitoring.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym CONSTRAINTS
Project Ecophysiological and biophysical constraints on domestication in crop plants
Researcher (PI) Cyrille (Fabrice) Violle
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), LS9, ERC-2014-STG
Summary A fundamental question in biology is how constraints drive phenotypic changes and the diversification of life. We know little about the role of these constraints on crop domestication, nor how artificial selection can escape them. CONSTRAINTS questions whether crop domestication has shifted ecophysiological and biophysical traits related to resource acquisition, use and partitioning, and how trade-offs between them have constrained domestication and can limit future improvements in both optimal and sub-optimal conditions.
The project is based on three objectives: 1. revealing the existence (or lack) of generic resource-use domestication syndrome in crop science; 2. elucidating ecophysiological and biophysical trade-offs within crop science and delineating the envelope of constraints for artificial selection; 3. examining the shape of ecophysiological and biophysical trade-offs in crop species when grown in sub-optimal environmental conditions. This project will be investigated within and across crop species thanks to a core panel of 12 studied species (maize, sunflower, Japanese rice, sorghum, durum wheat, bread wheat, alfalfa, orchardgrass, silvergrass, pea, colza, vine) for which data and collections (ca. 1,300 genotypes total) are already available to the PI, and additional high throughput phenotyping using automatons. Additional species will be used for specific tasks: (i) a panel of 30 species for a comparative analysis of crop species and their wild progenitors; (ii) 400 worldwide accessions of Arabidopsis thaliana for a genome-wide association study of resource-use traits. Collectively, we will use a multiple-tool approach by using: field measurement, high-throughput phenotyping, common-garden experiment, comparative analysis using databases, modelling, genomics.
The ground-breaking nature of the project holds in the nature of the questions asked and in the unique opportunity to transfer knowledge from ecology and evolutionary biology to crop species.
Summary
A fundamental question in biology is how constraints drive phenotypic changes and the diversification of life. We know little about the role of these constraints on crop domestication, nor how artificial selection can escape them. CONSTRAINTS questions whether crop domestication has shifted ecophysiological and biophysical traits related to resource acquisition, use and partitioning, and how trade-offs between them have constrained domestication and can limit future improvements in both optimal and sub-optimal conditions.
The project is based on three objectives: 1. revealing the existence (or lack) of generic resource-use domestication syndrome in crop science; 2. elucidating ecophysiological and biophysical trade-offs within crop science and delineating the envelope of constraints for artificial selection; 3. examining the shape of ecophysiological and biophysical trade-offs in crop species when grown in sub-optimal environmental conditions. This project will be investigated within and across crop species thanks to a core panel of 12 studied species (maize, sunflower, Japanese rice, sorghum, durum wheat, bread wheat, alfalfa, orchardgrass, silvergrass, pea, colza, vine) for which data and collections (ca. 1,300 genotypes total) are already available to the PI, and additional high throughput phenotyping using automatons. Additional species will be used for specific tasks: (i) a panel of 30 species for a comparative analysis of crop species and their wild progenitors; (ii) 400 worldwide accessions of Arabidopsis thaliana for a genome-wide association study of resource-use traits. Collectively, we will use a multiple-tool approach by using: field measurement, high-throughput phenotyping, common-garden experiment, comparative analysis using databases, modelling, genomics.
The ground-breaking nature of the project holds in the nature of the questions asked and in the unique opportunity to transfer knowledge from ecology and evolutionary biology to crop species.
Max ERC Funding
1 499 979 €
Duration
Start date: 2015-06-01, End date: 2020-05-31
Project acronym ConvergeAnt
Project An Integrative Approach to Understanding Convergent Evolution in Ant-eating Mammals
Researcher (PI) Frederic DELSUC
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS8, ERC-2015-CoG
Summary Despite its widespread occurrence across the tree of life, many questions still remain unanswered concerning the fascinating phenomenon of convergent evolution. Ant-eating mammals constitute a textbook example of morphological convergence with at least five independent origins in placentals (armadillos, anteaters, aardvarks, pangolins, and aardwolves). The large extent of convergent morphological evolution, the importance of molecular convergence, and the role of the host microbiome in diet adaptation are currently gaining acceptance. However, large-scale comparative studies combining morphology, host genomics, and metagenomics of the associated microbiome are still lacking. In the ConvergeAnt project, we propose taking advantage of the unique set of convergently evolved characters associated with the ant-eating diet to investigate the molecular mechanisms underlying phenotypical adaptation. By using state-of-the art phenotyping methods based on X-ray micro-computed tomography and Illumina sequencing technologies we will combine morphometric, genomic, and metagenomic approaches to evaluate the extent of convergent evolution in the skull of myrmecophagous placentals, in their genomes, and in their associated oral and gut microbiomes. With this ambitious research proposal, we aim at providing answers to longstanding but fundamental evolutionary questions pertaining to the mechanisms of convergent evolution. The ConvergeAnt project will be the first of its kind to apply such an integrative approach to investigate the complex interplay between the mammalian genome and its associated microbiome in a classical case of adaptive convergence driven by a highly specialized diet.
Summary
Despite its widespread occurrence across the tree of life, many questions still remain unanswered concerning the fascinating phenomenon of convergent evolution. Ant-eating mammals constitute a textbook example of morphological convergence with at least five independent origins in placentals (armadillos, anteaters, aardvarks, pangolins, and aardwolves). The large extent of convergent morphological evolution, the importance of molecular convergence, and the role of the host microbiome in diet adaptation are currently gaining acceptance. However, large-scale comparative studies combining morphology, host genomics, and metagenomics of the associated microbiome are still lacking. In the ConvergeAnt project, we propose taking advantage of the unique set of convergently evolved characters associated with the ant-eating diet to investigate the molecular mechanisms underlying phenotypical adaptation. By using state-of-the art phenotyping methods based on X-ray micro-computed tomography and Illumina sequencing technologies we will combine morphometric, genomic, and metagenomic approaches to evaluate the extent of convergent evolution in the skull of myrmecophagous placentals, in their genomes, and in their associated oral and gut microbiomes. With this ambitious research proposal, we aim at providing answers to longstanding but fundamental evolutionary questions pertaining to the mechanisms of convergent evolution. The ConvergeAnt project will be the first of its kind to apply such an integrative approach to investigate the complex interplay between the mammalian genome and its associated microbiome in a classical case of adaptive convergence driven by a highly specialized diet.
Max ERC Funding
1 880 570 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym COOPETITION
Project Cooperation and competition in vertical relations: the business strategies and industry oversight of supply agreements and buying patterns
Researcher (PI) Patrick Rey
Host Institution (HI) FONDATION JEAN-JACQUES LAFFONT,TOULOUSE SCIENCES ECONOMIQUES
Call Details Advanced Grant (AdG), SH1, ERC-2013-ADG
Summary "The application proposes to revisit the economics of cooperation and competition in industry vertical chains and develop new tools for industrial organization (IO). Modern IO theory treats firms as unitary, profit-maximizing entities, characterized by well-identified perimeters of activity, and clearly identified either as competitors or as complementors. Yet in practice:
- Industry structures are increasingly complex: Firms distribute for example their activities among partners across the globe, and moved to multiple, interlocking relationships.
- Firms competing for customers or suppliers are also cooperating in other dimensions, e.g., by setting-up common platforms, or by adopting joint common rules within which to compete.
- Supplier -customer relations often involve transaction costs other than pure search costs: adoption costs, learning or shopping costs, or expensive strategies to protect sensitive information.
Understanding the interplay between competition and cooperation is key to designing business strategies, but it has also implications for industry oversight: When should cooperation among competitors be limited or encouraged? Over which dimensions? This application proposes to cover three topics:
1. Allocation of tasks and the choice of partners.
2. Multilateral interlocking relations.
3. Cooperation and competition.
4. Transaction costs, buying patterns and business strategies
While the project falls primarily in the field of applied theory, some of the developments require new tools and interaction with game theorists. Furthermore, empirical validation will require the use of structural econometric modelling (based in particular on consumer panel data) and laboratory experiments. The project has also an interdisciplinary flavour and will benefit from work of and interactions with legal scholars and marketing experts."
Summary
"The application proposes to revisit the economics of cooperation and competition in industry vertical chains and develop new tools for industrial organization (IO). Modern IO theory treats firms as unitary, profit-maximizing entities, characterized by well-identified perimeters of activity, and clearly identified either as competitors or as complementors. Yet in practice:
- Industry structures are increasingly complex: Firms distribute for example their activities among partners across the globe, and moved to multiple, interlocking relationships.
- Firms competing for customers or suppliers are also cooperating in other dimensions, e.g., by setting-up common platforms, or by adopting joint common rules within which to compete.
- Supplier -customer relations often involve transaction costs other than pure search costs: adoption costs, learning or shopping costs, or expensive strategies to protect sensitive information.
Understanding the interplay between competition and cooperation is key to designing business strategies, but it has also implications for industry oversight: When should cooperation among competitors be limited or encouraged? Over which dimensions? This application proposes to cover three topics:
1. Allocation of tasks and the choice of partners.
2. Multilateral interlocking relations.
3. Cooperation and competition.
4. Transaction costs, buying patterns and business strategies
While the project falls primarily in the field of applied theory, some of the developments require new tools and interaction with game theorists. Furthermore, empirical validation will require the use of structural econometric modelling (based in particular on consumer panel data) and laboratory experiments. The project has also an interdisciplinary flavour and will benefit from work of and interactions with legal scholars and marketing experts."
Max ERC Funding
2 068 920 €
Duration
Start date: 2013-12-01, End date: 2018-11-30
Project acronym CORTEXSELFCONTROL
Project Self-Modulating Neurons in the Cerebral Cortex: From Molecular Mechanisms to Cortical Network Activities
Researcher (PI) Alberto Bacci
Host Institution (HI) INSTITUT DU CERVEAU ET DE LA MOELLE EPINIERE
Call Details Starting Grant (StG), LS4, ERC-2007-StG
Summary In the mammalian brain, the neocortex is the site where sensory information is integrated into complex cognitive functions. This is accomplished by the activity of both principal glutamatergic neurons and locally-projecting inhibitory GABAergic interneurons, interconnected in complex networks. Inhibitory neurons play several key roles in neocortical function. For example, they shape sensory receptive fields and drive several high frequency network oscillations. On the other hand, defects in their function can lead to devastating diseases, such as epilepsy and schizophrenia. Cortical interneurons represent a highly heterogeneous cell population. Understanding the specific role of each interneuron subtype within cortical microcircuits is still a crucial open question. We have examined properties of two major functional interneuron subclasses in neocortical layer V: fast-spiking (FS) and low-threshold spiking (LTS) cells. Our previous data indicate that each group expresses a novel form of self inhibition, namely autaptic inhibitory transmission in FS cells and an endocannabinoid-mediated slow self inhibition in LTS interneurons. In this proposal we will address three major questions relevant to self-inhibition of neocortical interneurons: 1) What is the role of FS cell autapses in coordinating fast network synchrony? 2) What are the molecular mechanisms underlying autaptic asynchronous release, prolonging FS cell self-inhibition by several seconds, and what is its relevance during physiological and pathological network activities? 3) What are the induction mechanisms, the molecular players involved and the functional roles within cortical microcircuits of the endocannabinoid-mediated long-lasting self-inhibition in LTS interneurons? Results of these experiments will lead to a better understanding of GABAergic interneuron regulation of neocortical excitability, relevant to both normal and pathological cortical function.
Summary
In the mammalian brain, the neocortex is the site where sensory information is integrated into complex cognitive functions. This is accomplished by the activity of both principal glutamatergic neurons and locally-projecting inhibitory GABAergic interneurons, interconnected in complex networks. Inhibitory neurons play several key roles in neocortical function. For example, they shape sensory receptive fields and drive several high frequency network oscillations. On the other hand, defects in their function can lead to devastating diseases, such as epilepsy and schizophrenia. Cortical interneurons represent a highly heterogeneous cell population. Understanding the specific role of each interneuron subtype within cortical microcircuits is still a crucial open question. We have examined properties of two major functional interneuron subclasses in neocortical layer V: fast-spiking (FS) and low-threshold spiking (LTS) cells. Our previous data indicate that each group expresses a novel form of self inhibition, namely autaptic inhibitory transmission in FS cells and an endocannabinoid-mediated slow self inhibition in LTS interneurons. In this proposal we will address three major questions relevant to self-inhibition of neocortical interneurons: 1) What is the role of FS cell autapses in coordinating fast network synchrony? 2) What are the molecular mechanisms underlying autaptic asynchronous release, prolonging FS cell self-inhibition by several seconds, and what is its relevance during physiological and pathological network activities? 3) What are the induction mechanisms, the molecular players involved and the functional roles within cortical microcircuits of the endocannabinoid-mediated long-lasting self-inhibition in LTS interneurons? Results of these experiments will lead to a better understanding of GABAergic interneuron regulation of neocortical excitability, relevant to both normal and pathological cortical function.
Max ERC Funding
996 000 €
Duration
Start date: 2008-10-01, End date: 2014-03-31
Project acronym CorticALS
Project Amyotrophic Lateral Sclerosis from a cortical perspective: towards alternative therapeutic strategies
Researcher (PI) Caroline Danielle Aline Rouaux
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS5, ERC-2014-STG
Summary Amyotrophic Lateral Sclerosis (ALS) is the most common adult-onset neurodegenerative disease of the motor system, with a prevalence of 2-3/100 000. In spite of intensive research efforts, ALS remains an incurable disease and presents with a very severe prognosis, leading to patient death within 2 to 5 years following diagnosis.
At the cellular level, ALS is characterized by the combined degeneration of both upper motor neurons (UMN, or corticospinal motor neurons) whose cell bodies are located in the cerebral cortex, and that extend axons to the medulla and spinal cord, and lower motor neurons (LMN, or spinal motor neurons) whose cell bodies are located in the medulla and spinal cord, and that connect to the skeletal muscles. This dual impairment allows to discriminate ALS from other, less severe diseases affecting either UMN or LMN. Despite this precise clinical description, it is striking to note that preclinical studies have so far mostly concentrated on LMN, leaving aside the role of UMN in ALS.
This project aims at shedding light on the contribution of the dysfunction and/or the loss of UMN in ALS, in order to design and test new therapeutic strategies based on the protection and/or the replacement of this exact neuronal type. This innovative question has never been directly asked so far. Our working hypothesis is that specific neurodegeneration of UMN, in the course of ALS, does not represent an isolated side effect, but rather actively contributes to the onset and progression of the disease. Based on the discovery of new molecular players, and the development of alternative therapies, this original thematic has the ambition to provide clinicians and patients with new answers and new therapeutic assets.
Summary
Amyotrophic Lateral Sclerosis (ALS) is the most common adult-onset neurodegenerative disease of the motor system, with a prevalence of 2-3/100 000. In spite of intensive research efforts, ALS remains an incurable disease and presents with a very severe prognosis, leading to patient death within 2 to 5 years following diagnosis.
At the cellular level, ALS is characterized by the combined degeneration of both upper motor neurons (UMN, or corticospinal motor neurons) whose cell bodies are located in the cerebral cortex, and that extend axons to the medulla and spinal cord, and lower motor neurons (LMN, or spinal motor neurons) whose cell bodies are located in the medulla and spinal cord, and that connect to the skeletal muscles. This dual impairment allows to discriminate ALS from other, less severe diseases affecting either UMN or LMN. Despite this precise clinical description, it is striking to note that preclinical studies have so far mostly concentrated on LMN, leaving aside the role of UMN in ALS.
This project aims at shedding light on the contribution of the dysfunction and/or the loss of UMN in ALS, in order to design and test new therapeutic strategies based on the protection and/or the replacement of this exact neuronal type. This innovative question has never been directly asked so far. Our working hypothesis is that specific neurodegeneration of UMN, in the course of ALS, does not represent an isolated side effect, but rather actively contributes to the onset and progression of the disease. Based on the discovery of new molecular players, and the development of alternative therapies, this original thematic has the ambition to provide clinicians and patients with new answers and new therapeutic assets.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-04-01, End date: 2021-03-31
Project acronym CoSpaDD
Project Competition for Space in Development and Diseases
Researcher (PI) Romain LEVAYER
Host Institution (HI) INSTITUT PASTEUR
Call Details Starting Grant (StG), LS3, ERC-2017-STG
Summary Developing tissues have a remarkable plasticity illustrated by their capacity to regenerate and form normal organs despite strong perturbations. This requires the adjustment of single cell behaviour to their neighbours and to tissue scale parameters. The modulation of cell growth and proliferation was suggested to be driven by mechanical inputs, however the mechanisms adjusting cell death are not well known. Recently it was shown that epithelial cells could be eliminated by spontaneous live-cell delamination following an increase of cell density. Studying cell delamination in the midline region of the Drosophila pupal notum, we confirmed that local tissue crowding is necessary and sufficient to drive cell elimination and found that Caspase 3 activation precedes and is required for cell delamination. This suggested that a yet unknown pathway is responsible for crowding sensing and activation of caspase, which does not involve already known mechanical sensing pathways. Moreover, we showed that fast growing clones in the notum could induce neighbouring cell elimination through crowding-induced death. This suggested that crowding-induced death could promote tissue invasion by pretumoural cells.
Here we will combine genetics, quantitative live imaging, statistics, laser perturbations and modelling to study crowding-induced death in Drosophila in order to: 1) find single cell deformations responsible for caspase activation; 2) find new pathways responsible for density sensing and apoptosis induction; 3) test their contribution to adult tissue homeostasis, morphogenesis and cell elimination coordination; 4) study the role of crowding induced death during competition between different cell types and tissue invasion 5) Explore theoretically the conditions required for efficient space competition between two cell populations.
This project will provide essential information for the understanding of epithelial homeostasis, mechanotransduction and tissue invasion by tumoural cells
Summary
Developing tissues have a remarkable plasticity illustrated by their capacity to regenerate and form normal organs despite strong perturbations. This requires the adjustment of single cell behaviour to their neighbours and to tissue scale parameters. The modulation of cell growth and proliferation was suggested to be driven by mechanical inputs, however the mechanisms adjusting cell death are not well known. Recently it was shown that epithelial cells could be eliminated by spontaneous live-cell delamination following an increase of cell density. Studying cell delamination in the midline region of the Drosophila pupal notum, we confirmed that local tissue crowding is necessary and sufficient to drive cell elimination and found that Caspase 3 activation precedes and is required for cell delamination. This suggested that a yet unknown pathway is responsible for crowding sensing and activation of caspase, which does not involve already known mechanical sensing pathways. Moreover, we showed that fast growing clones in the notum could induce neighbouring cell elimination through crowding-induced death. This suggested that crowding-induced death could promote tissue invasion by pretumoural cells.
Here we will combine genetics, quantitative live imaging, statistics, laser perturbations and modelling to study crowding-induced death in Drosophila in order to: 1) find single cell deformations responsible for caspase activation; 2) find new pathways responsible for density sensing and apoptosis induction; 3) test their contribution to adult tissue homeostasis, morphogenesis and cell elimination coordination; 4) study the role of crowding induced death during competition between different cell types and tissue invasion 5) Explore theoretically the conditions required for efficient space competition between two cell populations.
This project will provide essential information for the understanding of epithelial homeostasis, mechanotransduction and tissue invasion by tumoural cells
Max ERC Funding
1 489 147 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym CrIC
Project Molecular basis of the cross-talk between chronic inflammation and cancer
Researcher (PI) Nadine Laguette
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), LS6, ERC-2014-STG
Summary Cancer related inflammation (CRI) is a well-established hallmark of cancer. We recently demonstrated that the DNA damage repair SLX4 complex suppresses spontaneous and human immunodeficiency virus (HIV)-dependent pro-inflammatory cytokine production, revealing a role for this DNA repair complex in controlling innate immune responses. Bi-allelic mutations in SLX4 are involved in the onset of Fanconi Anemia (FA), a syndrome characterized, besides heightened cancer susceptibility, by severe defects of the immune system, resulting from increased pro-inflammatory cytokine levels and progressive bone marrow failure. Within this proposal, using SLX4-deficiency as a working model, I aim at investigating the molecular process underlying CRI. Based on our previous observation that the SLX4 complex binds to HIV-derived reverse-transcripts and promotes their degradation, my working hypothesis is that CRI results from the accumulation of endogenous pathological nucleic acids that are recognized by the innate immune system in the absence of SLX4. The present project should unveil the relationship between repression of pro-inflammatory cytokine production by proteins involved in DNA repair, DNA damage, and CRI, thereby opening unforeseen perspectives in the treatment of cancer patients.
Summary
Cancer related inflammation (CRI) is a well-established hallmark of cancer. We recently demonstrated that the DNA damage repair SLX4 complex suppresses spontaneous and human immunodeficiency virus (HIV)-dependent pro-inflammatory cytokine production, revealing a role for this DNA repair complex in controlling innate immune responses. Bi-allelic mutations in SLX4 are involved in the onset of Fanconi Anemia (FA), a syndrome characterized, besides heightened cancer susceptibility, by severe defects of the immune system, resulting from increased pro-inflammatory cytokine levels and progressive bone marrow failure. Within this proposal, using SLX4-deficiency as a working model, I aim at investigating the molecular process underlying CRI. Based on our previous observation that the SLX4 complex binds to HIV-derived reverse-transcripts and promotes their degradation, my working hypothesis is that CRI results from the accumulation of endogenous pathological nucleic acids that are recognized by the innate immune system in the absence of SLX4. The present project should unveil the relationship between repression of pro-inflammatory cytokine production by proteins involved in DNA repair, DNA damage, and CRI, thereby opening unforeseen perspectives in the treatment of cancer patients.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-04-01, End date: 2020-03-31
Project acronym CRISPAIR
Project Study of the interplay between CRISPR interference and DNA repair pathways towards the development of novel CRISPR tools
Researcher (PI) David Bikard
Host Institution (HI) INSTITUT PASTEUR
Call Details Starting Grant (StG), LS1, ERC-2015-STG
Summary CRISPR-Cas loci are the adaptive immune system of archaea and bacteria. They can capture pieces of invading DNA and use this information to degrade target DNA through the action of RNA-guided nucleases. The consequences of DNA cleavage by Cas nucleases, i.e. how breaks are processed and whether they can be repaired, remains to be investigated. A better understanding of the interplay between DNA repair and CRISPR-Cas is critical both to shed light on the evolution and biology of these fascinating systems and for the development of biotechnological tools based on Cas nucleases. CRISPR systems have indeed become a popular tool to edit Eukaryotic genomes. The strategies employed take advantage of different DNA repair pathways to introduce mutations upon DNA cleavage. In bacteria however, the introduction of breaks by Cas nucleases in the chromosome has been described to kill the cell. Preliminary data indicates that this might not always be the case and that some DNA repair pathways could compete with CRISPR immunity allowing cells to survive. Using a combination of bioinformatics and genetics approaches we will investigate the interplay between CRISPR and DNA repair in bacteria with a particular focus on the widely used CRISPR-Cas9 system. The knowledge gained from this study will then help us develop novel tools for bacterial genome engineering. In particular we will introduce a NHEJ pathway in E.coli making it possible to perform CRISPR knockout screens. Finally using CRISPR libraries and multiplexed targeting, we will generate for the first time all combinations of pair-wise gene knockouts in an organism, a task that for now remains elusive, even for large consortiums and with the use of automation. This will enable to decipher genome-scale genetic interaction networks, an important step for our understanding of bacteria as a system.
Summary
CRISPR-Cas loci are the adaptive immune system of archaea and bacteria. They can capture pieces of invading DNA and use this information to degrade target DNA through the action of RNA-guided nucleases. The consequences of DNA cleavage by Cas nucleases, i.e. how breaks are processed and whether they can be repaired, remains to be investigated. A better understanding of the interplay between DNA repair and CRISPR-Cas is critical both to shed light on the evolution and biology of these fascinating systems and for the development of biotechnological tools based on Cas nucleases. CRISPR systems have indeed become a popular tool to edit Eukaryotic genomes. The strategies employed take advantage of different DNA repair pathways to introduce mutations upon DNA cleavage. In bacteria however, the introduction of breaks by Cas nucleases in the chromosome has been described to kill the cell. Preliminary data indicates that this might not always be the case and that some DNA repair pathways could compete with CRISPR immunity allowing cells to survive. Using a combination of bioinformatics and genetics approaches we will investigate the interplay between CRISPR and DNA repair in bacteria with a particular focus on the widely used CRISPR-Cas9 system. The knowledge gained from this study will then help us develop novel tools for bacterial genome engineering. In particular we will introduce a NHEJ pathway in E.coli making it possible to perform CRISPR knockout screens. Finally using CRISPR libraries and multiplexed targeting, we will generate for the first time all combinations of pair-wise gene knockouts in an organism, a task that for now remains elusive, even for large consortiums and with the use of automation. This will enable to decipher genome-scale genetic interaction networks, an important step for our understanding of bacteria as a system.
Max ERC Funding
1 499 763 €
Duration
Start date: 2016-03-01, End date: 2021-02-28
Project acronym CytoBacLysis
Project Deciphering cytosolic antibacterial immunity: from triggering bacteriolysis to Aim2 inflammasome activation
Researcher (PI) Thomas Henry
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS6, ERC-2012-StG_20111109
Summary Bacteria replicating within host cells either multiply in membrane-bound compartment or escape into the host cytosol. The host cytosol has long been considered as a safe haven for bacteria. However, the host cytosol is armed with an array of innate immune receptors detecting cytosolic invasion. Furthermore, the macrophage cytosol displays a bacteriolytic activity, which is inducible by IFN. Surprisingly, the molecular mechanisms of this innate immune effector response are still largely uncharacterized. A ubiquitously expressed antimicrobial peptide, ubiquicidin has been described in the macrophage cytosol. Its relevance, its connection with macrophage-specific bacteriolytic activity and with IFN, remain to be deciphered. While cytosol-adapted bacteria are largely resistant to the bactericidal activity of the macrophage, lysis of a single bacterium triggers activation of the Aim2 inflammasome. Cytosolic bacteriolysis is thus key to orchestrate inflammasome-mediated innate immune responses. We propose here to characterize the bacteriolytic effector mechanisms, the regulation of this response and of the Aim2 inflammasome by IFN in infected macrophages. We will use two complementary bacterial models: F. tularensis, a cytosol-adapted bacterium and S. typhimurium sifA mutant, a bacterium lysed in the macrophage cytosol. We will develop three synergistic approaches:
i) the generation of novel tools to monitor cytosolic bacteriolysis
ii) hypothesis-driven investigations on the antimicrobial activity of the macrophage cytosol focusing on ubiquicidin to uncover the mechanisms of processing and targeting of this antimicrobial peptide
iii) screening of IFN-inducible genes to identify novel players involved in the cytosolic bacteriolytic activity and in inflammasome regulation.
We believe this project should reveal the innate immune effector mechanisms of the macrophage cytosol i.e. how the macrophage kills cytosolic bacteria and orchestrates further immune responses.
Summary
Bacteria replicating within host cells either multiply in membrane-bound compartment or escape into the host cytosol. The host cytosol has long been considered as a safe haven for bacteria. However, the host cytosol is armed with an array of innate immune receptors detecting cytosolic invasion. Furthermore, the macrophage cytosol displays a bacteriolytic activity, which is inducible by IFN. Surprisingly, the molecular mechanisms of this innate immune effector response are still largely uncharacterized. A ubiquitously expressed antimicrobial peptide, ubiquicidin has been described in the macrophage cytosol. Its relevance, its connection with macrophage-specific bacteriolytic activity and with IFN, remain to be deciphered. While cytosol-adapted bacteria are largely resistant to the bactericidal activity of the macrophage, lysis of a single bacterium triggers activation of the Aim2 inflammasome. Cytosolic bacteriolysis is thus key to orchestrate inflammasome-mediated innate immune responses. We propose here to characterize the bacteriolytic effector mechanisms, the regulation of this response and of the Aim2 inflammasome by IFN in infected macrophages. We will use two complementary bacterial models: F. tularensis, a cytosol-adapted bacterium and S. typhimurium sifA mutant, a bacterium lysed in the macrophage cytosol. We will develop three synergistic approaches:
i) the generation of novel tools to monitor cytosolic bacteriolysis
ii) hypothesis-driven investigations on the antimicrobial activity of the macrophage cytosol focusing on ubiquicidin to uncover the mechanisms of processing and targeting of this antimicrobial peptide
iii) screening of IFN-inducible genes to identify novel players involved in the cytosolic bacteriolytic activity and in inflammasome regulation.
We believe this project should reveal the innate immune effector mechanisms of the macrophage cytosol i.e. how the macrophage kills cytosolic bacteria and orchestrates further immune responses.
Max ERC Funding
1 404 688 €
Duration
Start date: 2012-11-01, End date: 2018-10-31
Project acronym Damocles
Project Modelling brain aneurysm to elucidate the role of platelets
Researcher (PI) Yacine BOULAFTALI
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS4, ERC-2017-STG
Summary In the European Union, 15 million people have an unruptured intracranial aneurysm (IA) that may rupture one day and lead to subarachnoid haemorrhage (SAH). The IA rupture event is ominous and lingers as a clinical quandary. No safe and effective non-invasive therapies have, as of yet, been identified and implemented in clinical practice mainly because of a lack of knowledge of the underlying mechanisms. Increasing evidence points to inflammation as one of the leading factors in the pathogenesis of IA. Intrasaccular clot formation is a common feature of IA occurring unruptured and ruptured IA. In addition to forming clots, activated platelets support leukocyte recruitment. Interestingly, platelets also prevent local hemorrhage in inflammatory situations independently of their ability to form a platelet plug.
We hypothesize that the role of platelet may evolve throughout the development of IA: initially playing a protective role of in the maintenance of vascular integrity in response to inflammation and contributing later to intrasaccular thrombus formation. What are the platelet signaling pathways and responses involved and to what extent do they contribute to the disease and the rupture event?
To answer these questions, we designed an interdisciplinary proposal, which gathers biophysical, pharmacological, and in-vivo approaches, with the following objectives: I) To investigate platelet functions from patients diagnosed with intracranial aneurysm at the sites of aneurysm sac. II) To delineate platelet mechanisms and responses in a cutting-edge technology of a 3D reconstruction of IA that will take into account the hemodynamic shear stress. III) To test in a preclinical mouse model of IA efficient anti-platelet therapies and define a therapeutic window to intervene on platelet activation. The proposed project will yield new insights in IA disease and in life science, from cell biology to the discovery of potential new targets in cardiovascular medicine.
Summary
In the European Union, 15 million people have an unruptured intracranial aneurysm (IA) that may rupture one day and lead to subarachnoid haemorrhage (SAH). The IA rupture event is ominous and lingers as a clinical quandary. No safe and effective non-invasive therapies have, as of yet, been identified and implemented in clinical practice mainly because of a lack of knowledge of the underlying mechanisms. Increasing evidence points to inflammation as one of the leading factors in the pathogenesis of IA. Intrasaccular clot formation is a common feature of IA occurring unruptured and ruptured IA. In addition to forming clots, activated platelets support leukocyte recruitment. Interestingly, platelets also prevent local hemorrhage in inflammatory situations independently of their ability to form a platelet plug.
We hypothesize that the role of platelet may evolve throughout the development of IA: initially playing a protective role of in the maintenance of vascular integrity in response to inflammation and contributing later to intrasaccular thrombus formation. What are the platelet signaling pathways and responses involved and to what extent do they contribute to the disease and the rupture event?
To answer these questions, we designed an interdisciplinary proposal, which gathers biophysical, pharmacological, and in-vivo approaches, with the following objectives: I) To investigate platelet functions from patients diagnosed with intracranial aneurysm at the sites of aneurysm sac. II) To delineate platelet mechanisms and responses in a cutting-edge technology of a 3D reconstruction of IA that will take into account the hemodynamic shear stress. III) To test in a preclinical mouse model of IA efficient anti-platelet therapies and define a therapeutic window to intervene on platelet activation. The proposed project will yield new insights in IA disease and in life science, from cell biology to the discovery of potential new targets in cardiovascular medicine.
Max ERC Funding
1 498 618 €
Duration
Start date: 2018-06-01, End date: 2023-05-31
Project acronym DARK
Project Dark matter of the human transcriptome: Functional study of the antisense Long Noncoding RNAs and Molecular Mechanisms of Action
Researcher (PI) Antonin Morillon
Host Institution (HI) INSTITUT CURIE
Call Details Consolidator Grant (CoG), LS2, ERC-2013-CoG
Summary 98% of the human genome is non-protein coding raising the question of the role of the dark matter of the genome. It is now admitted that pervasive transcription generates thousands of noncoding transcripts that regulate gene expression and have broad impacts on development and disease. Among the long non coding (lnc)RNAs, antisense transcripts have been poorly studied despite their putative regulatory importance. Several functional examples include X-chromosome inactivation, maintenance of pluripotency and transcriptional regulation. However, no systematic study has yet addressed the comprehensive functional description of human antisense ncRNA, mainly because of technological issues and their low abundance. Indeed, in budding yeast S. cerevisiae, our group showed the existence of an entire class of antisense regulatory lncRNA extremely sensitive to RNA decay pathways, impinging their study so far. The roles for yeast antisense lncRNAs in shaping the epigenome raises important questions: What are the molecular and biochemical mechanisms by which antisense lncRNAs carry out their functions and are they functionally conserved in human cells? We propose that the dark side of the non-coding genome is another layer of gene regulation complexity that needs to be deciphered.
With this proposal, we aim to draw the first exhaustive catalog of human antisense lncRNA in various cell types and tissues using up to date High throughput technologies and bioinformatics pipelines. Second, we propose to determine the functional role of antisense lncRNA on genome expression and stability in the context of cellular stress and cancer. We anticipate that powerful and modern genetic tools such DNA-mediated gene inactivation (ASO) and TALEN approaches will allow precise antisense genes manipulation never achieved so far. Our project is strongly supported by preliminary data indicating an unexpected large number of hidden antisense lncRNA in human cells controlled by RNA decay pathways.
Summary
98% of the human genome is non-protein coding raising the question of the role of the dark matter of the genome. It is now admitted that pervasive transcription generates thousands of noncoding transcripts that regulate gene expression and have broad impacts on development and disease. Among the long non coding (lnc)RNAs, antisense transcripts have been poorly studied despite their putative regulatory importance. Several functional examples include X-chromosome inactivation, maintenance of pluripotency and transcriptional regulation. However, no systematic study has yet addressed the comprehensive functional description of human antisense ncRNA, mainly because of technological issues and their low abundance. Indeed, in budding yeast S. cerevisiae, our group showed the existence of an entire class of antisense regulatory lncRNA extremely sensitive to RNA decay pathways, impinging their study so far. The roles for yeast antisense lncRNAs in shaping the epigenome raises important questions: What are the molecular and biochemical mechanisms by which antisense lncRNAs carry out their functions and are they functionally conserved in human cells? We propose that the dark side of the non-coding genome is another layer of gene regulation complexity that needs to be deciphered.
With this proposal, we aim to draw the first exhaustive catalog of human antisense lncRNA in various cell types and tissues using up to date High throughput technologies and bioinformatics pipelines. Second, we propose to determine the functional role of antisense lncRNA on genome expression and stability in the context of cellular stress and cancer. We anticipate that powerful and modern genetic tools such DNA-mediated gene inactivation (ASO) and TALEN approaches will allow precise antisense genes manipulation never achieved so far. Our project is strongly supported by preliminary data indicating an unexpected large number of hidden antisense lncRNA in human cells controlled by RNA decay pathways.
Max ERC Funding
1 998 884 €
Duration
Start date: 2014-12-01, End date: 2019-11-30
Project acronym DCBIOX
Project Phagosome functions and antigen cross presentation in primary dendritic cells
Researcher (PI) Sebastian Amigorena
Host Institution (HI) INSTITUT CURIE
Call Details Advanced Grant (AdG), LS6, ERC-2013-ADG
Summary T cell cross priming (the initiation of CD8+ T cell responses to antigen that are not expressed by dendritic cells, DCs) requires the phagocytosis of antigens by DCs and their presentation on MHC class I molecules, a process referred to as “cross presentation”. Here, we propose a series of integrated approaches to address the most fundamental mechanisms of cross presentation and explore the use of this process for translational purposes in human cancer.
This proposal will pursue three main objectives:
1) To analyze the mechanisms of control of antigen cross presentation and phagocytic functions in DCs. We will use genome wide screens and conditional KO mice, associated to quantitative assays for phagosomal functions and cross presentation, to investigate the molecular mechanisms of cross presentation in vitro and in vivo.
2) To study the epigenetic programing of cross presentation during the ontogeny of mouse DC subpopulations. We will define a “cross presentation gene signature” that will be validated by systematic gene silencing in vitro and we will analyze the epigenetic basis of control of cross presentation-related genes developing DCs.
3) To investigate the regulation of cross presentation in human primary DCs and to develop translational approaches in cancer. We will study cross presentation and phagosome functions in primary human DC subpopulations and its regulation by innate receptors for the development of original immunomodulation and vaccination strategies. We will explore the use of DCs cross presentation abilities in solid tumor infiltrating DCs and their use for prognosis in cancer.
The results of this project will unravel fundamental mechanisms of phagocytosis and its control by innate signals in mice and humans. The proposal also aims at defining new possible strategies for cancer treatment and prognosis.
Summary
T cell cross priming (the initiation of CD8+ T cell responses to antigen that are not expressed by dendritic cells, DCs) requires the phagocytosis of antigens by DCs and their presentation on MHC class I molecules, a process referred to as “cross presentation”. Here, we propose a series of integrated approaches to address the most fundamental mechanisms of cross presentation and explore the use of this process for translational purposes in human cancer.
This proposal will pursue three main objectives:
1) To analyze the mechanisms of control of antigen cross presentation and phagocytic functions in DCs. We will use genome wide screens and conditional KO mice, associated to quantitative assays for phagosomal functions and cross presentation, to investigate the molecular mechanisms of cross presentation in vitro and in vivo.
2) To study the epigenetic programing of cross presentation during the ontogeny of mouse DC subpopulations. We will define a “cross presentation gene signature” that will be validated by systematic gene silencing in vitro and we will analyze the epigenetic basis of control of cross presentation-related genes developing DCs.
3) To investigate the regulation of cross presentation in human primary DCs and to develop translational approaches in cancer. We will study cross presentation and phagosome functions in primary human DC subpopulations and its regulation by innate receptors for the development of original immunomodulation and vaccination strategies. We will explore the use of DCs cross presentation abilities in solid tumor infiltrating DCs and their use for prognosis in cancer.
The results of this project will unravel fundamental mechanisms of phagocytosis and its control by innate signals in mice and humans. The proposal also aims at defining new possible strategies for cancer treatment and prognosis.
Max ERC Funding
2 500 000 €
Duration
Start date: 2014-09-01, End date: 2019-08-31
Project acronym DECODE
Project Decoding the complexity of quantitative natural variation in Arabidopsis thaliana
Researcher (PI) Olivier Loudet
Host Institution (HI) INSTITUT NATIONAL DE LA RECHERCHE AGRONOMIQUE
Call Details Starting Grant (StG), LS2, ERC-2009-StG
Summary Following a long history of quantitative genetics in crop plants, it now becomes feasible to use naturally-occuring variation contained in Arabidopsis thaliana accessions (lines isolated from natural populations) as the source of quantitative genomics approaches, designed to map QTLs and resolve them at the gene level. Apart from being able to exploit in multiple genetic backgrounds allelic variation that cannot be easily generated by conventional mutagenesis, the (relatively few) success of the QTL studies has often been because of the use of quantitative phenotyping, as opposed to the qualitative gauges used in typical mutant screens. Among the various genetic mechanisms responsible for natural variation that have just started to be revealed, cis-acting regulation is potentially of large impact, despite remaining more difficult to recognize and confirm. The objective of this project is to apply genome-wide quantitative molecular genetics to both, a very integrative and classical quantitative trait (growth in interaction with the environment) and a molecular trait a priori more directly linked to the source of variation (gene expression under cis-regulation). We propose to use a combination of our unique high-troughput phenotyping robot, fine-mapping, complementation approaches and association genetics to pinpoint a significant number of QTLs and eQTLs to the gene level and identify causative polymorphisms and the molecular variation controlling natural diversity. Working at an unprecedented scale should finally allow to resolve enough quantitative loci and pay a significant contribution to drawing a general picture as to how and where in the pathways adaptation is shaping natural variation and improve our understanding of the transcriptional cis-regulatory code.
Summary
Following a long history of quantitative genetics in crop plants, it now becomes feasible to use naturally-occuring variation contained in Arabidopsis thaliana accessions (lines isolated from natural populations) as the source of quantitative genomics approaches, designed to map QTLs and resolve them at the gene level. Apart from being able to exploit in multiple genetic backgrounds allelic variation that cannot be easily generated by conventional mutagenesis, the (relatively few) success of the QTL studies has often been because of the use of quantitative phenotyping, as opposed to the qualitative gauges used in typical mutant screens. Among the various genetic mechanisms responsible for natural variation that have just started to be revealed, cis-acting regulation is potentially of large impact, despite remaining more difficult to recognize and confirm. The objective of this project is to apply genome-wide quantitative molecular genetics to both, a very integrative and classical quantitative trait (growth in interaction with the environment) and a molecular trait a priori more directly linked to the source of variation (gene expression under cis-regulation). We propose to use a combination of our unique high-troughput phenotyping robot, fine-mapping, complementation approaches and association genetics to pinpoint a significant number of QTLs and eQTLs to the gene level and identify causative polymorphisms and the molecular variation controlling natural diversity. Working at an unprecedented scale should finally allow to resolve enough quantitative loci and pay a significant contribution to drawing a general picture as to how and where in the pathways adaptation is shaping natural variation and improve our understanding of the transcriptional cis-regulatory code.
Max ERC Funding
1 742 113 €
Duration
Start date: 2010-02-01, End date: 2016-01-31
Project acronym DeCode
Project Dendrites and memory: role of dendritic spikes in information coding by hippocampal CA3 pyramidal neurons
Researcher (PI) Judit MAKARA
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Consolidator Grant (CoG), LS5, ERC-2017-COG
Summary The hippocampus is essential for building episodic memories. Coding of locations, contexts or events in the hippocampus is based on the correlated activity of neuronal ensembles; however, the mechanisms promoting the recruitment of individual neurons into information-coding ensembles are poorly understood.
In particular, the recurrent synaptic network of pyramidal cells (PCs) in the hippocampal CA3 area, receiving external inputs from the entorhinal cortex and the dentate gyrus, is thought to be essential for associative memory. Current models of the associative functions of CA3 are mainly based on plasticity of these synaptic connections. Recent work by us and others however suggests that active, voltage-dependent properties of CA3PC dendrites may also promote ensemble functions. Dendritic voltage-dependent ion channels allow nonlinear amplification of spatiotemporally correlated synaptic inputs (such as those produced by ensemble activity) and can even generate local dendritic spikes, which may elicit specific action potential patterns and induce synaptic plasticity. Furthermore, dendritic processing may be modulated by activity-dependent regulation of dendritic ion channels. However, still little is known about the active properties of CA3PC dendrites and their functions during spatial coding or memory tasks.
The general aim of my research program is to understand the cellular mechanisms that underlie the formation of hippocampal memory-coding neuronal ensembles. Specifically, we will test the hypothesis that active input integration by dendrites of individual CA3PCs plays an important role in their recruitment into specific context-coding ensembles. By combining in vitro (patch-clamp electrophysiology and two-photon (2P) microscopy in slices) and in vivo (2P imaging and activity-dependent labelling in behaving rodents) approaches, we will provide an in-depth understanding of the dendritic components contributing to the generation of the CA3 ensemble code.
Summary
The hippocampus is essential for building episodic memories. Coding of locations, contexts or events in the hippocampus is based on the correlated activity of neuronal ensembles; however, the mechanisms promoting the recruitment of individual neurons into information-coding ensembles are poorly understood.
In particular, the recurrent synaptic network of pyramidal cells (PCs) in the hippocampal CA3 area, receiving external inputs from the entorhinal cortex and the dentate gyrus, is thought to be essential for associative memory. Current models of the associative functions of CA3 are mainly based on plasticity of these synaptic connections. Recent work by us and others however suggests that active, voltage-dependent properties of CA3PC dendrites may also promote ensemble functions. Dendritic voltage-dependent ion channels allow nonlinear amplification of spatiotemporally correlated synaptic inputs (such as those produced by ensemble activity) and can even generate local dendritic spikes, which may elicit specific action potential patterns and induce synaptic plasticity. Furthermore, dendritic processing may be modulated by activity-dependent regulation of dendritic ion channels. However, still little is known about the active properties of CA3PC dendrites and their functions during spatial coding or memory tasks.
The general aim of my research program is to understand the cellular mechanisms that underlie the formation of hippocampal memory-coding neuronal ensembles. Specifically, we will test the hypothesis that active input integration by dendrites of individual CA3PCs plays an important role in their recruitment into specific context-coding ensembles. By combining in vitro (patch-clamp electrophysiology and two-photon (2P) microscopy in slices) and in vivo (2P imaging and activity-dependent labelling in behaving rodents) approaches, we will provide an in-depth understanding of the dendritic components contributing to the generation of the CA3 ensemble code.
Max ERC Funding
1 990 314 €
Duration
Start date: 2018-06-01, End date: 2023-05-31
Project acronym DECRYPT
Project Decrypting signals in the crypt
Researcher (PI) Philippe, Joseph Sansonetti
Host Institution (HI) INSTITUT PASTEUR
Call Details Advanced Grant (AdG), LS6, ERC-2013-ADG
Summary Pathogens and symbionts: War and Peace at mucosal surface in intestinal crypts.
In the proposed program called DECRYPT, I wish to strengthen novel orientations of our laboratory aimed at decrypting the dialogue between the microbiota and the host, while keeping a balance with the study of pathogens, both being analyzed at their interface with the gut mucosa to further our knowledge of the homeostatic and pathogenic mechanisms that respectively characterize a healthy and a diseased gut. The intestinal crypt is a key location to study this dialogue because it contains the stem cells, the differentiation and transit amplifying/proliferative compartments that are essential for epithelial regeneration at homeostasis, and restitution following an aggression. It is also embedded in a niche of immune cells that participate in homeostatic and pathological processes under microbial stimuli. Thus the breaking nature of my project will bear on the demonstration that crypt homeostasis depends on signals “emitted” by the microbiota, thereby stressing the depth of our symbiosis with the microbial world, and on the demonstration that the crypt is also the target of enteric pathogens like Shigella, thus introducing the novel paradigm that pathogenesis is not only matter of inflammatory destruction of infected tissues, but also of altered epithelial restitution. An extension of this paradigm is that loss or subversion of the microbiota-crypt homeostasis may account not only for inflammatory bowel diseases (IBD), but also for colon cancer. This fundamental knowledge will also be the basis for translational research, particularly the search for molecules that boost antimicrobial defenses and comfort homeostasis. In summary, I propose a balanced combination between the “cellular microbiology of pathogens” and the “cellular microbiology of symbionts”.
Summary
Pathogens and symbionts: War and Peace at mucosal surface in intestinal crypts.
In the proposed program called DECRYPT, I wish to strengthen novel orientations of our laboratory aimed at decrypting the dialogue between the microbiota and the host, while keeping a balance with the study of pathogens, both being analyzed at their interface with the gut mucosa to further our knowledge of the homeostatic and pathogenic mechanisms that respectively characterize a healthy and a diseased gut. The intestinal crypt is a key location to study this dialogue because it contains the stem cells, the differentiation and transit amplifying/proliferative compartments that are essential for epithelial regeneration at homeostasis, and restitution following an aggression. It is also embedded in a niche of immune cells that participate in homeostatic and pathological processes under microbial stimuli. Thus the breaking nature of my project will bear on the demonstration that crypt homeostasis depends on signals “emitted” by the microbiota, thereby stressing the depth of our symbiosis with the microbial world, and on the demonstration that the crypt is also the target of enteric pathogens like Shigella, thus introducing the novel paradigm that pathogenesis is not only matter of inflammatory destruction of infected tissues, but also of altered epithelial restitution. An extension of this paradigm is that loss or subversion of the microbiota-crypt homeostasis may account not only for inflammatory bowel diseases (IBD), but also for colon cancer. This fundamental knowledge will also be the basis for translational research, particularly the search for molecules that boost antimicrobial defenses and comfort homeostasis. In summary, I propose a balanced combination between the “cellular microbiology of pathogens” and the “cellular microbiology of symbionts”.
Max ERC Funding
2 499 992 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym DEEPEN
Project Deciphering deep architectures underlying structured perception in auditory networks
Researcher (PI) Brice Jean Philippe BATHELLIER
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS5, ERC-2017-COG
Summary The principles of sensory perception are still a large experimental and theoretical puzzle. A strong difficulty is that perception emerges from networks of recurrently connected brain areas whose activity and function are poorly approximated by current generic mathematical models. These models also fail to explain many of the fundamental structures effortlessly identified by the brain (shapes, objects, auditory or tactile categories). I here propose to establish a new approach combining high-throughput population recoding methods with a tailored theoretical framework to derive computational principles operating throughout sensory systems and leading to biologically structured perception. This approach follows on the recent mathematical proposal, suggested by Deep Machine Learning methods, that complex perceptual objects emerge through series of simple nonlinear operations combining increasingly complex sensory features along the sensory pathways. Starting with the mouse auditory system as a model pathway, we will recursively extract, with model-free methods, the main nonlinear sensory features encoded in genetically tagged output and local neurons at different processing stages, using optical and electrophysiological high density recording techniques in awake animals. The role of these features in perception will be identified with behavioural assays. Specific intra- and interareal feedback connections, typically not included in Deep Leaning models, will be opto- and chemogenetically perturbed to assess their contribution to precise nonlinearities of the system and their role in the emergence of complex perceptual structures. Based on these structural, functional and perturbation data, a new generation of well-constrained and predictive sensory processing models will be built, serving as a platform to extract general computational principles missing to link neural activity to perception and to fuel artificial neural networks technologies.
Summary
The principles of sensory perception are still a large experimental and theoretical puzzle. A strong difficulty is that perception emerges from networks of recurrently connected brain areas whose activity and function are poorly approximated by current generic mathematical models. These models also fail to explain many of the fundamental structures effortlessly identified by the brain (shapes, objects, auditory or tactile categories). I here propose to establish a new approach combining high-throughput population recoding methods with a tailored theoretical framework to derive computational principles operating throughout sensory systems and leading to biologically structured perception. This approach follows on the recent mathematical proposal, suggested by Deep Machine Learning methods, that complex perceptual objects emerge through series of simple nonlinear operations combining increasingly complex sensory features along the sensory pathways. Starting with the mouse auditory system as a model pathway, we will recursively extract, with model-free methods, the main nonlinear sensory features encoded in genetically tagged output and local neurons at different processing stages, using optical and electrophysiological high density recording techniques in awake animals. The role of these features in perception will be identified with behavioural assays. Specific intra- and interareal feedback connections, typically not included in Deep Leaning models, will be opto- and chemogenetically perturbed to assess their contribution to precise nonlinearities of the system and their role in the emergence of complex perceptual structures. Based on these structural, functional and perturbation data, a new generation of well-constrained and predictive sensory processing models will be built, serving as a platform to extract general computational principles missing to link neural activity to perception and to fuel artificial neural networks technologies.
Max ERC Funding
1 983 886 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym DEPREC
Project The Dependence Receptors notion: from a cell biology paradigm to anti-cancer targeted therapy
Researcher (PI) Patrick Mehlen
Host Institution (HI) UNIVERSITE LYON 1 CLAUDE BERNARD
Call Details Advanced Grant (AdG), LS4, ERC-2011-ADG_20110310
Summary "While it is assumed that transmembrane receptors are active only in the presence of ligand, we have proposed that some receptors may also be active in the absence of ligand stimulation. These receptors, named “dependence receptors” (DRs) share the ability to transmit two opposite signals: in the presence of ligand, these receptors transduce various classical “positive” signals, whereas in the absence of ligand, they trigger apoptosis. The expression of dependence receptors thus creates cellular states of dependence for survival on their respective ligands. To date, more than fifteen such receptors have been identified, including the netrin-1 receptors DCC (Deleted in Colorectal Cancer) and UNC5H1-4, some integrins, RET, EPHA4, TrkA, TrkC and the Sonic Hedgehog receptor Patched (Ptc). Even though the interest in this notion is increasing, two main questions remain poorly understood: (i) how very different receptors, with only modest homology, are able to trigger apoptosis when unengaged by their respective ligand, and (ii) what are the respective biological roles of this pro-apoptotic activity in vivo. We have hypothesized that the DRs pro-apoptotic activity is a mechanism that determines and regulates the territories of migration/localization of cells during embryonic development. We also demonstrated that this may be a mechanism that limits tumor growth and metastasis. The goal of the present project is, based on the study of a relatively small number of these receptors –i.e., DCC, UNC5H, RET, TrkC, Ptc- with a specifically larger emphasis on netrin-1 receptors, to address (i) the common and divergent cell signaling mechanisms triggering apoptosis downstream of these receptors and (ii) the physiological and pathological roles of these DRs on development of neoplasia in vivo. This latter goal will allow us to investigate how this pro-apoptotic activity can be of use to improve and diversify alternative anti-cancer therapeutic approaches."
Summary
"While it is assumed that transmembrane receptors are active only in the presence of ligand, we have proposed that some receptors may also be active in the absence of ligand stimulation. These receptors, named “dependence receptors” (DRs) share the ability to transmit two opposite signals: in the presence of ligand, these receptors transduce various classical “positive” signals, whereas in the absence of ligand, they trigger apoptosis. The expression of dependence receptors thus creates cellular states of dependence for survival on their respective ligands. To date, more than fifteen such receptors have been identified, including the netrin-1 receptors DCC (Deleted in Colorectal Cancer) and UNC5H1-4, some integrins, RET, EPHA4, TrkA, TrkC and the Sonic Hedgehog receptor Patched (Ptc). Even though the interest in this notion is increasing, two main questions remain poorly understood: (i) how very different receptors, with only modest homology, are able to trigger apoptosis when unengaged by their respective ligand, and (ii) what are the respective biological roles of this pro-apoptotic activity in vivo. We have hypothesized that the DRs pro-apoptotic activity is a mechanism that determines and regulates the territories of migration/localization of cells during embryonic development. We also demonstrated that this may be a mechanism that limits tumor growth and metastasis. The goal of the present project is, based on the study of a relatively small number of these receptors –i.e., DCC, UNC5H, RET, TrkC, Ptc- with a specifically larger emphasis on netrin-1 receptors, to address (i) the common and divergent cell signaling mechanisms triggering apoptosis downstream of these receptors and (ii) the physiological and pathological roles of these DRs on development of neoplasia in vivo. This latter goal will allow us to investigate how this pro-apoptotic activity can be of use to improve and diversify alternative anti-cancer therapeutic approaches."
Max ERC Funding
2 485 037 €
Duration
Start date: 2012-05-01, End date: 2017-04-30
Project acronym DIATOMIC
Project Untangling eco-evolutionary impacts on diatom genomes over timescales relevant to current climate change
Researcher (PI) Christopher Paul BOWLER
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS8, ERC-2018-ADG
Summary Diatoms are major contributors of primary production in the ocean and participate in carbon sequestration over geologically relevant timescales. As key components of the Earth’s carbon cycle and marine food webs we need to understand the eco-evolutionary underpinnings of their ecological success to forecast their fate in a future ocean impacted by anthropogenic change. Genomes and epigenomes from model diatoms, as well as hundreds of transcriptomes from multiple species, have revealed genetic and epigenetic processes regulating gene expression in response to changing environments. The Tara Oceans survey has in parallel generated resources to explore diatom abundance, diversity and gene expression in the world’s ocean in widely contrasting conditions. DIATOMIC will build on these resources to understand how evolutionary and ecological processes combine to influence diatom adaptations to their environment at unprecedented spatiotemporal scales. To examine these processes over timescales relevant to current climate change, DIATOMIC includes the pioneering exploration of ancient diatom DNA from the sub-seafloor to reveal the genetic and epigenetic bases of speciation and adaptation that have impacted their ecological success during the last 100,000 years, when Earth experienced major climatological events and an increase in anthropogenic impacts. As a model for exploring eco-evolutionary processes in the past and contemporary ocean we will focus primarily on Chaetoceros because this diatom genus is ancient, ubiquitous, abundant and contributes significantly to carbon export. Key findings will be additionally supported by lab-based studies using the diatom Phaeodactylum for which exemplar molecular tools exist. Specifically, the project will address:
1. What molecular features characterize genome evolution in diatoms?
2. Which processes determine diatom metapopulation structure?
3. What can ancient DNA tell us about diatom adaptations to environmental change in the past?
Summary
Diatoms are major contributors of primary production in the ocean and participate in carbon sequestration over geologically relevant timescales. As key components of the Earth’s carbon cycle and marine food webs we need to understand the eco-evolutionary underpinnings of their ecological success to forecast their fate in a future ocean impacted by anthropogenic change. Genomes and epigenomes from model diatoms, as well as hundreds of transcriptomes from multiple species, have revealed genetic and epigenetic processes regulating gene expression in response to changing environments. The Tara Oceans survey has in parallel generated resources to explore diatom abundance, diversity and gene expression in the world’s ocean in widely contrasting conditions. DIATOMIC will build on these resources to understand how evolutionary and ecological processes combine to influence diatom adaptations to their environment at unprecedented spatiotemporal scales. To examine these processes over timescales relevant to current climate change, DIATOMIC includes the pioneering exploration of ancient diatom DNA from the sub-seafloor to reveal the genetic and epigenetic bases of speciation and adaptation that have impacted their ecological success during the last 100,000 years, when Earth experienced major climatological events and an increase in anthropogenic impacts. As a model for exploring eco-evolutionary processes in the past and contemporary ocean we will focus primarily on Chaetoceros because this diatom genus is ancient, ubiquitous, abundant and contributes significantly to carbon export. Key findings will be additionally supported by lab-based studies using the diatom Phaeodactylum for which exemplar molecular tools exist. Specifically, the project will address:
1. What molecular features characterize genome evolution in diatoms?
2. Which processes determine diatom metapopulation structure?
3. What can ancient DNA tell us about diatom adaptations to environmental change in the past?
Max ERC Funding
2 495 753 €
Duration
Start date: 2019-11-01, End date: 2024-10-31
Project acronym DIATOMITE
Project Genome-enabled dissection of marine diatom ecophysiology
Researcher (PI) Chris Bowler
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS2, ERC-2011-ADG_20110310
Summary "Diatoms are the most successful group of eukaryotic phytoplankton in the modern ocean. Recently completed whole genome sequences have revealed a wealth of information about the evolutionary origins and metabolic adaptations that may have led to their ecological success. A major finding is that they have acquired genes both from their endosymbiotic ancestors and by horizontal gene transfer from marine bacteria. This unique melting pot of genes encodes novel and largely unexplored capacities for metabolic management. The project will address the current gap in knowledge about the physiological functions of diatom gene products and about the evolutionary mechanisms that have led to diatom success in contemporary oceans. We will exploit genome-enabled approaches to pioneer new research topics addressing:
1. How has diatom evolution enabled interactions between chloroplasts and mitochondria that have provided diatoms with physiological and metabolic innovations?
2. What are the relative contributions of DNA sequence variation and epigenetic processes in diatom adaptive dynamics?
By combining these questions, we will uniquely be able to identify sentinel genes that have driven major physiological and metabolic innovations in diatoms, and will explore the mechanisms that have selected and molded them during diatom evolution. We will focus our studies largely on diatom responses to nutrients, in particular nitrate and iron, and will exploit the advantages of Phaeodactylum tricornutum as a model diatom species for reverse genetics. The proposed studies will revisit textbook understanding of photosynthesis and nitrogen metabolism, and will refine hypotheses about why diatoms dominate in contemporary ocean settings. By placing our studies in evolutionary and ecological contexts, in particular by examining the contribution of epigenetic processes in diatoms, our work will furthermore provide insights into how the environment selects for fitness in phytoplankton."
Summary
"Diatoms are the most successful group of eukaryotic phytoplankton in the modern ocean. Recently completed whole genome sequences have revealed a wealth of information about the evolutionary origins and metabolic adaptations that may have led to their ecological success. A major finding is that they have acquired genes both from their endosymbiotic ancestors and by horizontal gene transfer from marine bacteria. This unique melting pot of genes encodes novel and largely unexplored capacities for metabolic management. The project will address the current gap in knowledge about the physiological functions of diatom gene products and about the evolutionary mechanisms that have led to diatom success in contemporary oceans. We will exploit genome-enabled approaches to pioneer new research topics addressing:
1. How has diatom evolution enabled interactions between chloroplasts and mitochondria that have provided diatoms with physiological and metabolic innovations?
2. What are the relative contributions of DNA sequence variation and epigenetic processes in diatom adaptive dynamics?
By combining these questions, we will uniquely be able to identify sentinel genes that have driven major physiological and metabolic innovations in diatoms, and will explore the mechanisms that have selected and molded them during diatom evolution. We will focus our studies largely on diatom responses to nutrients, in particular nitrate and iron, and will exploit the advantages of Phaeodactylum tricornutum as a model diatom species for reverse genetics. The proposed studies will revisit textbook understanding of photosynthesis and nitrogen metabolism, and will refine hypotheses about why diatoms dominate in contemporary ocean settings. By placing our studies in evolutionary and ecological contexts, in particular by examining the contribution of epigenetic processes in diatoms, our work will furthermore provide insights into how the environment selects for fitness in phytoplankton."
Max ERC Funding
2 423 320 €
Duration
Start date: 2012-06-01, End date: 2017-05-31
Project acronym DIRONAKI
Project Differentiation and role of Natural Killer cell subsets
Researcher (PI) Thierry Walzer
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS6, ERC-2011-StG_20101109
Summary "NK cells are innate lymphocytes that play a role in the early response against intracellular pathogens and against tumors. Several NK cell subsets have been described in peripheral organs that correspond to discrete stages of in vivo maturation. How NK cells differentiate from early precursors and what are the specific functions of each NK cell subset are unresolved issues. Here, we propose a three-aim program to address these questions. First, we want to revisit the partition of the NK cell population that is currently based on surface markers of undefined function by looking at the expression of transcription factors (TF) essential for NK cell development and maturation, such as T-bet and Eomes, using novel TF reporter mice. This strategy should also allow us to identify very early steps of NK cell development (NK cell progenitors) that remain ill defined. Second, we will try and identify molecular mechanisms that induce transition between NK cell maturation stages. For this we will take advantage of a previous gene profiling analysis that pointed at several pathways and TF that were highly regulated during NK cell maturation. The role of these pathways and TF in the differentiation of NK cells will be measured using a novel Cre/lox system allowing NK-specific gene deletion. Detailed analysis of mouse mutants will be used to delineate the role of selected genes and pathways in NK cell differentiation. Third, we will compare patterns of migration, cytokine secretion, in vivo cytotoxicity and global gene expression by individual NK cell subsets during an airway infection by Influenza to get insight on the specific functions of NK cell subsets during immune responses. Altogether, the results of this study should provide developmental, molecular and functional evidences to support the physiological relevance of NK cell subsets. This may improve strategies that aim at manipulating NK cell function for the benefit of patients with cancer or chronic infectious diseases"
Summary
"NK cells are innate lymphocytes that play a role in the early response against intracellular pathogens and against tumors. Several NK cell subsets have been described in peripheral organs that correspond to discrete stages of in vivo maturation. How NK cells differentiate from early precursors and what are the specific functions of each NK cell subset are unresolved issues. Here, we propose a three-aim program to address these questions. First, we want to revisit the partition of the NK cell population that is currently based on surface markers of undefined function by looking at the expression of transcription factors (TF) essential for NK cell development and maturation, such as T-bet and Eomes, using novel TF reporter mice. This strategy should also allow us to identify very early steps of NK cell development (NK cell progenitors) that remain ill defined. Second, we will try and identify molecular mechanisms that induce transition between NK cell maturation stages. For this we will take advantage of a previous gene profiling analysis that pointed at several pathways and TF that were highly regulated during NK cell maturation. The role of these pathways and TF in the differentiation of NK cells will be measured using a novel Cre/lox system allowing NK-specific gene deletion. Detailed analysis of mouse mutants will be used to delineate the role of selected genes and pathways in NK cell differentiation. Third, we will compare patterns of migration, cytokine secretion, in vivo cytotoxicity and global gene expression by individual NK cell subsets during an airway infection by Influenza to get insight on the specific functions of NK cell subsets during immune responses. Altogether, the results of this study should provide developmental, molecular and functional evidences to support the physiological relevance of NK cell subsets. This may improve strategies that aim at manipulating NK cell function for the benefit of patients with cancer or chronic infectious diseases"
Max ERC Funding
1 340 757 €
Duration
Start date: 2012-01-01, End date: 2017-12-31
Project acronym DIvA
Project Chromatin function in DNA Double Strand breaks repair: Prime, repair and restore DSB Inducible via AsiSI
Researcher (PI) Gaelle LEGUBE
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS1, ERC-2014-CoG
Summary "Among the types of damage, DNA Double Strands Breaks (DSBs) are the most deleterious, as illustrated by the variety of human diseases associated with DSB repair defects. Repair of DSB into the chromatin context raises several questions that we aim to address in this proposal. Firstly, it is likely that the chromatin environment where a break occurs influences the choice of repair pathway. Since the different DSB repair mechanisms can lead to different "scar" on the genome, further studies are required to elucidate how chromatin structure regulates the targeting of DSB repair machineries. Secondly, DNA packaging into chromatin hinders detection and repair of DSBs and many chromatin modifications were recently identified as induced around DSBs to facilitate repair. However, a complete picture of the chromatin landscape set up at DSB, and more specifically the set of histone modifications associated with each repair pathway ("repair histone code") is still awaited. In addition, whether and how damaged chromosomes are reorganized within the nucleus is still unknown. Finally, once repair has been completed, the initial chromatin landscape must be faithfully restored in order to maintain epigenome stability and cell fate.
Using an experimental system we recently developed (called DIvA for DSB Inducible via AsiSI), that allows the induction of multiple sequence-specific DSBs widespread across the genome, we propose to investigate these uncovered aspects of the relationship between chromatin and DSB repair. By high-throughput genomic and proteomic technologies, we will try (i) to understand the contribution of chromatin in the DSB repair pathway choice (PRIME), (ii) to describe more thoroughly the chromatin remodeling events and the spatial chromosomes reorganization, that occur concomitantly to DSB to promote adequate repair (REPAIR), and (iii) to elucidate the processes at work to restore epigenome integrity after DSB repair (RESTORE)."
Summary
"Among the types of damage, DNA Double Strands Breaks (DSBs) are the most deleterious, as illustrated by the variety of human diseases associated with DSB repair defects. Repair of DSB into the chromatin context raises several questions that we aim to address in this proposal. Firstly, it is likely that the chromatin environment where a break occurs influences the choice of repair pathway. Since the different DSB repair mechanisms can lead to different "scar" on the genome, further studies are required to elucidate how chromatin structure regulates the targeting of DSB repair machineries. Secondly, DNA packaging into chromatin hinders detection and repair of DSBs and many chromatin modifications were recently identified as induced around DSBs to facilitate repair. However, a complete picture of the chromatin landscape set up at DSB, and more specifically the set of histone modifications associated with each repair pathway ("repair histone code") is still awaited. In addition, whether and how damaged chromosomes are reorganized within the nucleus is still unknown. Finally, once repair has been completed, the initial chromatin landscape must be faithfully restored in order to maintain epigenome stability and cell fate.
Using an experimental system we recently developed (called DIvA for DSB Inducible via AsiSI), that allows the induction of multiple sequence-specific DSBs widespread across the genome, we propose to investigate these uncovered aspects of the relationship between chromatin and DSB repair. By high-throughput genomic and proteomic technologies, we will try (i) to understand the contribution of chromatin in the DSB repair pathway choice (PRIME), (ii) to describe more thoroughly the chromatin remodeling events and the spatial chromosomes reorganization, that occur concomitantly to DSB to promote adequate repair (REPAIR), and (iii) to elucidate the processes at work to restore epigenome integrity after DSB repair (RESTORE)."
Max ERC Funding
2 000 000 €
Duration
Start date: 2015-07-01, End date: 2020-06-30
Project acronym DNATRAFFIC
Project DNA traffic during bacterial cell division
Researcher (PI) François-Xavier Andre Fernand Barre
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), LS1, ERC-2011-StG_20101109
Summary The molecular mechanisms that serve to couple DNA replication, chromosome segregation and cell division are largely unknown in bacteria. This led a considerable interest to the study of Escherichia coli FtsK, an essential cell division protein that assembles into DNA-pumps to transfer chromosomal DNA between the two daughter cell compartments during septation. Indeed, our recent work suggests that FtsK might regulate the late stages of septation to ensure DNA is fully cleared from the septum before it is allowed to close. This would be the first example of a cell cycle checkpoint in bacteria.
FtsK-mediated DNA transfer is required in 15% of the cells at each generation in E. coli, in which it serves to promote the resolution of topological problems arising from the circularity of the chromosome by Xer recombination. However, the FtsK checkpoint could be a more general feature of the bacterial cell cycle since FtsK is highly conserved among eubacteria, including species that do not possess a Xer system. Indeed, preliminary results from the lab indicate that DNA transfer by FtsK is required independently of Xer recombination in Vibrio cholerae.
To confirm the existence and the generality of the FtsK checkpoint in bacteria, we will determine the different situations that lead to a requirement for FtsK-mediated DNA transfer by studying chromosome segregation and cell division in V. cholerae. In parallel, we will apply new fluorescent microscopy tools to follow the progression of cell division and chromosome segregation in single live bacterial cells. PALM will notably serve to probe the structure of the FtsK DNA-pumps at a high spatial resolution, FRET will be used to determine their timing of assembly and their interactions with the other cell division proteins, and TIRF will serve to follow in real time their activity with respect to the progression of chromosome dimer resolution, chromosome segregation, and septum closure.
Summary
The molecular mechanisms that serve to couple DNA replication, chromosome segregation and cell division are largely unknown in bacteria. This led a considerable interest to the study of Escherichia coli FtsK, an essential cell division protein that assembles into DNA-pumps to transfer chromosomal DNA between the two daughter cell compartments during septation. Indeed, our recent work suggests that FtsK might regulate the late stages of septation to ensure DNA is fully cleared from the septum before it is allowed to close. This would be the first example of a cell cycle checkpoint in bacteria.
FtsK-mediated DNA transfer is required in 15% of the cells at each generation in E. coli, in which it serves to promote the resolution of topological problems arising from the circularity of the chromosome by Xer recombination. However, the FtsK checkpoint could be a more general feature of the bacterial cell cycle since FtsK is highly conserved among eubacteria, including species that do not possess a Xer system. Indeed, preliminary results from the lab indicate that DNA transfer by FtsK is required independently of Xer recombination in Vibrio cholerae.
To confirm the existence and the generality of the FtsK checkpoint in bacteria, we will determine the different situations that lead to a requirement for FtsK-mediated DNA transfer by studying chromosome segregation and cell division in V. cholerae. In parallel, we will apply new fluorescent microscopy tools to follow the progression of cell division and chromosome segregation in single live bacterial cells. PALM will notably serve to probe the structure of the FtsK DNA-pumps at a high spatial resolution, FRET will be used to determine their timing of assembly and their interactions with the other cell division proteins, and TIRF will serve to follow in real time their activity with respect to the progression of chromosome dimer resolution, chromosome segregation, and septum closure.
Max ERC Funding
1 565 938 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym DOFOCO
Project Do forests cool the Earth? Reconciling sustained productivity and minimum climate response with portfolios of contrasting forest management strategies
Researcher (PI) Sebastiaan Luyssaert
Host Institution (HI) COMMISSARIAT A L ENERGIE ATOMIQUE ET AUX ENERGIES ALTERNATIVES
Call Details Starting Grant (StG), LS9, ERC-2009-StG
Summary Forests, of which globally 70% are managed, play a particularly important role in the global carbon cycle. Recently, forest management became a top priority on the agenda of the political negotiations to mitigate climate change because forest plantations may remove atmospheric CO2 and if used for energy production, the wood is a substitute for fossil fuel. However, this political imperative is at present running well ahead of the science required to deliver it. Despite the key implications of forest management on: 1) the carbon-energy-water balance, and 2) production, recreation and environmental protection, there are no integrated studies of its effects on the Earth s climate. The overall goal of DOFOCO is to quantify and understand the role of forest management in mitigating climate change. Specifically, I want to challenge the current focus on the carbon cycle and replace it with a total climate impact approach. Hence, the whole forest management spectrum ranging from short rotation coppice to old-growth forests will be analyzed for its effects on the water, energy and carbon cycles. Climate response of forest will be quantified by means of albedo, evapotranspiration, greenhouse gas sources and sinks and their resulting climate feedback mechanisms. The anticipated new quantitative results will be used to lay the foundations for a portfolio of management strategies which will sustain wood production while minimizing climate change impacts. DOFOCO is interdisciplinary and ground breaking because it brings together state-of-the art data and models from applied life and Earth system sciences; it will deliver the first quantitative insights into how forest management strategies can be linked to climate change mitigation.
Summary
Forests, of which globally 70% are managed, play a particularly important role in the global carbon cycle. Recently, forest management became a top priority on the agenda of the political negotiations to mitigate climate change because forest plantations may remove atmospheric CO2 and if used for energy production, the wood is a substitute for fossil fuel. However, this political imperative is at present running well ahead of the science required to deliver it. Despite the key implications of forest management on: 1) the carbon-energy-water balance, and 2) production, recreation and environmental protection, there are no integrated studies of its effects on the Earth s climate. The overall goal of DOFOCO is to quantify and understand the role of forest management in mitigating climate change. Specifically, I want to challenge the current focus on the carbon cycle and replace it with a total climate impact approach. Hence, the whole forest management spectrum ranging from short rotation coppice to old-growth forests will be analyzed for its effects on the water, energy and carbon cycles. Climate response of forest will be quantified by means of albedo, evapotranspiration, greenhouse gas sources and sinks and their resulting climate feedback mechanisms. The anticipated new quantitative results will be used to lay the foundations for a portfolio of management strategies which will sustain wood production while minimizing climate change impacts. DOFOCO is interdisciplinary and ground breaking because it brings together state-of-the art data and models from applied life and Earth system sciences; it will deliver the first quantitative insights into how forest management strategies can be linked to climate change mitigation.
Max ERC Funding
1 296 125 €
Duration
Start date: 2010-02-01, End date: 2015-10-31
Project acronym DRIVE
Project GUIDANCE AND FUNCTION OF REGENERATIVE FIBERS IN ADULT CNS
Researcher (PI) Homaira NAWABI
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS5, ERC-2017-STG
Summary The impairment of central nervous system (CNS) leads to irreversible loss of vital functions because, unlike young neurons, mature neurons are not able to regenerate. Thus, understanding the detailed mechanisms of axonal growth and repair remains one of the greatest challenges of neurobiology and for society. If the extrinsic factors fail to reach levels required for regeneration, manipulating intrinsic pathways has shown promising results. Particularly, my work demonstrated that the simultaneous activation of mTOR, JAK/STAT and c-myc pathways allows exceptional regeneration with axons close to their targets. However it also exacerbates previously described phenomenon of misguidance with potential aberrant circuit formation. My unique model opens up the possibility to explore these fundamental questions of CNS regeneration. I propose to address the yet unexplored problem of the guidance of regenerating axons in adults in order to promote the formation of a functional new circuit after injury. Indeed what are the modalities of guidance in the adult? Are axons still responsive to developmental guidance cues and are they still expressed? Can regenerative axons form connections with their targets and are these connections functional?
To answer these critical questions, I will use the combination of state of the art biochemistry, imaging, and electrophysiology in an in-vivo and ex-vivo model of the visual system to 1) Understand axon guidance in mature system in order to properly drive regenerative axons to their brain targets and avoid aberrant projections, and 2) Analyze the formation of a functional optic nerve circuit after injury. Altogether, these results will generate major breakthroughs in a fundamental but uncovered mechanism of axon guidance during regeneration and the functionality of de novo formed circuits. They will open up new ways for innovative therapeutic development after CNS trauma but also to the large spectrum of neurodegenerative diseases.
Summary
The impairment of central nervous system (CNS) leads to irreversible loss of vital functions because, unlike young neurons, mature neurons are not able to regenerate. Thus, understanding the detailed mechanisms of axonal growth and repair remains one of the greatest challenges of neurobiology and for society. If the extrinsic factors fail to reach levels required for regeneration, manipulating intrinsic pathways has shown promising results. Particularly, my work demonstrated that the simultaneous activation of mTOR, JAK/STAT and c-myc pathways allows exceptional regeneration with axons close to their targets. However it also exacerbates previously described phenomenon of misguidance with potential aberrant circuit formation. My unique model opens up the possibility to explore these fundamental questions of CNS regeneration. I propose to address the yet unexplored problem of the guidance of regenerating axons in adults in order to promote the formation of a functional new circuit after injury. Indeed what are the modalities of guidance in the adult? Are axons still responsive to developmental guidance cues and are they still expressed? Can regenerative axons form connections with their targets and are these connections functional?
To answer these critical questions, I will use the combination of state of the art biochemistry, imaging, and electrophysiology in an in-vivo and ex-vivo model of the visual system to 1) Understand axon guidance in mature system in order to properly drive regenerative axons to their brain targets and avoid aberrant projections, and 2) Analyze the formation of a functional optic nerve circuit after injury. Altogether, these results will generate major breakthroughs in a fundamental but uncovered mechanism of axon guidance during regeneration and the functionality of de novo formed circuits. They will open up new ways for innovative therapeutic development after CNS trauma but also to the large spectrum of neurodegenerative diseases.
Max ERC Funding
1 499 410 €
Duration
Start date: 2018-03-01, End date: 2023-02-28
Project acronym DRIWGHP
Project The Distribution and Redistribution of Income and Wealth: A Global and Historical Perspective
Researcher (PI) Thomas Piketty
Host Institution (HI) ECOLE D'ECONOMIE DE PARIS
Call Details Advanced Grant (AdG), SH1, ERC-2013-ADG
Summary "Income and wealth inequality has widened significantly in many developed countries during the past 40 years, particularly in the Anglo-Saxon world. In some countries, e.g. the US, income concentration is now higher than in the early decades of the 20th century. EU trends are less strong, but push in the same direction. Yet, despite these puzzling facts, we still know very little about the forces behind the long run evolution of income and wealth distribution. The central objective of this proposal is to better understand the rise in inequality, and more generally to develop a unified empirical and theoretical approach to the distribution and redistribution of income and wealth.
First, I propose to construct a new ""World Wealth and Income Database"" (WWID) that will be made public through a dedicated website. Existing inequality data sets are insufficient, first because many countries are not well covered, and mostly because available series concentrate on income inequality and usually do not cover wealth inequality. This is unfortunate, because the theoretical forces at play are very different for income and wealth distributions. The WWID will remedy both deficiencies and allow for a better articulation between available data and theoretical models.
Next, I will use this new database to test for the various mechanisms explaining the rise in inequality. In particular, I will explore the extent to which low growth and high returns to wealth naturally push towards higher wealth-income ratios as well as rising wealth concentration. In the near future this mechanism is likely to be particularly strong in low growth Europe (especially in countries with negative population growth). In the long run it can also operate at the level of the global distribution of wealth. I will also develop new theoretical models of optimal taxation of income and wealth. These models will be using a ""sufficient statistics"" approach and will be calibrated using WWID data."
Summary
"Income and wealth inequality has widened significantly in many developed countries during the past 40 years, particularly in the Anglo-Saxon world. In some countries, e.g. the US, income concentration is now higher than in the early decades of the 20th century. EU trends are less strong, but push in the same direction. Yet, despite these puzzling facts, we still know very little about the forces behind the long run evolution of income and wealth distribution. The central objective of this proposal is to better understand the rise in inequality, and more generally to develop a unified empirical and theoretical approach to the distribution and redistribution of income and wealth.
First, I propose to construct a new ""World Wealth and Income Database"" (WWID) that will be made public through a dedicated website. Existing inequality data sets are insufficient, first because many countries are not well covered, and mostly because available series concentrate on income inequality and usually do not cover wealth inequality. This is unfortunate, because the theoretical forces at play are very different for income and wealth distributions. The WWID will remedy both deficiencies and allow for a better articulation between available data and theoretical models.
Next, I will use this new database to test for the various mechanisms explaining the rise in inequality. In particular, I will explore the extent to which low growth and high returns to wealth naturally push towards higher wealth-income ratios as well as rising wealth concentration. In the near future this mechanism is likely to be particularly strong in low growth Europe (especially in countries with negative population growth). In the long run it can also operate at the level of the global distribution of wealth. I will also develop new theoretical models of optimal taxation of income and wealth. These models will be using a ""sufficient statistics"" approach and will be calibrated using WWID data."
Max ERC Funding
2 489 576 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym Drug-Seq
Project Unravelling the Genomic Targets of Drugs Using High-Throughput Sequencing
Researcher (PI) Raphaël Rodriguez
Host Institution (HI) INSTITUT CURIE
Call Details Consolidator Grant (CoG), LS7, ERC-2014-CoG
Summary This proposal is centred on the development of small molecule probes derived from DNA damaging agents to identify their genomic targets using a novel unbiased approach. Although, several genotoxic drugs have been used for decades to treat cancers, the exact mechanisms by which they operate are not fully understood. It is established that these compounds interfere with the processes of transcription and replication, thereby promoting genomic instability and cell death. However, there is as yet no genome-wide map of the exact location of sites that are putative targets for these drugs in vivo. This information is critical to understand and rationalize cellular responses to genotoxic agents. Here, we propose to develop an innovative discovery- based methodology that will combine click chemistry in situ, affinity pull-down techniques and high throughput DNA sequencing (Drug-Seq), to identify the genomic interactome of DNA damaging drugs in order to elucidate their cellular activity at the molecular level. Two independent lines of enquiry will be followed. Firstly, we will establish the genomic interacting landscape of landmark drugs including etoposide, camptothecin and cisplatin using Drug-Seq. Secondly, we will perform regular chromatin immuno- precipitation sequencing (ChIP-Seq) of selected proteins linked to the cellular response of interest to validate Drug-Seq and further identify druggable genomic sites. An important aim of this proposal is to establish a universal methodology to decipher small molecule/genome interactions in vivo that trigger a particular response in disease-relevant models. We also seek to interrogate the role of chromatin in regulating drug/genome interactions and to define whether it is possible to act on the epigenome to modulate the activity and specificity of these drugs. Collectively, we anticipate our study will lay down the foundation for personalized medicine with the implementation of rational rather than empirical clinical protocols.
Summary
This proposal is centred on the development of small molecule probes derived from DNA damaging agents to identify their genomic targets using a novel unbiased approach. Although, several genotoxic drugs have been used for decades to treat cancers, the exact mechanisms by which they operate are not fully understood. It is established that these compounds interfere with the processes of transcription and replication, thereby promoting genomic instability and cell death. However, there is as yet no genome-wide map of the exact location of sites that are putative targets for these drugs in vivo. This information is critical to understand and rationalize cellular responses to genotoxic agents. Here, we propose to develop an innovative discovery- based methodology that will combine click chemistry in situ, affinity pull-down techniques and high throughput DNA sequencing (Drug-Seq), to identify the genomic interactome of DNA damaging drugs in order to elucidate their cellular activity at the molecular level. Two independent lines of enquiry will be followed. Firstly, we will establish the genomic interacting landscape of landmark drugs including etoposide, camptothecin and cisplatin using Drug-Seq. Secondly, we will perform regular chromatin immuno- precipitation sequencing (ChIP-Seq) of selected proteins linked to the cellular response of interest to validate Drug-Seq and further identify druggable genomic sites. An important aim of this proposal is to establish a universal methodology to decipher small molecule/genome interactions in vivo that trigger a particular response in disease-relevant models. We also seek to interrogate the role of chromatin in regulating drug/genome interactions and to define whether it is possible to act on the epigenome to modulate the activity and specificity of these drugs. Collectively, we anticipate our study will lay down the foundation for personalized medicine with the implementation of rational rather than empirical clinical protocols.
Max ERC Funding
1 999 900 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym DYMOCHRO
Project Dynamics of modified chromatin domains
Researcher (PI) Fabian, Roman ERDEL
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), LS1, ERC-2018-STG
Summary Cellular identity is defined by complex patterns of DNA and histone modifications, which partition our chromosomes and determine how cells interpret the genetic information. For cells to remember who they are, these modifications have to be tightly regulated over time and through cell division. Loss of cellular identity promotes different types of disease including neurological disorders and cancer.
Histone modifications can spread along chromatin and can be transmitted through cell division, giving rise to chromatin position effects and cellular memory. However, the underlying molecular mechanisms are not understood. In particular, we do not know how the size and stability of modified domains is controlled, and we currently lack techniques to study these processes in real-time.
Here, I propose to develop the single-molecule ‘chromatin curtains’ platform to directly visualize spreading and maintenance of histone methylation in a reconstituted system and to compare the resulting domains to those found on individual chromatin fibers isolated from cells. In a complementary approach, I will devise and set up a tunable synthetic circuit that installs and reinforces orthogonal epigenetic modifications in living cells to define the functional modules that are necessary and sufficient for chromatin position effects and cellular memory.
My project combines molecular biophysics with synthetic biology to elucidate the fundamental principles that govern the dynamics of histone modifications to establish and preserve cellular identity. The chromatin curtains platform I will develop complements sequencing-based methods and will make it for the first time possible to directly assess the dynamics of histone modification patterns on single chromatin fibers under native conditions. I anticipate that the insights gained in my project will aid in the design of future strategies to control histone modifications in disease.
Summary
Cellular identity is defined by complex patterns of DNA and histone modifications, which partition our chromosomes and determine how cells interpret the genetic information. For cells to remember who they are, these modifications have to be tightly regulated over time and through cell division. Loss of cellular identity promotes different types of disease including neurological disorders and cancer.
Histone modifications can spread along chromatin and can be transmitted through cell division, giving rise to chromatin position effects and cellular memory. However, the underlying molecular mechanisms are not understood. In particular, we do not know how the size and stability of modified domains is controlled, and we currently lack techniques to study these processes in real-time.
Here, I propose to develop the single-molecule ‘chromatin curtains’ platform to directly visualize spreading and maintenance of histone methylation in a reconstituted system and to compare the resulting domains to those found on individual chromatin fibers isolated from cells. In a complementary approach, I will devise and set up a tunable synthetic circuit that installs and reinforces orthogonal epigenetic modifications in living cells to define the functional modules that are necessary and sufficient for chromatin position effects and cellular memory.
My project combines molecular biophysics with synthetic biology to elucidate the fundamental principles that govern the dynamics of histone modifications to establish and preserve cellular identity. The chromatin curtains platform I will develop complements sequencing-based methods and will make it for the first time possible to directly assess the dynamics of histone modification patterns on single chromatin fibers under native conditions. I anticipate that the insights gained in my project will aid in the design of future strategies to control histone modifications in disease.
Max ERC Funding
1 447 860 €
Duration
Start date: 2019-07-01, End date: 2024-06-30
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 DYNACOTINE
Project Signal transduction and allosteric modulation of nicotinic acetylcholine receptors:from ion channel electrophysiology to atomic 3D structures
Researcher (PI) Pierre-Jean CORRINGER
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS1, ERC-2017-ADG
Summary Nicotinic acetylcholine receptors (nAChRs) mediate neuronal synaptic transmission and modulation. They contribute to higher brain functions such as cognition and reward and are important drug targets. Recent studies have revealed that these acetylcholine-gated ion channels display an unanticipated conformational plasticity, adopting multiple allosteric states that shape the time course of their electrophysiological response. To date, a single nAChR structure has been solved at high resolution, and our understanding of the conformational transitions remains so far elusive.
To address this challenge, we propose to develop a top-down approach starting from the study of the conformational transitions of nAChRs functionally expressed in cells, and then dissecting the molecular mechanisms on purified proteins. In WP1, we will develop an innovative fluorescence quenching approach to follow the protein motions concomitant with channel opening at the cell membrane. In WP2, we will further exploit this technique on purified proteins, to study the role/requirement of lipids, and their pharmacological crosstalk with allosteric modulators acting at the transmembrane domain. In WP3, the gained knowledge will open original routes to solve 3D structures of nAChRs, in novel conformations and in complex with allosteric modulators. The research will be centered on the major brain nAChRs, primarily the homomeric α7 and also the heteromeric α4β2 nAChRs that are major physiological players and key potential therapeutic targets.
This multidisciplinary project combines electrophysiology, fluorescence, pharmacology, membrane protein biochemistry and structural biology, together with in silico modeling, molecular dynamics and ligand docking. The results will provide fundamental insights into the allosteric mechanisms underlying both nAChR function and its modulation by allosteric modulators that hold promises in therapeutics.
Summary
Nicotinic acetylcholine receptors (nAChRs) mediate neuronal synaptic transmission and modulation. They contribute to higher brain functions such as cognition and reward and are important drug targets. Recent studies have revealed that these acetylcholine-gated ion channels display an unanticipated conformational plasticity, adopting multiple allosteric states that shape the time course of their electrophysiological response. To date, a single nAChR structure has been solved at high resolution, and our understanding of the conformational transitions remains so far elusive.
To address this challenge, we propose to develop a top-down approach starting from the study of the conformational transitions of nAChRs functionally expressed in cells, and then dissecting the molecular mechanisms on purified proteins. In WP1, we will develop an innovative fluorescence quenching approach to follow the protein motions concomitant with channel opening at the cell membrane. In WP2, we will further exploit this technique on purified proteins, to study the role/requirement of lipids, and their pharmacological crosstalk with allosteric modulators acting at the transmembrane domain. In WP3, the gained knowledge will open original routes to solve 3D structures of nAChRs, in novel conformations and in complex with allosteric modulators. The research will be centered on the major brain nAChRs, primarily the homomeric α7 and also the heteromeric α4β2 nAChRs that are major physiological players and key potential therapeutic targets.
This multidisciplinary project combines electrophysiology, fluorescence, pharmacology, membrane protein biochemistry and structural biology, together with in silico modeling, molecular dynamics and ligand docking. The results will provide fundamental insights into the allosteric mechanisms underlying both nAChR function and its modulation by allosteric modulators that hold promises in therapeutics.
Max ERC Funding
2 282 105 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym DYNMECH
Project Dynamic Mechanisms
Researcher (PI) Daniel Ferguson Garrett
Host Institution (HI) FONDATION JEAN-JACQUES LAFFONT,TOULOUSE SCIENCES ECONOMIQUES
Call Details Starting Grant (StG), SH1, ERC-2016-STG
Summary This project studies dynamic mechanisms. By “dynamic mechanisms”, we mean policies to which a principal (e.g., a seller, an employer, or a regulator) can commit to induce the agents (e.g., buyers, employees, or regulated firms) to take the desired actions over time. Several components of the project are envisaged:
- Competition in dynamic mechanisms.
o I propose a competitive setting in which agents (e.g., buyers or workers) learn about the offers of different principals over time. Agents may receive more than one offer at a time, leading to direct competition between mechanisms. Received offers are agents’ private information, permitting strategic delay of acceptance (for instance, an agent may want to wait to evaluate new offers that received in the future).
- Robust predictions for a rich class of stochastic processes.
o We study optimal dynamic mechanisms for agents whose preferences evolve stochastically with time. We develop an approach to partially characterizing these mechanisms which (unlike virtually all of the existing literature) does not depend on ad-hoc restrictions on the stochastic process for preferences.
- Efficient bilateral trade with budget balance: dynamic arrival of traders
o I study bilateral trade with budget balance, when traders (i) arrive over time, and (ii) have preferences which evolve stochastically with time. The project aims at an impossibility result in this setting: contrary to the existing literature which does not account for dynamic arrivals, budget-balanced efficient trade is typically impossible, even for very patient traders.
- Pre-event ticket sales and complementary investments
o We provide a rationale for the early allocation of capacity to customers for events such as flights and concerts based on customers’ demand for pre-event complementary investments (such as booking a hotel or a babysitter). We examine efficient and profit-maximizing mechanisms.
Summary
This project studies dynamic mechanisms. By “dynamic mechanisms”, we mean policies to which a principal (e.g., a seller, an employer, or a regulator) can commit to induce the agents (e.g., buyers, employees, or regulated firms) to take the desired actions over time. Several components of the project are envisaged:
- Competition in dynamic mechanisms.
o I propose a competitive setting in which agents (e.g., buyers or workers) learn about the offers of different principals over time. Agents may receive more than one offer at a time, leading to direct competition between mechanisms. Received offers are agents’ private information, permitting strategic delay of acceptance (for instance, an agent may want to wait to evaluate new offers that received in the future).
- Robust predictions for a rich class of stochastic processes.
o We study optimal dynamic mechanisms for agents whose preferences evolve stochastically with time. We develop an approach to partially characterizing these mechanisms which (unlike virtually all of the existing literature) does not depend on ad-hoc restrictions on the stochastic process for preferences.
- Efficient bilateral trade with budget balance: dynamic arrival of traders
o I study bilateral trade with budget balance, when traders (i) arrive over time, and (ii) have preferences which evolve stochastically with time. The project aims at an impossibility result in this setting: contrary to the existing literature which does not account for dynamic arrivals, budget-balanced efficient trade is typically impossible, even for very patient traders.
- Pre-event ticket sales and complementary investments
o We provide a rationale for the early allocation of capacity to customers for events such as flights and concerts based on customers’ demand for pre-event complementary investments (such as booking a hotel or a babysitter). We examine efficient and profit-maximizing mechanisms.
Max ERC Funding
1 321 625 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym DYSMOIA
Project Dynamic Structural Economic Models: Identification and Estimation
Researcher (PI) Thierry Jean Magnac
Host Institution (HI) FONDATION JEAN-JACQUES LAFFONT,TOULOUSE SCIENCES ECONOMIQUES
Call Details Advanced Grant (AdG), SH1, ERC-2011-ADG_20110406
Summary The objective of this project is to enhance knowledge in the construction, identification and estimation of dynamic structural microeconomic models that are used for policy evaluation. This research proposal is built up having specific economic applications in mind as these applications involve inter-temporal trade-offs for a single or several decision makers. It first seeks to develop original identification results in these applications and attaches special attention to partial identification issues and constructive identification results so as to easily derive estimation techniques. In each specific application, empirical estimates using micro-data will then be used to construct and analyse counterfactuals. The whole sequence of original identification, estimation and prediction results aims at enhancing the quality and credibility of economic policy evaluations.
These research questions will be addressed in frameworks in which dynamic choices are continuous such as the ones regarding human capital investments or discrete such as the college choice decisions. This extends to dynamic games as in the analysis of firms' entry into a market.
This research proposal develops micro-econometric analyses devoted to earning dynamics, consumption smoothing and incomplete markets, firms' entry, school matching mechanisms as well as to the dynamics of undergraduate studies and the dynamics of retirement. It involves studies in labor economics, consumer behavior as well as financial econometrics, empirical industrial organization and the economics of education. One last theme of this project is devoted to research in theoretical econometrics analyzing questions derived from the empirical projects. Each empirical project will cross fertilize others and will feed up theoretical econometric analyses related to point or partial identification in various dimensions. In turn, theoretical analyses will inform identification and estimation in each of those specific applications.
Summary
The objective of this project is to enhance knowledge in the construction, identification and estimation of dynamic structural microeconomic models that are used for policy evaluation. This research proposal is built up having specific economic applications in mind as these applications involve inter-temporal trade-offs for a single or several decision makers. It first seeks to develop original identification results in these applications and attaches special attention to partial identification issues and constructive identification results so as to easily derive estimation techniques. In each specific application, empirical estimates using micro-data will then be used to construct and analyse counterfactuals. The whole sequence of original identification, estimation and prediction results aims at enhancing the quality and credibility of economic policy evaluations.
These research questions will be addressed in frameworks in which dynamic choices are continuous such as the ones regarding human capital investments or discrete such as the college choice decisions. This extends to dynamic games as in the analysis of firms' entry into a market.
This research proposal develops micro-econometric analyses devoted to earning dynamics, consumption smoothing and incomplete markets, firms' entry, school matching mechanisms as well as to the dynamics of undergraduate studies and the dynamics of retirement. It involves studies in labor economics, consumer behavior as well as financial econometrics, empirical industrial organization and the economics of education. One last theme of this project is devoted to research in theoretical econometrics analyzing questions derived from the empirical projects. Each empirical project will cross fertilize others and will feed up theoretical econometric analyses related to point or partial identification in various dimensions. In turn, theoretical analyses will inform identification and estimation in each of those specific applications.
Max ERC Funding
1 722 000 €
Duration
Start date: 2012-06-01, End date: 2018-05-31
Project acronym E-DOHAD
Project Environmentally-induced Developmental Origins of Health and Disease
Researcher (PI) Remy (Fernand) Slama
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS7, ERC-2012-StG_20111109
Summary The DOHaD (Developmental Origins of Health and Disease) hypothesis states that, because of developmental plasticity, in utero and early postnatal stressors can increase chronic disease risk in childhood and later. Alterations of epigenetic marks, impacting on gene expression, are one mechanism that could explain such long-term impact. These hypotheses have so far been tested mostly in animals, and little for atmospheric pollutants, for which animal evidence is scarce.
We aim to characterize the impact of environmental exposures on childhood health. Our focus is on two families of pollutants with a highly prevalent and controllable exposure in humans: atmospheric pollutants and specific high-volume non-persistent chemicals (Bisphenol A, other phenols and phthalates). These pollutants are archetypal of modern life pollutants challenging environmental health research. We will set up a new type of mother-child cohort with early recruitment in pregnancy, intense follow-up (including geolocalisation of subjects with GPS combined with fine-scale air pollution modelling), personal exposure monitoring, repeated collection of biological samples. Transcriptomic analysis, non-invasive clinical examinations (Doppler and ultrasound imaging, ECG, early postnatal evaluation of lung function) will bring clues regarding target functions. This observational approach in humans will be supplemented by an animal experiment aiming at characterizing the impact of in utero exposure to traffic-related atmospheric pollutants on foetal development and health in adulthood, and characterizing target functions and organs more finely than the human study can allow.
E-DOHaD spans over the whole range of environmental health disciplines, with epidemiologic and toxicologic studies being conducted in parallel to ease comparability and results synthesis. E-DOHaD is expected to have far-reaching implications in environmental health research and for public health.
Summary
The DOHaD (Developmental Origins of Health and Disease) hypothesis states that, because of developmental plasticity, in utero and early postnatal stressors can increase chronic disease risk in childhood and later. Alterations of epigenetic marks, impacting on gene expression, are one mechanism that could explain such long-term impact. These hypotheses have so far been tested mostly in animals, and little for atmospheric pollutants, for which animal evidence is scarce.
We aim to characterize the impact of environmental exposures on childhood health. Our focus is on two families of pollutants with a highly prevalent and controllable exposure in humans: atmospheric pollutants and specific high-volume non-persistent chemicals (Bisphenol A, other phenols and phthalates). These pollutants are archetypal of modern life pollutants challenging environmental health research. We will set up a new type of mother-child cohort with early recruitment in pregnancy, intense follow-up (including geolocalisation of subjects with GPS combined with fine-scale air pollution modelling), personal exposure monitoring, repeated collection of biological samples. Transcriptomic analysis, non-invasive clinical examinations (Doppler and ultrasound imaging, ECG, early postnatal evaluation of lung function) will bring clues regarding target functions. This observational approach in humans will be supplemented by an animal experiment aiming at characterizing the impact of in utero exposure to traffic-related atmospheric pollutants on foetal development and health in adulthood, and characterizing target functions and organs more finely than the human study can allow.
E-DOHaD spans over the whole range of environmental health disciplines, with epidemiologic and toxicologic studies being conducted in parallel to ease comparability and results synthesis. E-DOHaD is expected to have far-reaching implications in environmental health research and for public health.
Max ERC Funding
1 499 870 €
Duration
Start date: 2013-02-01, End date: 2018-01-31
Project acronym EARLYLIFE
Project LEARNING TO SURVIVE IN A CHANGING WORLD
Researcher (PI) Henri Weimerskirch
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS8, ERC-2012-ADG_20120314
Summary "This project has the ambitious goal of providing new groundbreaking information on a poorly known, yet critical stage of the life of any animal, the EARLYLIFE. The originality and novelty of EARLYLIFE is to combine two fields in ecology generally considered independently: foraging ecology and demography.With the data obtained for a range of marine predators with contrasted life histories I will be in a position to test several long lasting hypotheses in ecology, in particular on the causes of mortality of young animals, learning abilities, as well as on the significance of delayed maturity in long lived animals. Ultimately the project will allow a robust evaluation of the potential effects of global changes on young animals, and its consequences for population dynamics and conservation of marine air breathing predators.
I will carry out EARLYLIFE in three steps, with my CNRS research group that brings together top researchers with complementary specialties and skills in animal tracking, foraging ecology, demographic modeling. First, using newly developed state of the art developments in bio-telemetry, bio-logging and biochemical markers, I will study for the first time the foraging ecology (movement, habitat, energetic, diet) of 13 species of seabirds and seals during their first years of life at sea after independence. This will allow estimating critical parameters such as the timing and causes of death, the spatial variation in mortality, as well as the extent of improvement in foraging skills at early stages. Second I will model the ontogenic changes occurring during the early stage of life, and contrast them with the foraging abilities of mature animals using our tracking data base. Finally, using our unique long term demographic data base I will examine to what extent, and how, environmental variability affects juvenile mortality, and models the potential effects of climate change and human activities on population dynamics."
Summary
"This project has the ambitious goal of providing new groundbreaking information on a poorly known, yet critical stage of the life of any animal, the EARLYLIFE. The originality and novelty of EARLYLIFE is to combine two fields in ecology generally considered independently: foraging ecology and demography.With the data obtained for a range of marine predators with contrasted life histories I will be in a position to test several long lasting hypotheses in ecology, in particular on the causes of mortality of young animals, learning abilities, as well as on the significance of delayed maturity in long lived animals. Ultimately the project will allow a robust evaluation of the potential effects of global changes on young animals, and its consequences for population dynamics and conservation of marine air breathing predators.
I will carry out EARLYLIFE in three steps, with my CNRS research group that brings together top researchers with complementary specialties and skills in animal tracking, foraging ecology, demographic modeling. First, using newly developed state of the art developments in bio-telemetry, bio-logging and biochemical markers, I will study for the first time the foraging ecology (movement, habitat, energetic, diet) of 13 species of seabirds and seals during their first years of life at sea after independence. This will allow estimating critical parameters such as the timing and causes of death, the spatial variation in mortality, as well as the extent of improvement in foraging skills at early stages. Second I will model the ontogenic changes occurring during the early stage of life, and contrast them with the foraging abilities of mature animals using our tracking data base. Finally, using our unique long term demographic data base I will examine to what extent, and how, environmental variability affects juvenile mortality, and models the potential effects of climate change and human activities on population dynamics."
Max ERC Funding
2 484 000 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym ECCENTRIC
Project Epigenetic challenges in centromere inheritance during the cell cycle
Researcher (PI) Geneviève Almouzni - Pettinotti
Host Institution (HI) INSTITUT CURIE
Call Details Advanced Grant (AdG), LS1, ERC-2009-AdG
Summary Studies concerning the mechanism of DNA replication have advanced our understanding of genetic transmission through multiple cell cycles. Recent work has shed light on possible means to ensure the stable transmission of information beyond just DNA and the concept of epigenetic inheritance has emerged. Considering chromatin-based information, key candidates have arisen as epigenetic marks including DNA and histone modifications, histone variants, non-histone chromatin proteins, nuclear RNA as well as higher-order chromatin organization. Thus, understanding the dynamics and stability of these marks following disruptive events during replication and repair and throughout the cell cycle becomes of critical importance for the maintenance of any given chromatin state. To approach these issues, we propose to study the maintenance of heterochromatin at centromeres, key chromosomal regions for the proper chromosome segregation. Our current goal is to access to the sophisticated mechanisms that have evolved in order to facilitate inheritance of epigenetic marks not only at the replication fork, but also at other stages of the cell cycle, during repair and development. Beyond inheritance of DNA methylation, understanding how inheritance of histone variants and their modifications can be controlled either coupled or not coupled to DNA replication will be a major focus of this project. Our studies will build on the expertise and tools developed over the years in a strategy that integrates molecular, cellular, and biochemical approaches. This will be combined with the use of new technologies to monitor cell cycle (Fucci), protein dynamics (SNAP-Tagging) together with single molecule analysis involving DNA and chromatin combing. We wish to define a possible framework for an understanding of both the stability and reversibility of epigenetic marks and their dynamics at centromeres. These lessons may teach us general principles of inheritance of epigenetic states.
Summary
Studies concerning the mechanism of DNA replication have advanced our understanding of genetic transmission through multiple cell cycles. Recent work has shed light on possible means to ensure the stable transmission of information beyond just DNA and the concept of epigenetic inheritance has emerged. Considering chromatin-based information, key candidates have arisen as epigenetic marks including DNA and histone modifications, histone variants, non-histone chromatin proteins, nuclear RNA as well as higher-order chromatin organization. Thus, understanding the dynamics and stability of these marks following disruptive events during replication and repair and throughout the cell cycle becomes of critical importance for the maintenance of any given chromatin state. To approach these issues, we propose to study the maintenance of heterochromatin at centromeres, key chromosomal regions for the proper chromosome segregation. Our current goal is to access to the sophisticated mechanisms that have evolved in order to facilitate inheritance of epigenetic marks not only at the replication fork, but also at other stages of the cell cycle, during repair and development. Beyond inheritance of DNA methylation, understanding how inheritance of histone variants and their modifications can be controlled either coupled or not coupled to DNA replication will be a major focus of this project. Our studies will build on the expertise and tools developed over the years in a strategy that integrates molecular, cellular, and biochemical approaches. This will be combined with the use of new technologies to monitor cell cycle (Fucci), protein dynamics (SNAP-Tagging) together with single molecule analysis involving DNA and chromatin combing. We wish to define a possible framework for an understanding of both the stability and reversibility of epigenetic marks and their dynamics at centromeres. These lessons may teach us general principles of inheritance of epigenetic states.
Max ERC Funding
2 490 483 €
Duration
Start date: 2010-06-01, End date: 2015-12-31
Project acronym ECOFEED
Project Altered eco-evolutionary feedbacks in a future climate
Researcher (PI) Julien COTE
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS8, ERC-2018-COG
Summary Current scenarios predict an accelerated biodiversity erosion with climate change. However, uncertainties in predictions remain large because the multitude of climate change effects from genes to ecosystems and their interdependencies are still overlooked. This incomplete vision hampers the development of effective mitigation strategies to sustain biodiversity.
Climate change can directly modify the phenotype and performance of individuals through phenotypic plasticity and evolution on contemporary time scales. The microevolution of keystone species can spread throughout the whole ecological network due to changes in species interactions and further translate into an altered ecosystem functioning. Conversely, direct impacts on communities and ecosystems can have ripple effects on the phenotypic distribution and evolution of all species of ecological networks.
Climate-driven changes at individual and population levels can shape community composition and ecosystem functioning, and vice versa, altering eco-evolutionary feedbacks, namely the reciprocal interactions between ecological and evolutionary processes. Climate-driven ecological and evolutionary dynamics are yet often investigated separately. The role of eco-evolutionary feedbacks in climate change impacts on biological systems therefore hinges on little concrete empirical evidence contrasting with a profuse theoretical development.
ECOFEED will investigate climate-dependent eco-evolutionary feedbacks using a 6 year-long realistic warming experiment reproducing natural conditions and thus allowing for both evolutionary and ecological dynamics to occur under a predicted climate change scenario. Complementary laboratory experiments will quantify reciprocal impacts of climate-dependent evolutionary and ecological changes on each other. ECOFEED will provide unprecedented insights on the eco-evolutionary feedbacks in a future climate and will ultimately help refine predictions on the future of biodiversity.
Summary
Current scenarios predict an accelerated biodiversity erosion with climate change. However, uncertainties in predictions remain large because the multitude of climate change effects from genes to ecosystems and their interdependencies are still overlooked. This incomplete vision hampers the development of effective mitigation strategies to sustain biodiversity.
Climate change can directly modify the phenotype and performance of individuals through phenotypic plasticity and evolution on contemporary time scales. The microevolution of keystone species can spread throughout the whole ecological network due to changes in species interactions and further translate into an altered ecosystem functioning. Conversely, direct impacts on communities and ecosystems can have ripple effects on the phenotypic distribution and evolution of all species of ecological networks.
Climate-driven changes at individual and population levels can shape community composition and ecosystem functioning, and vice versa, altering eco-evolutionary feedbacks, namely the reciprocal interactions between ecological and evolutionary processes. Climate-driven ecological and evolutionary dynamics are yet often investigated separately. The role of eco-evolutionary feedbacks in climate change impacts on biological systems therefore hinges on little concrete empirical evidence contrasting with a profuse theoretical development.
ECOFEED will investigate climate-dependent eco-evolutionary feedbacks using a 6 year-long realistic warming experiment reproducing natural conditions and thus allowing for both evolutionary and ecological dynamics to occur under a predicted climate change scenario. Complementary laboratory experiments will quantify reciprocal impacts of climate-dependent evolutionary and ecological changes on each other. ECOFEED will provide unprecedented insights on the eco-evolutionary feedbacks in a future climate and will ultimately help refine predictions on the future of biodiversity.
Max ERC Funding
1 983 565 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym ECOMATCH
Project Economics of Matching Markets: Theoretical and Empirical Investigations
Researcher (PI) Alfred Galichon
Host Institution (HI) FONDATION NATIONALE DES SCIENCES POLITIQUES
Call Details Starting Grant (StG), SH1, ERC-2012-StG_20111124
Summary This project offers a theoretical and empirical investigation of matching markets. Matching is, broadly speaking, the study of complementarities, which explains the formation of coalitions. Matching models are found in many applied fields within Economics: Labour Economics, Family Economics, Consumer theory of differentiated goods (hedonic models), Trade, etc. Desirable properties of these coalitions, such as stability, lead to testable implications of the surplus that individuals generate in a match, allowing for structural estimation of matching models.
The goal of this proposal is to expand the frontiers of the theory of matching to design a very general and highly flexible model of matching that will lend itself to estimation and thus lead to empirical findings in various fields of Economics. Based on promising work initiated by the PI, this proposal seeks to bridge the gap between the theory and the empirics of matching markets that was traditionally observed in this literature.
Particular focus will be given to situations where stable outcomes may not exist (such as unipartite, or one-to-many matching models), frictions, taxes. In these cases, a thorough investigation is carried on what solution concept should be used, and what are the testable implications.
Applications will be given to various empirical issues or policy relevant questions such as:
- The nature of the complementarities between senior and junior employees within teams,
- The role played by the marriage market in the problem of rural depletion in China,
- The impact of CEO risk aversion on assignment to firms, and on the CEO compensation package,
- The pricing of attributes of French wines.
Summary
This project offers a theoretical and empirical investigation of matching markets. Matching is, broadly speaking, the study of complementarities, which explains the formation of coalitions. Matching models are found in many applied fields within Economics: Labour Economics, Family Economics, Consumer theory of differentiated goods (hedonic models), Trade, etc. Desirable properties of these coalitions, such as stability, lead to testable implications of the surplus that individuals generate in a match, allowing for structural estimation of matching models.
The goal of this proposal is to expand the frontiers of the theory of matching to design a very general and highly flexible model of matching that will lend itself to estimation and thus lead to empirical findings in various fields of Economics. Based on promising work initiated by the PI, this proposal seeks to bridge the gap between the theory and the empirics of matching markets that was traditionally observed in this literature.
Particular focus will be given to situations where stable outcomes may not exist (such as unipartite, or one-to-many matching models), frictions, taxes. In these cases, a thorough investigation is carried on what solution concept should be used, and what are the testable implications.
Applications will be given to various empirical issues or policy relevant questions such as:
- The nature of the complementarities between senior and junior employees within teams,
- The role played by the marriage market in the problem of rural depletion in China,
- The impact of CEO risk aversion on assignment to firms, and on the CEO compensation package,
- The pricing of attributes of French wines.
Max ERC Funding
1 119 000 €
Duration
Start date: 2013-01-01, End date: 2018-09-30
Project acronym Econ_Prejudice
Project The Economics of Ethnic Prejudice
Researcher (PI) Ekaterina Zhuravskaya
Host Institution (HI) ECOLE D'ECONOMIE DE PARIS
Call Details Consolidator Grant (CoG), SH1, ERC-2014-CoG
Summary Why do ethnic differences matter in some cases and not in others? What determines the strength of ethnic self-identification? This question is central to understanding the consequences of ethnic divisions for conflict and economic development and their policy implications but it was neglected by economic research until now. This project aims at filling this gap by endogenizing ethnic identity. We study how the salience of ethnic differences depends on economic and social context and policies of nation building. Our research program is organized around 3 pillars focusing on social, economic, and political determinants of ethnic tensions, respectively. The first pillar tests social psychology theories of ethnic identity using natural experiments, generated by forced mass movements of ethnic groups in Eastern Europe and from Eastern Europe to Central Asia as a result of WWII. The second pillar studies how market interactions between representatives of different ethnic groups and, in particular, ethnic occupational segregation affects ethnic tensions in the context of historical anti-Jewish violence following agro-climatic income shocks in the 19th and 20th century Eastern Europe. The third pillar focuses on the effects of political manipulation on ethnic conflict in the context of the historical experiment of nation building in Central Asia. It studies how political empowerment of a certain ethnic elite in a multi-ethnic traditional society coupled with a set of nation-building policies affects ethnic conflicts depending on the pre-existing ethnic mix and the distribution of political power among ethnic elites. This research will shed light on factors that make ethnic diversity important for conflict and economic development.
Summary
Why do ethnic differences matter in some cases and not in others? What determines the strength of ethnic self-identification? This question is central to understanding the consequences of ethnic divisions for conflict and economic development and their policy implications but it was neglected by economic research until now. This project aims at filling this gap by endogenizing ethnic identity. We study how the salience of ethnic differences depends on economic and social context and policies of nation building. Our research program is organized around 3 pillars focusing on social, economic, and political determinants of ethnic tensions, respectively. The first pillar tests social psychology theories of ethnic identity using natural experiments, generated by forced mass movements of ethnic groups in Eastern Europe and from Eastern Europe to Central Asia as a result of WWII. The second pillar studies how market interactions between representatives of different ethnic groups and, in particular, ethnic occupational segregation affects ethnic tensions in the context of historical anti-Jewish violence following agro-climatic income shocks in the 19th and 20th century Eastern Europe. The third pillar focuses on the effects of political manipulation on ethnic conflict in the context of the historical experiment of nation building in Central Asia. It studies how political empowerment of a certain ethnic elite in a multi-ethnic traditional society coupled with a set of nation-building policies affects ethnic conflicts depending on the pre-existing ethnic mix and the distribution of political power among ethnic elites. This research will shed light on factors that make ethnic diversity important for conflict and economic development.
Max ERC Funding
1 598 308 €
Duration
Start date: 2015-11-01, End date: 2020-10-31