Project acronym ABATSYNAPSE
Project Evolution of Alzheimer’s Disease: From dynamics of single synapses to memory loss
Researcher (PI) Inna Slutsky
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS5, ERC-2011-StG_20101109
Summary A persistent challenge in unravelling mechanisms that regulate memory function is how to bridge the gap between inter-molecular dynamics of single proteins, activity of individual synapses and emerging properties of neuronal circuits. The prototype condition of disintegrating neuronal circuits is Alzheimer’s Disease (AD). Since the early time of Alois Alzheimer at the turn of the 20th century, scientists have been searching for a molecular entity that is in the roots of the cognitive deficits. Although diverse lines of evidence suggest that the amyloid-beta peptide (Abeta) plays a central role in synaptic dysfunctions of AD, several key questions remain unresolved. First, endogenous Abeta peptides are secreted by neurons throughout life, but their physiological functions are largely unknown. Second, experience-dependent physiological mechanisms that initiate the changes in Abeta composition in sporadic, the most frequent form of AD, are unidentified. And finally, molecular mechanisms that trigger Abeta-induced synaptic failure and memory decline remain elusive.
To target these questions, I propose to develop an integrative approach to correlate structure and function at the level of single synapses in hippocampal circuits. State-of-the-art techniques will enable the simultaneous real-time visualization of inter-molecular dynamics within signalling complexes and functional synaptic modifications. Utilizing FRET spectroscopy, high-resolution optical imaging, electrophysiology, molecular biology and biochemistry we will determine the casual relationship between ongoing neuronal activity, temporo-spatial dynamics and molecular composition of Abeta, structural rearrangements within the Abeta signalling complexes and plasticity of single synapses and whole networks. The proposed research will elucidate fundamental principles of neuronal circuits function and identify critical steps that initiate primary synaptic dysfunctions at the very early stages of sporadic AD.
Summary
A persistent challenge in unravelling mechanisms that regulate memory function is how to bridge the gap between inter-molecular dynamics of single proteins, activity of individual synapses and emerging properties of neuronal circuits. The prototype condition of disintegrating neuronal circuits is Alzheimer’s Disease (AD). Since the early time of Alois Alzheimer at the turn of the 20th century, scientists have been searching for a molecular entity that is in the roots of the cognitive deficits. Although diverse lines of evidence suggest that the amyloid-beta peptide (Abeta) plays a central role in synaptic dysfunctions of AD, several key questions remain unresolved. First, endogenous Abeta peptides are secreted by neurons throughout life, but their physiological functions are largely unknown. Second, experience-dependent physiological mechanisms that initiate the changes in Abeta composition in sporadic, the most frequent form of AD, are unidentified. And finally, molecular mechanisms that trigger Abeta-induced synaptic failure and memory decline remain elusive.
To target these questions, I propose to develop an integrative approach to correlate structure and function at the level of single synapses in hippocampal circuits. State-of-the-art techniques will enable the simultaneous real-time visualization of inter-molecular dynamics within signalling complexes and functional synaptic modifications. Utilizing FRET spectroscopy, high-resolution optical imaging, electrophysiology, molecular biology and biochemistry we will determine the casual relationship between ongoing neuronal activity, temporo-spatial dynamics and molecular composition of Abeta, structural rearrangements within the Abeta signalling complexes and plasticity of single synapses and whole networks. The proposed research will elucidate fundamental principles of neuronal circuits function and identify critical steps that initiate primary synaptic dysfunctions at the very early stages of sporadic AD.
Max ERC Funding
2 000 000 €
Duration
Start date: 2011-12-01, End date: 2017-09-30
Project acronym ANGIOFAT
Project New mechanisms of angiogenesis modulators in switching between white and brown adipose tissues
Researcher (PI) Yihai Cao
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Advanced Grant (AdG), LS4, ERC-2009-AdG
Summary Understanding the molecular mechanisms underlying adipose blood vessel growth or regression opens new fundamentally insight into novel therapeutic options for the treatment of obesity and its related metabolic diseases such as type 2 diabetes and cancer. Unlike any other tissues in the body, the adipose tissue constantly experiences expansion and shrinkage throughout the adult life. Adipocytes in the white adipose tissue have the ability to switch into metabolically highly active brown-like adipocytes. Brown adipose tissue (BAT) contains significantly higher numbers of microvessels than white adipose tissue (WAT) in order to adopt the high rates of metabolism. Thus, an angiogenic phenotype has to be switched on during the transition from WAT into BAT. We have found that acclimation of mice in cold could induce transition from inguinal and epidedymal WAT into BAT by upregulation of angiogenic factor expression and down-regulations of angiogenesis inhibitors (Xue et al, Cell Metabolism, 2009). The transition from WAT into BAT is dependent on vascular endothelial growth factor (VEGF) that primarily targets on vascular endothelial cells via a tissue hypoxia-independent mechanism. VEGF blockade significantly alters adipose tissue metabolism. In another genetic model, we show similar findings that angiogenesis is crucial to mediate the transition from WAT into BAT (Xue et al, PNAS, 2008). Here we propose that the vascular tone determines the metabolic switch between WAT and BAT. Characterization of these novel angiogenic pathways may reveal new mechanisms underlying development of obesity- and metabolism-related disease complications and may define novel therapeutic targets. Thus, the benefit of this research proposal is enormous and is aimed to treat the most common and highly risk human health conditions in the modern time.
Summary
Understanding the molecular mechanisms underlying adipose blood vessel growth or regression opens new fundamentally insight into novel therapeutic options for the treatment of obesity and its related metabolic diseases such as type 2 diabetes and cancer. Unlike any other tissues in the body, the adipose tissue constantly experiences expansion and shrinkage throughout the adult life. Adipocytes in the white adipose tissue have the ability to switch into metabolically highly active brown-like adipocytes. Brown adipose tissue (BAT) contains significantly higher numbers of microvessels than white adipose tissue (WAT) in order to adopt the high rates of metabolism. Thus, an angiogenic phenotype has to be switched on during the transition from WAT into BAT. We have found that acclimation of mice in cold could induce transition from inguinal and epidedymal WAT into BAT by upregulation of angiogenic factor expression and down-regulations of angiogenesis inhibitors (Xue et al, Cell Metabolism, 2009). The transition from WAT into BAT is dependent on vascular endothelial growth factor (VEGF) that primarily targets on vascular endothelial cells via a tissue hypoxia-independent mechanism. VEGF blockade significantly alters adipose tissue metabolism. In another genetic model, we show similar findings that angiogenesis is crucial to mediate the transition from WAT into BAT (Xue et al, PNAS, 2008). Here we propose that the vascular tone determines the metabolic switch between WAT and BAT. Characterization of these novel angiogenic pathways may reveal new mechanisms underlying development of obesity- and metabolism-related disease complications and may define novel therapeutic targets. Thus, the benefit of this research proposal is enormous and is aimed to treat the most common and highly risk human health conditions in the modern time.
Max ERC Funding
2 411 547 €
Duration
Start date: 2010-03-01, End date: 2015-02-28
Project acronym ARISE
Project The Ecology of Antibiotic Resistance
Researcher (PI) Roy Kishony
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS8, ERC-2011-StG_20101109
Summary Main goal. We aim to understand the puzzling coexistence of antibiotic-resistant and antibiotic-sensitive species in natural soil environments, using novel quantitative experimental techniques and mathematical analysis. The ecological insights gained will be translated into novel treatment strategies for combating antibiotic resistance.
Background. Microbial soil ecosystems comprise communities of species interacting through copious secretion of antibiotics and other chemicals. Defence mechanisms, i.e. resistance to antibiotics, are ubiquitous in these wild communities. However, in sharp contrast to clinical settings, resistance does not take over the population. Our hypothesis is that the ecological setting provides natural mechanisms that keep antibiotic resistance in check. We are motivated by our recent finding that specific antibiotic combinations can generate selection against resistance and that soil microbial strains produce compounds that directly target antibiotic resistant mechanisms.
Approaches. We will: (1) Isolate natural bacterial species from individual grains of soil, characterize their ability to produce and resist antibiotics and identify the spatial scale for correlations between resistance and production. (2) Systematically measure interactions between species and identify interaction patterns enriched in co-existing communities derived from the same grain of soil. (3) Introducing fluorescently-labelled resistant and sensitive strains into natural soil, we will measure the fitness cost and benefit of antibiotic resistance in situ and identify natural compounds that select against resistance. (4) Test whether such “selection-inverting” compounds can slow evolution of resistance to antibiotics in continuous culture experiments.
Conclusions. These findings will provide insights into the ecological processes that keep antibiotic resistance in check, and will suggest novel antimicrobial treatment strategies.
Summary
Main goal. We aim to understand the puzzling coexistence of antibiotic-resistant and antibiotic-sensitive species in natural soil environments, using novel quantitative experimental techniques and mathematical analysis. The ecological insights gained will be translated into novel treatment strategies for combating antibiotic resistance.
Background. Microbial soil ecosystems comprise communities of species interacting through copious secretion of antibiotics and other chemicals. Defence mechanisms, i.e. resistance to antibiotics, are ubiquitous in these wild communities. However, in sharp contrast to clinical settings, resistance does not take over the population. Our hypothesis is that the ecological setting provides natural mechanisms that keep antibiotic resistance in check. We are motivated by our recent finding that specific antibiotic combinations can generate selection against resistance and that soil microbial strains produce compounds that directly target antibiotic resistant mechanisms.
Approaches. We will: (1) Isolate natural bacterial species from individual grains of soil, characterize their ability to produce and resist antibiotics and identify the spatial scale for correlations between resistance and production. (2) Systematically measure interactions between species and identify interaction patterns enriched in co-existing communities derived from the same grain of soil. (3) Introducing fluorescently-labelled resistant and sensitive strains into natural soil, we will measure the fitness cost and benefit of antibiotic resistance in situ and identify natural compounds that select against resistance. (4) Test whether such “selection-inverting” compounds can slow evolution of resistance to antibiotics in continuous culture experiments.
Conclusions. These findings will provide insights into the ecological processes that keep antibiotic resistance in check, and will suggest novel antimicrobial treatment strategies.
Max ERC Funding
1 900 000 €
Duration
Start date: 2012-09-01, End date: 2018-08-31
Project acronym BATESON
Project Dissecting genotype-phenotype relationships using high-throughput genomics and carefully selected study populations
Researcher (PI) Leif Andersson
Host Institution (HI) UPPSALA UNIVERSITET
Call Details Advanced Grant (AdG), LS2, ERC-2011-ADG_20110310
Summary A major aim in genome research is to reveal how genetic variation affects phenotypic variation. Here I propose to use high-throughput genomics (whole genome sequencing, transcriptome and epigenome analysis) to screen carefully selected study populations where the chances are particularly favourable to obtain novel insight into genotype-phenotype relationships. The ambition is to take discoveries all the way from phenotypic characterization to the identification of the genes and the actual genetic variant causing a phenotypic effect and to understanding the underlying functional mechanisms. The program will involve a fish (the Atlantic herring), a bird (the domestic chicken) and a mammal (the European rabbit). The Atlantic herring will be studied because it provides unique opportunities to study the genetics of adaptation in a natural population and because of the possibilities to revolutionize the fishery management of this economically important marine fish. We will generate a draft assembly of the herring genome and then perform whole genome resequencing of different populations to reveal the population structure and the loci underlying genetic adaptation. The European rabbit is an excellent model for studying the genetics of speciation due to the presence of two distinct subspecies on the Iberian Peninsula. The domestication of the rabbit is also particularly interesting because it is a recent event (about 1500 years ago) and it is well established that domestication happened from the wild rabbit population in southern France. Finally, the domestic chicken provides excellent opportunities for in depth functional studies since it is both a domestic animal harbouring a rich genetic diversity and an experimental organism.
(BATESON is the acronym for this proposal because Bateson (1902) pioneered the study of genotype-phenotype relationships in animals and used the chicken for this work.)
Summary
A major aim in genome research is to reveal how genetic variation affects phenotypic variation. Here I propose to use high-throughput genomics (whole genome sequencing, transcriptome and epigenome analysis) to screen carefully selected study populations where the chances are particularly favourable to obtain novel insight into genotype-phenotype relationships. The ambition is to take discoveries all the way from phenotypic characterization to the identification of the genes and the actual genetic variant causing a phenotypic effect and to understanding the underlying functional mechanisms. The program will involve a fish (the Atlantic herring), a bird (the domestic chicken) and a mammal (the European rabbit). The Atlantic herring will be studied because it provides unique opportunities to study the genetics of adaptation in a natural population and because of the possibilities to revolutionize the fishery management of this economically important marine fish. We will generate a draft assembly of the herring genome and then perform whole genome resequencing of different populations to reveal the population structure and the loci underlying genetic adaptation. The European rabbit is an excellent model for studying the genetics of speciation due to the presence of two distinct subspecies on the Iberian Peninsula. The domestication of the rabbit is also particularly interesting because it is a recent event (about 1500 years ago) and it is well established that domestication happened from the wild rabbit population in southern France. Finally, the domestic chicken provides excellent opportunities for in depth functional studies since it is both a domestic animal harbouring a rich genetic diversity and an experimental organism.
(BATESON is the acronym for this proposal because Bateson (1902) pioneered the study of genotype-phenotype relationships in animals and used the chicken for this work.)
Max ERC Funding
2 300 000 €
Duration
Start date: 2012-05-01, End date: 2017-04-30
Project acronym BBBARRIER
Project Mechanisms of regulation of the blood-brain barrier; towards opening and closing the barrier on demand
Researcher (PI) Björn Christer Betsholtz
Host Institution (HI) UPPSALA UNIVERSITET
Call Details Advanced Grant (AdG), LS4, ERC-2011-ADG_20110310
Summary In the bone-enclosed CNS, increased vascular permeability may cause life-threatening tissue swelling, and/or ischemia and inflammation which compromise tissue repair after trauma or stroke. The brain vasculature possesses several unique features collectively named the blood-brain barrier (BBB) in which passive permeability is almost completely abolished and replaced by a complex of specific transport mechanisms. The BBB is necessary to uphold the specific milieu necessary for neuronal function. Whereas breakdown of the BBB is part of many CNS diseases, including stroke, neuroinflammation, trauma and neurodegenerative disorders, its molecular mechanisms and consequences are unclear and debated. Conversely, the intact BBB is a huge obstacle for drug delivery to the brain. Research on the BBB therefore has two seemingly opposing aims: 1) to seal a damaged BBB and protect the brain from toxic blood products, and 2) to open the BBB “on demand” for drug delivery. A major problem in the BBB field has been the lack of in vivo animal models for molecular and functional studies. So far, available in vitro models are not recapitulating the in vivo BBB. Our recent work on mouse models lacking pericytes, a BBB-associated cell type, demonstrates a specific role for pericytes in the development and regulation of the mammalian BBB. These animal models are the first ones showing a general and significant BBB impairment in adulthood, and as such they provide a unique opportunity to address molecular mechanisms of BBB disruption in disease and in drug transport across the BBB. Importantly, the new models and tools that we have developed allow us to search for relevant druggable mechanisms and molecular targets in the BBB. The long-term goals of this proposal are to develop molecular strategies and tools to open and close the BBB “on demand” for drug delivery to the CNS, and to explore the importance and mechanisms of BBB dysfunction in neurodegenerative diseases and stroke.
Summary
In the bone-enclosed CNS, increased vascular permeability may cause life-threatening tissue swelling, and/or ischemia and inflammation which compromise tissue repair after trauma or stroke. The brain vasculature possesses several unique features collectively named the blood-brain barrier (BBB) in which passive permeability is almost completely abolished and replaced by a complex of specific transport mechanisms. The BBB is necessary to uphold the specific milieu necessary for neuronal function. Whereas breakdown of the BBB is part of many CNS diseases, including stroke, neuroinflammation, trauma and neurodegenerative disorders, its molecular mechanisms and consequences are unclear and debated. Conversely, the intact BBB is a huge obstacle for drug delivery to the brain. Research on the BBB therefore has two seemingly opposing aims: 1) to seal a damaged BBB and protect the brain from toxic blood products, and 2) to open the BBB “on demand” for drug delivery. A major problem in the BBB field has been the lack of in vivo animal models for molecular and functional studies. So far, available in vitro models are not recapitulating the in vivo BBB. Our recent work on mouse models lacking pericytes, a BBB-associated cell type, demonstrates a specific role for pericytes in the development and regulation of the mammalian BBB. These animal models are the first ones showing a general and significant BBB impairment in adulthood, and as such they provide a unique opportunity to address molecular mechanisms of BBB disruption in disease and in drug transport across the BBB. Importantly, the new models and tools that we have developed allow us to search for relevant druggable mechanisms and molecular targets in the BBB. The long-term goals of this proposal are to develop molecular strategies and tools to open and close the BBB “on demand” for drug delivery to the CNS, and to explore the importance and mechanisms of BBB dysfunction in neurodegenerative diseases and stroke.
Max ERC Funding
2 499 427 €
Duration
Start date: 2012-08-01, End date: 2017-07-31
Project acronym BRAINGAIN
Project NOVEL STRATEGIES FOR BRAIN REGENERATION
Researcher (PI) Andras Simon
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Starting Grant (StG), LS3, ERC-2011-StG_20101109
Summary In contrast to mammals, newts possess exceptional capacities among vertebrates to rebuild complex structures, such as the brain. Our goal is to bridge the gap in the regenerative outcomes between newts and mammals. My group has made significant contributions towards this goal. We created a novel experimental system, which recapitulates central features of Parkinson’s disease in newts, and provides a unique model for understanding regeneration in the adult midbrain. We showed an unexpected but key feature of the newt brain that it is akin to the mammalian brain in terms of the extent of homeostatic cell turn over, but distinct in terms of its injury response, showing the regenerative capacity of the adult vertebrate brain by activating neurogenesis in normally quiescent regions. Further we established a critical role for the neurotransmitter dopamine in controlling quiescence in the midbrain, thereby preventing neurogenesis during homeostasis and terminating neurogenesis once the correct number of neurons has been produced during regeneration. Here we aim to identify key molecular pathways that regulate adult neurogenesis, to define lineage relationships between neuronal stem and progenitor cells, and to identify essential differences between newts and mammals. We will combine pharmacological modulation of neurotransmitter signaling with extensive cellular fate mapping approaches, and molecular manipulations. Ultimately we will test hypotheses derived from newt studies with mammalian systems including newt/mouse cross species complementation approaches. We expect that our findings will provide new regenerative strategies, and reveal fundamental aspects of cell fate determination, tissue growth, and tissue maintenance in normal and pathological conditions.
Summary
In contrast to mammals, newts possess exceptional capacities among vertebrates to rebuild complex structures, such as the brain. Our goal is to bridge the gap in the regenerative outcomes between newts and mammals. My group has made significant contributions towards this goal. We created a novel experimental system, which recapitulates central features of Parkinson’s disease in newts, and provides a unique model for understanding regeneration in the adult midbrain. We showed an unexpected but key feature of the newt brain that it is akin to the mammalian brain in terms of the extent of homeostatic cell turn over, but distinct in terms of its injury response, showing the regenerative capacity of the adult vertebrate brain by activating neurogenesis in normally quiescent regions. Further we established a critical role for the neurotransmitter dopamine in controlling quiescence in the midbrain, thereby preventing neurogenesis during homeostasis and terminating neurogenesis once the correct number of neurons has been produced during regeneration. Here we aim to identify key molecular pathways that regulate adult neurogenesis, to define lineage relationships between neuronal stem and progenitor cells, and to identify essential differences between newts and mammals. We will combine pharmacological modulation of neurotransmitter signaling with extensive cellular fate mapping approaches, and molecular manipulations. Ultimately we will test hypotheses derived from newt studies with mammalian systems including newt/mouse cross species complementation approaches. We expect that our findings will provide new regenerative strategies, and reveal fundamental aspects of cell fate determination, tissue growth, and tissue maintenance in normal and pathological conditions.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym CARDIOPREVENT
Project INTEGRATION OF GENOMICS AND CARDIOMETABOLIC PLASMA BIOMARKERS FOR IMPROVED PREDICTION AND PRIMARY PREVENTION OF CARDIOVASCULAR DISEASE
Researcher (PI) Olle Sten Melander
Host Institution (HI) LUNDS UNIVERSITET
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary "By taking advantage of great experience in genetic and cardiovascular epidemiology and some of the largest cohorts in the world including 60 000 unique individuals, the applicant aims at (1) improving CVD risk prediction and (2) identifying mechanisms causally related to CVD development in order to provide novel targets for drug discovery and targeted life style interventions for use in primary prevention.
In SUBPROJECT 1 we aim at identifying disease causing alleles of loci implicated in CVD by Genome Wide Association Studies (GWAS) and to identify rare alleles with large impact on human CVD. We thus perform whole exome and targeted sequencing in early CVD cases and healthy controls and evaluate all identified variants by relating them to incident CVD in 60.000 individuals. Further, we will create a score of all validated CVD gene variants and test whether such a score improves clinical risk assessment over and above traditional risk factors.
In SUBPROJECT 2 we test whether the plasma metabolome- a phenotype representing the product of dietary intake and inherent (e.g. genetic) metabolism- differs between incident CVD cases and controls and between individuals with high and low CVD genetic risk. We further test whether a life style intervention differentially alters the plasma metabolome between individuals with high and low CVD genetic risk. Finally, we will elucidate the mechanisms underlying CVD genetic associations by testing whether myocardial expression of such genes are affected by experimental myocardial infarction (MI) and whether heart function, MI size and the plasma metabolome are affected by adenoviral myocardial CVD gene transfer in rats.
In SUBPROJECT 3 we test whether glucose metabolism and CVD risk factors can be ameliorated by suppressing vasopressin (VP) by increased water intake in humans. Finally, we test which of the 3 VP receptors is responsible for adverse glucometabolic VP effects in rats by specific VP receptor pharmacological studies."
Summary
"By taking advantage of great experience in genetic and cardiovascular epidemiology and some of the largest cohorts in the world including 60 000 unique individuals, the applicant aims at (1) improving CVD risk prediction and (2) identifying mechanisms causally related to CVD development in order to provide novel targets for drug discovery and targeted life style interventions for use in primary prevention.
In SUBPROJECT 1 we aim at identifying disease causing alleles of loci implicated in CVD by Genome Wide Association Studies (GWAS) and to identify rare alleles with large impact on human CVD. We thus perform whole exome and targeted sequencing in early CVD cases and healthy controls and evaluate all identified variants by relating them to incident CVD in 60.000 individuals. Further, we will create a score of all validated CVD gene variants and test whether such a score improves clinical risk assessment over and above traditional risk factors.
In SUBPROJECT 2 we test whether the plasma metabolome- a phenotype representing the product of dietary intake and inherent (e.g. genetic) metabolism- differs between incident CVD cases and controls and between individuals with high and low CVD genetic risk. We further test whether a life style intervention differentially alters the plasma metabolome between individuals with high and low CVD genetic risk. Finally, we will elucidate the mechanisms underlying CVD genetic associations by testing whether myocardial expression of such genes are affected by experimental myocardial infarction (MI) and whether heart function, MI size and the plasma metabolome are affected by adenoviral myocardial CVD gene transfer in rats.
In SUBPROJECT 3 we test whether glucose metabolism and CVD risk factors can be ameliorated by suppressing vasopressin (VP) by increased water intake in humans. Finally, we test which of the 3 VP receptors is responsible for adverse glucometabolic VP effects in rats by specific VP receptor pharmacological studies."
Max ERC Funding
1 500 000 €
Duration
Start date: 2011-12-01, End date: 2016-11-30
Project acronym CELLREPROGRAMMING
Project Uncovering the Mechanisms of Epigenetic Reprogramming of Pluripotent and Somatic Cell States
Researcher (PI) Yaqub Hanna
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS3, ERC-2011-StG_20101109
Summary The generation of animals by nuclear transfer demonstrated that the epigenetic state of somatic cells could be reset to an embryonic state, capable of directing the development of a new organism. The nuclear cloning technology is of interest for transplantation medicine, but any application is hampered by the inefficiency and ethical problems. A breakthrough solving these issues has been the in vitro derivation of reprogrammed Induced Pluripotent Stem “iPS” cells by the ectopic expression of defined transcription factors in somatic cells. iPS cells recapitulate all defining features of embryo-derived pluripotent stem cells, including the ability to differentiate into all somatic cell types. Further, recent publications have demonstrated the ability to directly trans-differentiate somatic cell types by ectopic expression of lineage specification factors. Thus, it is becoming increasingly clear that an ultimate goal in the stem cell field is to enable scientists to have the power to safely manipulate somatic cells by “reprogramming” their behavior at will. However, to frame this challenge, we must understand the basic mechanisms underlying the generation of reprogrammed cells in parallel to designing strategies for their medical application and their use in human disease specific research. In this ERC Starting Grant proposal, I describe comprehensive lines of experimentation that I plan to conduct in my new lab scheduled to open in April 2011 at the Weizmann Institute of Science. We will utilize exacting transgenic mammalian models and high throughput sequencing and genomic screening tools for in depth characterization of the molecular “rules” of rewiring the epigenome of somatic and pluripotent cell states. The proposed research endeavors will not only contribute to the development of safer strategies for cell reprogramming, but will also help decipher how diverse gene expression programs lead to cellular specification during normal development.
Summary
The generation of animals by nuclear transfer demonstrated that the epigenetic state of somatic cells could be reset to an embryonic state, capable of directing the development of a new organism. The nuclear cloning technology is of interest for transplantation medicine, but any application is hampered by the inefficiency and ethical problems. A breakthrough solving these issues has been the in vitro derivation of reprogrammed Induced Pluripotent Stem “iPS” cells by the ectopic expression of defined transcription factors in somatic cells. iPS cells recapitulate all defining features of embryo-derived pluripotent stem cells, including the ability to differentiate into all somatic cell types. Further, recent publications have demonstrated the ability to directly trans-differentiate somatic cell types by ectopic expression of lineage specification factors. Thus, it is becoming increasingly clear that an ultimate goal in the stem cell field is to enable scientists to have the power to safely manipulate somatic cells by “reprogramming” their behavior at will. However, to frame this challenge, we must understand the basic mechanisms underlying the generation of reprogrammed cells in parallel to designing strategies for their medical application and their use in human disease specific research. In this ERC Starting Grant proposal, I describe comprehensive lines of experimentation that I plan to conduct in my new lab scheduled to open in April 2011 at the Weizmann Institute of Science. We will utilize exacting transgenic mammalian models and high throughput sequencing and genomic screening tools for in depth characterization of the molecular “rules” of rewiring the epigenome of somatic and pluripotent cell states. The proposed research endeavors will not only contribute to the development of safer strategies for cell reprogramming, but will also help decipher how diverse gene expression programs lead to cellular specification during normal development.
Max ERC Funding
1 960 000 €
Duration
Start date: 2011-11-01, End date: 2016-10-31
Project acronym CM TURNOVER
Project Uncovering the Mechanisms of Cardiomyocyte Differentiation and Dedifferentiation
Researcher (PI) Eldad Tzahor
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary The quest to restore damaged organs is one of the major challenges in medicine. Recent studies in both animals and in humans suggest that the heart has a limited capacity to replenish its own cardiomyocytes (CMs) throughout life, albeit inadequate to compensate for major injuries such as acute myocardial infarction (MI). Most therapeutic research in regenerative cardiogenesis is geared toward stem cell therapy as a means to replace lost CMs associated with ischemic heart disease. Clinical data evaluating the efficacy of cell-based therapy for heart disease are relatively disappointing. This proposal encompasses multidisciplinary and novel approaches to study the molecular and cellular mechanisms that govern the proliferation, differentiation and dedifferentiation of endogenous CMs, combining developmental-, systems- and cell-biology methodologies in vitro and in vivo, in chick, rodent, and human tissue samples. First, we will perform combinatorial perturbations of signaling pathways in chick embryos, focusing primarily on the FGF-ERK pathway, to investigate the molecular switch between cardiac progenitors and CMs (Aim 1). Because adult CMs have limited proliferative capacity, mainly due to mechanical constraints, in Aim 2, we will apply state-of-the-art techniques in cell biology, to determine whether specific mechno-transduction stimuli can prime the proliferation of differentiated CMs. In order to gain deeper insights into the capacity of adult CMs to renew themselves under normal and pathological conditions, in Aim 3, we will employ a novel cell lineage methodology in mouse and human tissue, based on information encoded in genome. Using this methodology, we hope to shed light on the maintenance, renewal and regenerative capacities of adult CMs in vivo. The expected outcome will be a significantly greater understanding of the bidirectional transition from proliferating cardiac progenitors into differentiated CMs, in embryonic and adult hearts.
Summary
The quest to restore damaged organs is one of the major challenges in medicine. Recent studies in both animals and in humans suggest that the heart has a limited capacity to replenish its own cardiomyocytes (CMs) throughout life, albeit inadequate to compensate for major injuries such as acute myocardial infarction (MI). Most therapeutic research in regenerative cardiogenesis is geared toward stem cell therapy as a means to replace lost CMs associated with ischemic heart disease. Clinical data evaluating the efficacy of cell-based therapy for heart disease are relatively disappointing. This proposal encompasses multidisciplinary and novel approaches to study the molecular and cellular mechanisms that govern the proliferation, differentiation and dedifferentiation of endogenous CMs, combining developmental-, systems- and cell-biology methodologies in vitro and in vivo, in chick, rodent, and human tissue samples. First, we will perform combinatorial perturbations of signaling pathways in chick embryos, focusing primarily on the FGF-ERK pathway, to investigate the molecular switch between cardiac progenitors and CMs (Aim 1). Because adult CMs have limited proliferative capacity, mainly due to mechanical constraints, in Aim 2, we will apply state-of-the-art techniques in cell biology, to determine whether specific mechno-transduction stimuli can prime the proliferation of differentiated CMs. In order to gain deeper insights into the capacity of adult CMs to renew themselves under normal and pathological conditions, in Aim 3, we will employ a novel cell lineage methodology in mouse and human tissue, based on information encoded in genome. Using this methodology, we hope to shed light on the maintenance, renewal and regenerative capacities of adult CMs in vivo. The expected outcome will be a significantly greater understanding of the bidirectional transition from proliferating cardiac progenitors into differentiated CMs, in embryonic and adult hearts.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym CORALWARM
Project Corals and global warming: The Mediterranean versus the Red Sea
Researcher (PI) Zvy Dubinsky
Host Institution (HI) BAR ILAN UNIVERSITY
Call Details Advanced Grant (AdG), LS8, ERC-2009-AdG
Summary CoralWarm will generate for the first time projections of temperate and subtropical coral survival by integrating sublethal temperature increase effects on metabolic and skeletal processes in Mediterranean and Red Sea key species. CoralWarm unique approach is from the nano- to the macro-scale, correlating molecular events to environmental processes. This will show new pathways to future investigations on cellular mechanisms linking environmental factors to final phenotype, potentially improving prediction powers and paleoclimatological interpretation. Biological and chemical expertise will merge, producing new interdisciplinary approaches for ecophysiology and biomineralization. Field transplantations will be combined with controlled experiments under IPCC scenarios. Corals will be grown in aquaria, exposing the Mediterranean species native to cooler waters to higher temperatures, and the Red Sea ones to gradually increasing above ambient warming seawater. Virtually all state-of-the-art methods will be used, by uniquely combining the investigators expertise. Expected results include responses of algal symbionts photosynthesis, host, symbiont and holobiont respiration, biomineralization rates and patterns, including colony architecture, and reproduction to temperature and pH gradients and combinations. Integration of molecular aspects of potential replacement of symbiont clades, changes in skeletal crystallography, with biochemical and physiological aspects of temperature response, will lead to a novel mechanistic model predicting changes in coral ecology and survival prospect. High-temperature tolerant clades and species will be revealed, allowing future bioremediation actions and establishment of coral refuges, saving corals and coral reefs for future generations.
Summary
CoralWarm will generate for the first time projections of temperate and subtropical coral survival by integrating sublethal temperature increase effects on metabolic and skeletal processes in Mediterranean and Red Sea key species. CoralWarm unique approach is from the nano- to the macro-scale, correlating molecular events to environmental processes. This will show new pathways to future investigations on cellular mechanisms linking environmental factors to final phenotype, potentially improving prediction powers and paleoclimatological interpretation. Biological and chemical expertise will merge, producing new interdisciplinary approaches for ecophysiology and biomineralization. Field transplantations will be combined with controlled experiments under IPCC scenarios. Corals will be grown in aquaria, exposing the Mediterranean species native to cooler waters to higher temperatures, and the Red Sea ones to gradually increasing above ambient warming seawater. Virtually all state-of-the-art methods will be used, by uniquely combining the investigators expertise. Expected results include responses of algal symbionts photosynthesis, host, symbiont and holobiont respiration, biomineralization rates and patterns, including colony architecture, and reproduction to temperature and pH gradients and combinations. Integration of molecular aspects of potential replacement of symbiont clades, changes in skeletal crystallography, with biochemical and physiological aspects of temperature response, will lead to a novel mechanistic model predicting changes in coral ecology and survival prospect. High-temperature tolerant clades and species will be revealed, allowing future bioremediation actions and establishment of coral refuges, saving corals and coral reefs for future generations.
Max ERC Funding
3 332 032 €
Duration
Start date: 2010-06-01, End date: 2016-05-31
Project acronym CRYOPRESERVATION
Project Improved Cryopreservation using Ice Binding Proteins
Researcher (PI) Ido Braslavsky
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS9, ERC-2011-StG_20101109
Summary Several organisms have evolved specialized ice binding proteins (IBPs) that prevent their body fluids from freezing (antifreeze proteins, AFPs), inhibit recrystallization of ice in frozen tissues, or initiate freezing at moderate supercooling temperatures (ice nucleating proteins, INPs). These proteins have many potential applications in agriculture, food preservation, cryobiology, and biomedical science. The ubiquitous presence of IBPs in such organisms indicates the power of these molecules to enable survival under cold conditions. Despite this key role in nature, however, IBPs have been effectively exploited in only one cryopreservation application, namely, recrystallization inhibition in ice cream. Several terrestrial organisms, including insects, have developed very active forms of AFPs. These hyperactive AFPs (hypAFPs) have not been utilized significantly thus far in cryopreservation techniques. The gap between the obvious potential of IBPs and their actual applications stems from a lack of knowledge regarding the mechanisms by which IBPs interact with ice surfaces and how these proteins can assist in cryoprotection. I propose to investigate the mechanism by which IBPs inhibit ice crystallization and the use of such proteins for cryopreserving cells, tissues, and organisms. My group has a strong record in the study of the interactions between IBPs and ice using novel methods that we have developed, including fluorescence microscopy techniques combined with cooled microfluidic devices. We will investigate the interactions of AFPs with ice and the use of hypAFPs in cryopreservation procedures. This research will contribute to an understanding of the mechanisms by which IBPs act, and apply the acquired knowledge to cryopreservation. The successful implementation of IBPs in cryopreservation would revolutionize the field of cryobiology, with enormous implications for cryopreservation applications in general and the frozen and chilled food industry in particular.
Summary
Several organisms have evolved specialized ice binding proteins (IBPs) that prevent their body fluids from freezing (antifreeze proteins, AFPs), inhibit recrystallization of ice in frozen tissues, or initiate freezing at moderate supercooling temperatures (ice nucleating proteins, INPs). These proteins have many potential applications in agriculture, food preservation, cryobiology, and biomedical science. The ubiquitous presence of IBPs in such organisms indicates the power of these molecules to enable survival under cold conditions. Despite this key role in nature, however, IBPs have been effectively exploited in only one cryopreservation application, namely, recrystallization inhibition in ice cream. Several terrestrial organisms, including insects, have developed very active forms of AFPs. These hyperactive AFPs (hypAFPs) have not been utilized significantly thus far in cryopreservation techniques. The gap between the obvious potential of IBPs and their actual applications stems from a lack of knowledge regarding the mechanisms by which IBPs interact with ice surfaces and how these proteins can assist in cryoprotection. I propose to investigate the mechanism by which IBPs inhibit ice crystallization and the use of such proteins for cryopreserving cells, tissues, and organisms. My group has a strong record in the study of the interactions between IBPs and ice using novel methods that we have developed, including fluorescence microscopy techniques combined with cooled microfluidic devices. We will investigate the interactions of AFPs with ice and the use of hypAFPs in cryopreservation procedures. This research will contribute to an understanding of the mechanisms by which IBPs act, and apply the acquired knowledge to cryopreservation. The successful implementation of IBPs in cryopreservation would revolutionize the field of cryobiology, with enormous implications for cryopreservation applications in general and the frozen and chilled food industry in particular.
Max ERC Funding
1 500 000 €
Duration
Start date: 2011-11-01, End date: 2016-10-31
Project acronym DEPICT
Project Design principles and controllability of protein circuits
Researcher (PI) Uri Alon
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS2, ERC-2009-AdG
Summary Cells use circuits of interacting proteins to respond to their environment. In the past decades, molecular biology has provided detailed knowledge on the proteins in these circuits and their interactions. To fully understand circuit function requires, in addition to molecular knowledge, new concepts that explain how multiple components work together to perform systems level functions. Our lab has been a leader in defining such concepts, based on combined experimental and theoretical study of well characterized circuits in bacteria and human cells. In this proposal we aim to find novel principles on how circuits resist fluctuations and errors, and how they can be controlled by drugs: (1) Why do key regulatory systems use bifunctional enzymes that catalyze antagonistic reactions (e.g. both kinase and phosphatase)? We will test the role of bifunctional enzymes in making circuits robust to variations in protein levels. (2) Why are some genes regulated by a repressor and others by an activator? We will test this in the context of reduction of errors in transcription control. (3) Are there principles that describe how drugs combine to affect protein dynamics in human cells? We will use a novel dynamic proteomics approach developed in our lab to explore how protein dynamics can be controlled by drug combinations. This research will define principles that unite our understanding of seemingly distinct biological systems, and explain their particular design in terms of systems-level functions. This understanding will help form the basis for a future medicine that rationally controls the state of the cell based on a detailed blueprint of their circuit design, and quantitative principles for the effects of drugs on this circuitry.
Summary
Cells use circuits of interacting proteins to respond to their environment. In the past decades, molecular biology has provided detailed knowledge on the proteins in these circuits and their interactions. To fully understand circuit function requires, in addition to molecular knowledge, new concepts that explain how multiple components work together to perform systems level functions. Our lab has been a leader in defining such concepts, based on combined experimental and theoretical study of well characterized circuits in bacteria and human cells. In this proposal we aim to find novel principles on how circuits resist fluctuations and errors, and how they can be controlled by drugs: (1) Why do key regulatory systems use bifunctional enzymes that catalyze antagonistic reactions (e.g. both kinase and phosphatase)? We will test the role of bifunctional enzymes in making circuits robust to variations in protein levels. (2) Why are some genes regulated by a repressor and others by an activator? We will test this in the context of reduction of errors in transcription control. (3) Are there principles that describe how drugs combine to affect protein dynamics in human cells? We will use a novel dynamic proteomics approach developed in our lab to explore how protein dynamics can be controlled by drug combinations. This research will define principles that unite our understanding of seemingly distinct biological systems, and explain their particular design in terms of systems-level functions. This understanding will help form the basis for a future medicine that rationally controls the state of the cell based on a detailed blueprint of their circuit design, and quantitative principles for the effects of drugs on this circuitry.
Max ERC Funding
2 261 440 €
Duration
Start date: 2010-03-01, End date: 2015-02-28
Project acronym DIADRUG
Project Insulin resistance and diabetic nephropathy - development of novel in vivo models for drug discovery
Researcher (PI) Sanna Lehtonen
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Starting Grant (StG), LS9, ERC-2009-StG
Summary Up to one third of diabetic patients develop nephropathy, a serious complication of diabetes. Microalbuminuria is the earliest sign of the complication, which may ultimately develop to end-stage renal disease requiring dialysis or a kidney transplant. Insulin resistance and metabolic syndrome are associated with an increased risk for diabetic nephropathy. Interestingly, glomerular epithelial cells or podocytes have recently been shown to be insulin responsive. Further, nephrin, a key structural component of podocytes, is essential for insulin action in these cells. Our novel findings show that adaptor protein CD2AP, an interaction partner of nephrin, associates with regulators of insulin signaling and glucose transport in glomeruli. The results suggest that nephrin and CD2AP are involved, by association with these proteins, in the regulation of insulin signaling and glucose transport in podocytes. We hypothesize that podocytes can develop insulin resistance and that disturbances in insulin response affect podocyte function and contribute to the development of diabetic nephropathy. The aim of this project is to clarify the mechanisms leading to development of insulin resistance in podocytes and to study the association between insulin resistance and the development of diabetic nephropathy. For this we will develop transgenic zebrafish and mouse models by overexpressing/knocking down insulin signaling-associated proteins specifically in podocytes. Further, we aim to identify novel drug leads to treat insulin resistance and diabetic nephropathy by performing high-throughput small molecule library screens on the developed transgenic fish models. The ultimate goal is to find a treatment to combat the early stages of diabetic nephropathy in humans.
Summary
Up to one third of diabetic patients develop nephropathy, a serious complication of diabetes. Microalbuminuria is the earliest sign of the complication, which may ultimately develop to end-stage renal disease requiring dialysis or a kidney transplant. Insulin resistance and metabolic syndrome are associated with an increased risk for diabetic nephropathy. Interestingly, glomerular epithelial cells or podocytes have recently been shown to be insulin responsive. Further, nephrin, a key structural component of podocytes, is essential for insulin action in these cells. Our novel findings show that adaptor protein CD2AP, an interaction partner of nephrin, associates with regulators of insulin signaling and glucose transport in glomeruli. The results suggest that nephrin and CD2AP are involved, by association with these proteins, in the regulation of insulin signaling and glucose transport in podocytes. We hypothesize that podocytes can develop insulin resistance and that disturbances in insulin response affect podocyte function and contribute to the development of diabetic nephropathy. The aim of this project is to clarify the mechanisms leading to development of insulin resistance in podocytes and to study the association between insulin resistance and the development of diabetic nephropathy. For this we will develop transgenic zebrafish and mouse models by overexpressing/knocking down insulin signaling-associated proteins specifically in podocytes. Further, we aim to identify novel drug leads to treat insulin resistance and diabetic nephropathy by performing high-throughput small molecule library screens on the developed transgenic fish models. The ultimate goal is to find a treatment to combat the early stages of diabetic nephropathy in humans.
Max ERC Funding
2 000 000 €
Duration
Start date: 2009-11-01, End date: 2014-10-31
Project acronym ENGVASC
Project Engineering Vascularized Tissues
Researcher (PI) Shulamit Levenberg
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Vascularization, the process in which new blood vessels assemble, is fundamental to tissue vitality. Vessel network assembly within 3D tissues can be induced in-vitro by means of multicellular culturing of endothelial cells (EC), fibroblasts and cells specific to the tissue of interest. This approach supports formation of endothelial vessels and promotes EC and tissue-specific cell interactions. Such EC-dependent tube-like openings may also form the basis for improved media penetration to the inner regions of thick 3D constructs, allowing for enhanced construct survival and for effective engineering of large complex tissues in the lab. Moreover, our own breakthrough results describe the beneficial impact of in vitro prevascularization of engineered muscle tissue on its survival and vascularization upon implantation. These studies have also demonstrated that implanted vascular networks of in vitro engineered constructs, can anastomose with host vasculature and form functional blood vessels in vivo. However, the mechanisms underlying enhanced vascularization of endothelialized engineered constructs and implant-host vessel integration remain unclear. In this proposal, our research objectives are (1) to uncover the mechanisms governing in vitro vessel network formation in engineered 3D tissues and (2) to elucidate the process of graft-host vessel network integration and implant vessel-stimulated promotion of neovascularization in vivo. In addition, the impact of construct prevascularization on implant survival and function will be explored in animal disease models. While there are still many challenges ahead, should we succeed, our research could lay the foundation for significantly enhanced tissue construct vascularization procedures and for their application in regenerative medicine. In addition, it may provide alternative models for studying the vascularization processes in embryogenesis and disease.
Summary
Vascularization, the process in which new blood vessels assemble, is fundamental to tissue vitality. Vessel network assembly within 3D tissues can be induced in-vitro by means of multicellular culturing of endothelial cells (EC), fibroblasts and cells specific to the tissue of interest. This approach supports formation of endothelial vessels and promotes EC and tissue-specific cell interactions. Such EC-dependent tube-like openings may also form the basis for improved media penetration to the inner regions of thick 3D constructs, allowing for enhanced construct survival and for effective engineering of large complex tissues in the lab. Moreover, our own breakthrough results describe the beneficial impact of in vitro prevascularization of engineered muscle tissue on its survival and vascularization upon implantation. These studies have also demonstrated that implanted vascular networks of in vitro engineered constructs, can anastomose with host vasculature and form functional blood vessels in vivo. However, the mechanisms underlying enhanced vascularization of endothelialized engineered constructs and implant-host vessel integration remain unclear. In this proposal, our research objectives are (1) to uncover the mechanisms governing in vitro vessel network formation in engineered 3D tissues and (2) to elucidate the process of graft-host vessel network integration and implant vessel-stimulated promotion of neovascularization in vivo. In addition, the impact of construct prevascularization on implant survival and function will be explored in animal disease models. While there are still many challenges ahead, should we succeed, our research could lay the foundation for significantly enhanced tissue construct vascularization procedures and for their application in regenerative medicine. In addition, it may provide alternative models for studying the vascularization processes in embryogenesis and disease.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym EvoDevoQuorum
Project Evolution and Development of Bacterial Communication
Researcher (PI) Avigdor Eldar
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS2, ERC-2011-StG_20101109
Summary Bacterial cooperation underlies many bacterial traits of practical interest. Many social traits of bacteria are regulated by inter-cellular signalling pathways, generally known as quorum sensing (QS). QS has been proposed as novel target for anti-virulence treatment. To this aim, there is a need to better understand the mechanisms of QS and their social and evolutionary impact.
While the basic schemes of a single quorum sensing pathway acting in homogenous conditions are well understood, the system’s level function of QS regulatory networks can only be appreciated by considering the role phenotypic and genetic variability has on shaping the network’s structure and function. Phenotypic variability in complex communities may arise from division of labour between cells and environmental gradients and substantially impact the way cells secrete and interpret QS signals. Genetic variability in QS networks may lead to multiple social relations between cells of different genotypes including cross-talks, interception, manipulation and quenching of signals. This will affect the population structure and performance.
The proposed project will study the function of QS signalling in heterogeneous communities. Phenotypic variability and its impact on QS function will be studied in a spatially inhomogeneous cooperating system. Genetic variability will be studied at the macro and micro-scales in a bacterial species showing rapid diversification of their QS networks. Finally, we will rationally design strains with superior ‘cheating’ strategies that can invade and eliminate a cooperative population.
Throughout this project, we will use a combination of experimental techniques from microbiology, socio-biology, genetics and microscopy together with mathematical analysis tools from systems biology, population genetics and game theory, to study bacterial cooperation and its dependence on the underlying communication network, social complexity and environmental variation.
Summary
Bacterial cooperation underlies many bacterial traits of practical interest. Many social traits of bacteria are regulated by inter-cellular signalling pathways, generally known as quorum sensing (QS). QS has been proposed as novel target for anti-virulence treatment. To this aim, there is a need to better understand the mechanisms of QS and their social and evolutionary impact.
While the basic schemes of a single quorum sensing pathway acting in homogenous conditions are well understood, the system’s level function of QS regulatory networks can only be appreciated by considering the role phenotypic and genetic variability has on shaping the network’s structure and function. Phenotypic variability in complex communities may arise from division of labour between cells and environmental gradients and substantially impact the way cells secrete and interpret QS signals. Genetic variability in QS networks may lead to multiple social relations between cells of different genotypes including cross-talks, interception, manipulation and quenching of signals. This will affect the population structure and performance.
The proposed project will study the function of QS signalling in heterogeneous communities. Phenotypic variability and its impact on QS function will be studied in a spatially inhomogeneous cooperating system. Genetic variability will be studied at the macro and micro-scales in a bacterial species showing rapid diversification of their QS networks. Finally, we will rationally design strains with superior ‘cheating’ strategies that can invade and eliminate a cooperative population.
Throughout this project, we will use a combination of experimental techniques from microbiology, socio-biology, genetics and microscopy together with mathematical analysis tools from systems biology, population genetics and game theory, to study bacterial cooperation and its dependence on the underlying communication network, social complexity and environmental variation.
Max ERC Funding
1 497 996 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym EXPRES
Project Chromatin and transcription in ES cells: from single cells to genome wide views
Researcher (PI) Eran Meshorer
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS2, ERC-2011-StG_20101109
Summary How embryonic stem cells (ESCs) maintain their dual capacity to self-renew and to differentiate into all cell types is one of the fundamental questions in biology. Although this question remains largely open, there is growing evidence suggesting that chromatin plasticity is a fundamental hallmark of ESCs, providing their necessary flexibility.
Previously we found that ESCs possess a relatively open chromatin conformation, giving rise to permissive transcriptional program. Here I propose to investigate the mechanisms that support chromatin plasticity and pluripotency in ESCs.
Using a simple biochemical assay which I developed (DCAP: Differential Chromatin Associated Proteins), based on micrococcal nuclease (MNase) digestion combined with multi-dimensional protein identification technology (MudPIT), I seek to identify ESC-specific chromatin proteins. Selected proteins will be knocked-down (or out) and their ESC function will be evaluated.
In addition, I will conduct a hypothesis-driven research using mutant ESCs and epigenetic-related drugs to search for potential mechanisms, (i.e. histone modifications, DNA methylation), that may support chromatin plasticity in ESCs. Based on our intriguing preliminary data, I will also focus on the link between the nuclear lamina and ESC plasticity.
Thirdly, we will analyze non-polyadenylated transcription using genome-wide tiling arrays and RNA-seq. We will design custom microarrays containing the identified sequences, which will allow us to reveal, using ChIP-chip experiments, the mechanistic regulation of the non-polyadenylated transcripts. Finally, we will knockout, using zinc-finger nuclease technology, selected highly conserved candidates in search of their function.
Understanding chromatin regulation, plasticity and function will enable one to intelligently manipulate ESCs to transition between the pluripotent, multipotent and unipotent states and to expedite their use in the clinic.
Summary
How embryonic stem cells (ESCs) maintain their dual capacity to self-renew and to differentiate into all cell types is one of the fundamental questions in biology. Although this question remains largely open, there is growing evidence suggesting that chromatin plasticity is a fundamental hallmark of ESCs, providing their necessary flexibility.
Previously we found that ESCs possess a relatively open chromatin conformation, giving rise to permissive transcriptional program. Here I propose to investigate the mechanisms that support chromatin plasticity and pluripotency in ESCs.
Using a simple biochemical assay which I developed (DCAP: Differential Chromatin Associated Proteins), based on micrococcal nuclease (MNase) digestion combined with multi-dimensional protein identification technology (MudPIT), I seek to identify ESC-specific chromatin proteins. Selected proteins will be knocked-down (or out) and their ESC function will be evaluated.
In addition, I will conduct a hypothesis-driven research using mutant ESCs and epigenetic-related drugs to search for potential mechanisms, (i.e. histone modifications, DNA methylation), that may support chromatin plasticity in ESCs. Based on our intriguing preliminary data, I will also focus on the link between the nuclear lamina and ESC plasticity.
Thirdly, we will analyze non-polyadenylated transcription using genome-wide tiling arrays and RNA-seq. We will design custom microarrays containing the identified sequences, which will allow us to reveal, using ChIP-chip experiments, the mechanistic regulation of the non-polyadenylated transcripts. Finally, we will knockout, using zinc-finger nuclease technology, selected highly conserved candidates in search of their function.
Understanding chromatin regulation, plasticity and function will enable one to intelligently manipulate ESCs to transition between the pluripotent, multipotent and unipotent states and to expedite their use in the clinic.
Max ERC Funding
1 500 000 €
Duration
Start date: 2011-12-01, End date: 2016-11-30
Project acronym FUTUREGENES
Project Gene transfer techniques in the treatment of cardiovascular diseases and malignant glioma
Researcher (PI) Seppo Yla-Herttuala
Host Institution (HI) ITA-SUOMEN YLIOPISTO
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary Background: Poor angiogenesis and collateral vessel formation lead to coronary heart disease, claudication, infarctions and amputations while malignant glioma is one of the most aggressive proangiogenic tumors leading to death in a few months. For these diseases either stimulation or blocking, respectively, of angiogenesis may provide novel treatment options. Advancing State-of-the-Art: Our hypothesis is that in ischemia it will be possible to support natural growth of blood vessels with Therapeutic angiogenesis and lymphangiogenesis by using local gene transfer of the new members of vascular endothelial growth factor (VEGF) family and their receptors. New co-receptors, designer mutants and PCR suffling products of VEGFs will be used. New vector technology will be used to achieve long-lasting effects of VEGFs. We aim to develop novel site-specifically integrating, targeted, regulated vectors to precisely express the new VEGFs, their soluble decoy receptors and single-chain therapeutic antibodies (scFv) for pro- and anti-angiogenic purposes. As novel approaches, we have developed metabolically biotinylated lenti- and adenoviruses suitable for targeting and Epigenetherapy where siRNA/miRNAs and short nuclear RNAs regulate endogenous gene expression at the VEGF promoter level via modification of histone code. scFv library for endothelial cells and lentivirus-siRNA library directed to all human and mouse kinases will be screened to identify new mediators of angiogenesis in order to develop next generation pro- and antiangiogenic therapies. Based on our strong track record in Clinical applications, the best new pro- and antiangiogenic approaches will be taken to phase I clinical studies in myocardial ischemia and malignant glioma. Significance: This work should lead to significant advances and new therapies for severe ischemia and malignant glioma. Epigenetherapy and new site-specifically integrating, regulated vectors should be widely applicable in medicine.
Summary
Background: Poor angiogenesis and collateral vessel formation lead to coronary heart disease, claudication, infarctions and amputations while malignant glioma is one of the most aggressive proangiogenic tumors leading to death in a few months. For these diseases either stimulation or blocking, respectively, of angiogenesis may provide novel treatment options. Advancing State-of-the-Art: Our hypothesis is that in ischemia it will be possible to support natural growth of blood vessels with Therapeutic angiogenesis and lymphangiogenesis by using local gene transfer of the new members of vascular endothelial growth factor (VEGF) family and their receptors. New co-receptors, designer mutants and PCR suffling products of VEGFs will be used. New vector technology will be used to achieve long-lasting effects of VEGFs. We aim to develop novel site-specifically integrating, targeted, regulated vectors to precisely express the new VEGFs, their soluble decoy receptors and single-chain therapeutic antibodies (scFv) for pro- and anti-angiogenic purposes. As novel approaches, we have developed metabolically biotinylated lenti- and adenoviruses suitable for targeting and Epigenetherapy where siRNA/miRNAs and short nuclear RNAs regulate endogenous gene expression at the VEGF promoter level via modification of histone code. scFv library for endothelial cells and lentivirus-siRNA library directed to all human and mouse kinases will be screened to identify new mediators of angiogenesis in order to develop next generation pro- and antiangiogenic therapies. Based on our strong track record in Clinical applications, the best new pro- and antiangiogenic approaches will be taken to phase I clinical studies in myocardial ischemia and malignant glioma. Significance: This work should lead to significant advances and new therapies for severe ischemia and malignant glioma. Epigenetherapy and new site-specifically integrating, regulated vectors should be widely applicable in medicine.
Max ERC Funding
2 200 000 €
Duration
Start date: 2010-06-01, End date: 2015-05-31
Project acronym GENANX
Project A cross-species neurogenomics approach to anxiety
Researcher (PI) Iiris Maaria Hovatta
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Starting Grant (StG), LS2, ERC-2011-StG_20101109
Summary My goal is to identify novel genetic factors that regulate normal and pathological anxiety. Anxiety disorders are complex diseases with genetic and environmental susceptibility factors. Stressful life events, especially in childhood are well-known risk factors for anxiety disorders. We have previously used inbred mouse strains to identify genes underlying innate anxiety. Here I propose a new approach using a mouse model of anxiety induced by social stress to identify gene regulatory networks, to show the causality of the selected networks in vivo, and to investigate involvement of these networks in the genetic predisposition to human anxiety disorders. Gene regulatory networks will be identified by using massively parallel sequencing methods (i.e. miRNA-seq and mRNA-seq). We will generate from four brain regions known to regulate anxiety a complete gene and miRNA expression atlas, which is used to identify genes, transcripts, miRNAs and isomiRs differentially expressed between anxious and control animals. Pathway and network analysis tools are employed to identify molecular pathways affected by induced anxiety. Datasets consisting of different levels of data from individual brain regions will then be integrated in light of existing molecular and anatomical information to construct draft network models that will be curated, filtered and enhanced. On the basis of these models, we will generate specific hypotheses that are tested in vivo in mice to show the causality of the most interesting networks. To identify the networks important for human anxiety disorders, we have access to a unique population-based cohort from Finland characterized for anxiety disorders. Candidate genes and miRNAs from selected networks are tested in a genetic association analysis. This multidisciplinary project will increase understanding of the genetic and neurobiological basis of anxiety in mouse and human, and has potential to provide targets for the development of improved anxiolytics.
Summary
My goal is to identify novel genetic factors that regulate normal and pathological anxiety. Anxiety disorders are complex diseases with genetic and environmental susceptibility factors. Stressful life events, especially in childhood are well-known risk factors for anxiety disorders. We have previously used inbred mouse strains to identify genes underlying innate anxiety. Here I propose a new approach using a mouse model of anxiety induced by social stress to identify gene regulatory networks, to show the causality of the selected networks in vivo, and to investigate involvement of these networks in the genetic predisposition to human anxiety disorders. Gene regulatory networks will be identified by using massively parallel sequencing methods (i.e. miRNA-seq and mRNA-seq). We will generate from four brain regions known to regulate anxiety a complete gene and miRNA expression atlas, which is used to identify genes, transcripts, miRNAs and isomiRs differentially expressed between anxious and control animals. Pathway and network analysis tools are employed to identify molecular pathways affected by induced anxiety. Datasets consisting of different levels of data from individual brain regions will then be integrated in light of existing molecular and anatomical information to construct draft network models that will be curated, filtered and enhanced. On the basis of these models, we will generate specific hypotheses that are tested in vivo in mice to show the causality of the most interesting networks. To identify the networks important for human anxiety disorders, we have access to a unique population-based cohort from Finland characterized for anxiety disorders. Candidate genes and miRNAs from selected networks are tested in a genetic association analysis. This multidisciplinary project will increase understanding of the genetic and neurobiological basis of anxiety in mouse and human, and has potential to provide targets for the development of improved anxiolytics.
Max ERC Funding
1 499 863 €
Duration
Start date: 2012-02-01, End date: 2017-07-31
Project acronym GENCON
Project The evolutionary implications of genetic conflict
Researcher (PI) Göran Arnqvist
Host Institution (HI) UPPSALA UNIVERSITET
Call Details Advanced Grant (AdG), LS8, ERC-2011-ADG_20110310
Summary The study of genetic conflict is developing at an almost explosive rate. The recognition that genes or alleles residing in individuals of the two sexes may have conflicting interests is transforming evolutionary biology and, likewise, conflict between genes showing different modes of transmission may fundamentally affect adaptive evolution. The research proposed here will push the frontiers of genetic conflict research and establish new domains. It is aimed at exploring the novel possibility that conflict between mitochondrial and nuclear genes have far-reaching implications for adaptive evolution and at advancing our understanding of the biological consequences of sexual conflict. The project consists of several interrelated parts and will employ insects as model systems. First, I will assess to what extent genetic variation in fitness is sexually antagonistic and what life history traits contribute to sexually antagonistic variation. Second, I will elucidate the genomics of metabolic rate and measure selection on metabolic phenotypes. Third, I will test whether sexually antagonistic epistatic interactions between mitochondrial and nuclear genes generate conflict over metabolic rate. Fourth, I will test the hypothesis that sexual conflict contribute to the evolution of primary and secondary sexual traits. Fifth, I will shed light on the complicated evolutionary interplay between sexual conflict and mating system evolution. I will employ an innovative research strategy, ‘experimental genomics’, in which genomic data is used to guide experimental evolutionary work with distinct genotypes. The research outlined here will collectively provide an unprecedented wealth of information into the role of genetic conflict in several horizons of adaptive evolution, ranging from DNA sequence evolution over life history evolution to speciation, and will set the standard for a new generation of insightful studies aimed at bridging the gap between phenotypic selection and genomics.
Summary
The study of genetic conflict is developing at an almost explosive rate. The recognition that genes or alleles residing in individuals of the two sexes may have conflicting interests is transforming evolutionary biology and, likewise, conflict between genes showing different modes of transmission may fundamentally affect adaptive evolution. The research proposed here will push the frontiers of genetic conflict research and establish new domains. It is aimed at exploring the novel possibility that conflict between mitochondrial and nuclear genes have far-reaching implications for adaptive evolution and at advancing our understanding of the biological consequences of sexual conflict. The project consists of several interrelated parts and will employ insects as model systems. First, I will assess to what extent genetic variation in fitness is sexually antagonistic and what life history traits contribute to sexually antagonistic variation. Second, I will elucidate the genomics of metabolic rate and measure selection on metabolic phenotypes. Third, I will test whether sexually antagonistic epistatic interactions between mitochondrial and nuclear genes generate conflict over metabolic rate. Fourth, I will test the hypothesis that sexual conflict contribute to the evolution of primary and secondary sexual traits. Fifth, I will shed light on the complicated evolutionary interplay between sexual conflict and mating system evolution. I will employ an innovative research strategy, ‘experimental genomics’, in which genomic data is used to guide experimental evolutionary work with distinct genotypes. The research outlined here will collectively provide an unprecedented wealth of information into the role of genetic conflict in several horizons of adaptive evolution, ranging from DNA sequence evolution over life history evolution to speciation, and will set the standard for a new generation of insightful studies aimed at bridging the gap between phenotypic selection and genomics.
Max ERC Funding
2 497 442 €
Duration
Start date: 2012-05-01, End date: 2017-04-30
Project acronym GENOVAR
Project Sequence based strategies to identify genetic variation associated with mental retardation and schizophrenia
Researcher (PI) Lars Feuk
Host Institution (HI) UPPSALA UNIVERSITET
Call Details Starting Grant (StG), LS2, ERC-2011-StG_20101109
Summary Mental retardation (MR) and schizophrenia (SCZ) are disorders of the brain that affect 2-3% and 1% of the population, respectively. Both disorders are considered to be highly heritable, but exhibit heterogeneous genetic etiology. Recent genetic studies have led to discoveries that the same variants that can give rise to different neuropsychiatric disorders, including MR and SCZ. In this proposal, sequencing will be used to identify novel genes involved in MR and SCZ, and to explore the potential overlap between these disorders. The specific goals of the research plan include:
1. Genetic characterization of patients from large pedigrees with SCZ and MR.
Five pedigrees have been collected in which multiple individuals are affected by SCZ or MR. The pedigrees vary in size, with the largest spanning 12 generations including 3,400 individuals. Exome and whole genome sequencing will be performed to identify the genetic variants associated with disease. Candidate genes identified will be screened in large independent cohorts of MR and SCZ patients. In addition, RNA sequencing will be performed on cell lines established for patients and controls from two of the pedigrees.
2. Screening of trios to identify novel genes causing MR
Mental retardation (MR) patients are typically referred for array-based analysis. With current genetic screening using microarray, a clinically significant rearrangement is identified in 15-20% of patients. I propose use high throughput sequencing to screen MR patients and their parents with the goal of identifying new MR genes and to investigate to what extent the diagnostic yield can be increased.
By combining sequencing, bioinformatics and carefully selected clinical material, the work presented in this proposal will lead to an increased understanding of disease mechanisms and enable the development of novel targets and strategies for molecular diagnostic screening.
Summary
Mental retardation (MR) and schizophrenia (SCZ) are disorders of the brain that affect 2-3% and 1% of the population, respectively. Both disorders are considered to be highly heritable, but exhibit heterogeneous genetic etiology. Recent genetic studies have led to discoveries that the same variants that can give rise to different neuropsychiatric disorders, including MR and SCZ. In this proposal, sequencing will be used to identify novel genes involved in MR and SCZ, and to explore the potential overlap between these disorders. The specific goals of the research plan include:
1. Genetic characterization of patients from large pedigrees with SCZ and MR.
Five pedigrees have been collected in which multiple individuals are affected by SCZ or MR. The pedigrees vary in size, with the largest spanning 12 generations including 3,400 individuals. Exome and whole genome sequencing will be performed to identify the genetic variants associated with disease. Candidate genes identified will be screened in large independent cohorts of MR and SCZ patients. In addition, RNA sequencing will be performed on cell lines established for patients and controls from two of the pedigrees.
2. Screening of trios to identify novel genes causing MR
Mental retardation (MR) patients are typically referred for array-based analysis. With current genetic screening using microarray, a clinically significant rearrangement is identified in 15-20% of patients. I propose use high throughput sequencing to screen MR patients and their parents with the goal of identifying new MR genes and to investigate to what extent the diagnostic yield can be increased.
By combining sequencing, bioinformatics and carefully selected clinical material, the work presented in this proposal will lead to an increased understanding of disease mechanisms and enable the development of novel targets and strategies for molecular diagnostic screening.
Max ERC Funding
1 496 574 €
Duration
Start date: 2012-08-01, End date: 2017-07-31
Project acronym GLUCOSE SENSING
Project Transcriptional networks in glucose sensing
Researcher (PI) Ville Ilmari Hietakangas
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary Glucose is key source of nutritional energy and raw material for biosynthetic processes. Maintaining glucose homeostasis requires a regulatory network that functions both in the systemic level through hormonal signaling and locally at the intracellular level. Insulin signalling is the main hormonal mechanism involved in maintaining the levels of circulating glucose through regulation of cellular glucose intake and metabolism. While the signalling pathways mediating the effects of insulin have been thoroughly studied, the transcriptional networks downstream of insulin signalling are not comprehensively understood. In addition to insulin signalling, intracellular glucose sensing mechanisms, including transcription factor complex MondoA/B-Mlx, have recently emerged as important regulators of glucose metabolism. In the proposed project we aim to take a systematic approach to characterize the transcriptional regulators involved in glucose sensing and metabolism in physiological context, using Drosophila as the main model system. We will use several complementary screening strategies, both in vivo and in cell culture, to identify transcription factors regulated by insulin and intracellular glucose. Identified transcription factors will be exposed to a panel of in vivo tests measuring parameters related to glucose and energy metabolism, aiming to identify those transcriptional regulators most essential in maintaining glucose homeostasis. With these factors, we will proceed to in-depth analysis, generating mutant alleles, analysing their metabolic profile and physiologically important target genes as well as functional conservation in mammals. Our aim is to identify and characterize several novel transcriptional regulators involved in glucose metabolism and to achieve a comprehensive overview on how these transcriptional regulators act together to achieve metabolic homeostasis in response to fluctuating dietary glucose intake.
Summary
Glucose is key source of nutritional energy and raw material for biosynthetic processes. Maintaining glucose homeostasis requires a regulatory network that functions both in the systemic level through hormonal signaling and locally at the intracellular level. Insulin signalling is the main hormonal mechanism involved in maintaining the levels of circulating glucose through regulation of cellular glucose intake and metabolism. While the signalling pathways mediating the effects of insulin have been thoroughly studied, the transcriptional networks downstream of insulin signalling are not comprehensively understood. In addition to insulin signalling, intracellular glucose sensing mechanisms, including transcription factor complex MondoA/B-Mlx, have recently emerged as important regulators of glucose metabolism. In the proposed project we aim to take a systematic approach to characterize the transcriptional regulators involved in glucose sensing and metabolism in physiological context, using Drosophila as the main model system. We will use several complementary screening strategies, both in vivo and in cell culture, to identify transcription factors regulated by insulin and intracellular glucose. Identified transcription factors will be exposed to a panel of in vivo tests measuring parameters related to glucose and energy metabolism, aiming to identify those transcriptional regulators most essential in maintaining glucose homeostasis. With these factors, we will proceed to in-depth analysis, generating mutant alleles, analysing their metabolic profile and physiologically important target genes as well as functional conservation in mammals. Our aim is to identify and characterize several novel transcriptional regulators involved in glucose metabolism and to achieve a comprehensive overview on how these transcriptional regulators act together to achieve metabolic homeostasis in response to fluctuating dietary glucose intake.
Max ERC Funding
1 496 930 €
Duration
Start date: 2012-01-01, End date: 2017-02-28
Project acronym HOPSEP
Project Harnessing Oxygenic Photosynthesis for Sustainable Energy Production
Researcher (PI) Nathan Nelson
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Advanced Grant (AdG), LS9, ERC-2011-ADG_20110310
Summary Oxygenic photosynthesis, that takes place in cyanobacteria algae and plants, provides most of the food and fuel on earth. The light stage of this process is driven by two photosystems. Photosystem II (PSII) that oxidizes water to O2 and 4 H+ and photosystem I (PSI) which in the light provides the most negative redox potential in nature that can drive numerous reactions including CO2 assimilation and hydrogen (H2) production. The structure of most of the complexes involved in oxygenic photosynthesis was solved in several laboratories including our own. Utilizing our plant PSI crystals we were able to generate a light dependent electric potential of up to 100 V. We will develop this system for designing biological based photoelectric devices. Recently, we discovered a marine phage that carries an operon encoding all PSI subunits. Generation, in synechocystis, of a phage-like PSI enabled the mutated complex to accept electrons from additional sources like respiratory cytochromes. This way a novel photorespiration, where PSI can substitute for cytochrome oxidase, is created. The wild type and mutant synechocystis PSI were crystallized and solved, confirming the suggested structural consequences. Moreover, several structural alterations in the mesophilic PSI were recorded. We designed a hydrogen producing bioreactor where the novel photorespiration will enable to utilize the organic material of the cell as an electron source for H2 production. We propose that in conjunction of engineering a Cyanobacterium strain with a temperature sensitive PSII, enhancing rates in its respiratory chain an efficient and sustainable hydrogen production can be achieved.
Summary
Oxygenic photosynthesis, that takes place in cyanobacteria algae and plants, provides most of the food and fuel on earth. The light stage of this process is driven by two photosystems. Photosystem II (PSII) that oxidizes water to O2 and 4 H+ and photosystem I (PSI) which in the light provides the most negative redox potential in nature that can drive numerous reactions including CO2 assimilation and hydrogen (H2) production. The structure of most of the complexes involved in oxygenic photosynthesis was solved in several laboratories including our own. Utilizing our plant PSI crystals we were able to generate a light dependent electric potential of up to 100 V. We will develop this system for designing biological based photoelectric devices. Recently, we discovered a marine phage that carries an operon encoding all PSI subunits. Generation, in synechocystis, of a phage-like PSI enabled the mutated complex to accept electrons from additional sources like respiratory cytochromes. This way a novel photorespiration, where PSI can substitute for cytochrome oxidase, is created. The wild type and mutant synechocystis PSI were crystallized and solved, confirming the suggested structural consequences. Moreover, several structural alterations in the mesophilic PSI were recorded. We designed a hydrogen producing bioreactor where the novel photorespiration will enable to utilize the organic material of the cell as an electron source for H2 production. We propose that in conjunction of engineering a Cyanobacterium strain with a temperature sensitive PSII, enhancing rates in its respiratory chain an efficient and sustainable hydrogen production can be achieved.
Max ERC Funding
2 487 000 €
Duration
Start date: 2012-01-01, End date: 2017-12-31
Project acronym ID-CAB
Project Individual differences in Collective Animal Behaviour
Researcher (PI) David Sumpter
Host Institution (HI) UPPSALA UNIVERSITET
Call Details Starting Grant (StG), LS8, ERC-2009-StG
Summary One of the key challenges in scientific research is to link together our understanding of different levels of biological organisation. This challenge is fundamental to the scientific endeavour: from understand how genes interact to drive the cell, to how cells interact to form organisms, up to how organisms interact to form groups and societies. My own and the research of others has addressed this question in the context of the collective behaviour of animals. Mathematical models of complex systems have been used to successfully predict experimental outcome. Most previous studies are however limited in one important aspect: individuals are treated as identical units. The aim of the proposed research proposed is to investigate features which produce differences within the units. The model systems of our study will be sticklebacks, homing pigeons and house sparrows. Individuals can differ from each other on a range of time scales, from information acquired within the last few minutes, through socially learnt information, to genetically inherited differences. Through a series of experiments on each of the study species, the development of mathematical models which incorporate between individual differences, and novel forms of data analysis, we will begin to understand the role played by individual differences within groups. We will look at the rules of motion for fish and birds; the role of personality in decision-making and how short term information differences improve decision-making accuracy. Achieving the project objectives will greatly enhance our understanding of the relationship between individual animals and the groups they live in, as well as impacting on our understanding of individual differences in other areas of biology.
Summary
One of the key challenges in scientific research is to link together our understanding of different levels of biological organisation. This challenge is fundamental to the scientific endeavour: from understand how genes interact to drive the cell, to how cells interact to form organisms, up to how organisms interact to form groups and societies. My own and the research of others has addressed this question in the context of the collective behaviour of animals. Mathematical models of complex systems have been used to successfully predict experimental outcome. Most previous studies are however limited in one important aspect: individuals are treated as identical units. The aim of the proposed research proposed is to investigate features which produce differences within the units. The model systems of our study will be sticklebacks, homing pigeons and house sparrows. Individuals can differ from each other on a range of time scales, from information acquired within the last few minutes, through socially learnt information, to genetically inherited differences. Through a series of experiments on each of the study species, the development of mathematical models which incorporate between individual differences, and novel forms of data analysis, we will begin to understand the role played by individual differences within groups. We will look at the rules of motion for fish and birds; the role of personality in decision-making and how short term information differences improve decision-making accuracy. Achieving the project objectives will greatly enhance our understanding of the relationship between individual animals and the groups they live in, as well as impacting on our understanding of individual differences in other areas of biology.
Max ERC Funding
977 768 €
Duration
Start date: 2010-02-01, End date: 2015-01-31
Project acronym IMMUNOSWITCH
Project Switch recombination: a model system for DNA editing and repair in human lymphocytes with relevance for primary immunodeficiency and cancer formation
Researcher (PI) Qiang Pan Hammarström
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Starting Grant (StG), LS6, ERC-2009-StG
Summary The aim of this project is to try to understand the complex molecular mechanisms involved in DNA editing, repair and recombination during immunoglobulin class switch recombination (CSR) and somatic hypermutation (SHM). We have developed a series of PCR-based assays to study in vivo generated CSR junctions and the pattern of mutations introduced in the immunoglobulin variable region genes in human B cells, allowing us to characterize CSR and SHM in patients with immunodeficiency due to defect(s) in DNA repair/recombination. Novel in vitro CSR assays, based on GFP expression, allowing quantitative measurement of substrate recombination, are also being developed. In addition, we have initiated an evolutionary analysis of the function and structure of activation-induced deaminase, an essential molecule involved both in CSR and SHM, aiming to identify CSR specific-cofactor(s). Combining these approaches, we will be able to define the DNA repair pathways involved in CSR and SHM. The suggested project requires access to patients with various defects in the DNA repair pathways. Many of these diseases are exceedingly rare. However, through worldwide collaboration, we have obtained samples from a majority of the diagnosed patients. We are also refining the existing screening methods and developing novel methods, that will allow identification of additional patients both with recognized and new diseases caused by mutations in DNA repair pathways. Finally, we hope to be able to address the question whether illegitimate CSR events are associated with predisposition to lymphomagenesis in patients with immunodeficiency/DNA repair defect(s), by analyzing the CSR induced chromosomal breaks and translocations in these patients. A large-scale sequencing project is also planned to characterize the CSRnome in B-cell lymphoma samples.
Summary
The aim of this project is to try to understand the complex molecular mechanisms involved in DNA editing, repair and recombination during immunoglobulin class switch recombination (CSR) and somatic hypermutation (SHM). We have developed a series of PCR-based assays to study in vivo generated CSR junctions and the pattern of mutations introduced in the immunoglobulin variable region genes in human B cells, allowing us to characterize CSR and SHM in patients with immunodeficiency due to defect(s) in DNA repair/recombination. Novel in vitro CSR assays, based on GFP expression, allowing quantitative measurement of substrate recombination, are also being developed. In addition, we have initiated an evolutionary analysis of the function and structure of activation-induced deaminase, an essential molecule involved both in CSR and SHM, aiming to identify CSR specific-cofactor(s). Combining these approaches, we will be able to define the DNA repair pathways involved in CSR and SHM. The suggested project requires access to patients with various defects in the DNA repair pathways. Many of these diseases are exceedingly rare. However, through worldwide collaboration, we have obtained samples from a majority of the diagnosed patients. We are also refining the existing screening methods and developing novel methods, that will allow identification of additional patients both with recognized and new diseases caused by mutations in DNA repair pathways. Finally, we hope to be able to address the question whether illegitimate CSR events are associated with predisposition to lymphomagenesis in patients with immunodeficiency/DNA repair defect(s), by analyzing the CSR induced chromosomal breaks and translocations in these patients. A large-scale sequencing project is also planned to characterize the CSRnome in B-cell lymphoma samples.
Max ERC Funding
1 888 166 €
Duration
Start date: 2009-12-01, End date: 2014-11-30
Project acronym INFOTROPHIC
Project Algal Bloom Dynamics: From Cellular Mechanisms to Trophic Level Interactions
Researcher (PI) Assaf Vardi
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS8, ERC-2011-StG_20101109
Summary Marine phytoplankton are the basis of marine food webs and are responsible for nearly 50% of the global annual carbon-based primary production. Since phytoplankton exert such a global-scale influence on climate, we are interested in understanding what controls their cell fate during bloom “boom and bust” dynamics. Despite their importance, the molecular basis for their ecological success is still in its infancy. In recent years, the wealth of genomic information from marine microbes, coupled with molecular resources and analytical tools, provide an unprecedented opportunity to address fundamental questions about their unique evolutionary history and ecological role. Nevertheless, there is a critical need to “decode” the genomic resources and translate them into cellular mechanisms, community structure and, eventually, to their role in ecosystem function. This proposed research aims to provide novel insights into the role of a chemical-based “arms race” that mediates and structures the microbial interactions in the marine environment. We will dissect unexplored signaling pathways employed by phytoplankton during sensing and acclimation to changes in their environment. Our overarching objective is to unravel the role of infochemicals and related gene products in regulating phytoplankton surveillance systems in response to environmental stress conditions. We will focus on the three major biotic interactions of key dominant algal groups in the oceans; intercellular signaling in diatoms, host-virus interactions in coccolithophores and predator-prey interactions. We will provide a suite of cellular probes and metabolic biomarkers that will allow in situ detection of chemical signaling and biotic interactions in the oceans and will highlight their role in shaping microbial food webs. Our vision is to provide novel cellular concepts and molecular tools to the link between the intricate mechanisms of cell signaling and stress response, and large biogeochemical cycles.
Summary
Marine phytoplankton are the basis of marine food webs and are responsible for nearly 50% of the global annual carbon-based primary production. Since phytoplankton exert such a global-scale influence on climate, we are interested in understanding what controls their cell fate during bloom “boom and bust” dynamics. Despite their importance, the molecular basis for their ecological success is still in its infancy. In recent years, the wealth of genomic information from marine microbes, coupled with molecular resources and analytical tools, provide an unprecedented opportunity to address fundamental questions about their unique evolutionary history and ecological role. Nevertheless, there is a critical need to “decode” the genomic resources and translate them into cellular mechanisms, community structure and, eventually, to their role in ecosystem function. This proposed research aims to provide novel insights into the role of a chemical-based “arms race” that mediates and structures the microbial interactions in the marine environment. We will dissect unexplored signaling pathways employed by phytoplankton during sensing and acclimation to changes in their environment. Our overarching objective is to unravel the role of infochemicals and related gene products in regulating phytoplankton surveillance systems in response to environmental stress conditions. We will focus on the three major biotic interactions of key dominant algal groups in the oceans; intercellular signaling in diatoms, host-virus interactions in coccolithophores and predator-prey interactions. We will provide a suite of cellular probes and metabolic biomarkers that will allow in situ detection of chemical signaling and biotic interactions in the oceans and will highlight their role in shaping microbial food webs. Our vision is to provide novel cellular concepts and molecular tools to the link between the intricate mechanisms of cell signaling and stress response, and large biogeochemical cycles.
Max ERC Funding
1 999 648 €
Duration
Start date: 2011-11-01, End date: 2016-10-31
Project acronym LATELIFEHEALTH
Project Mapping the late-life health promoting mechanisms in worms and mammals
Researcher (PI) Ehud Cohen
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS5, ERC-2011-StG_20101109
Summary Aberrant protein aggregation (proteotoxicity) is an underlying mechanistic event common to numerous late-onset human neurodegenerative maladies including Alzheimer’s (AD) disease. Recent studies indicated that the ageing process plays key roles in enabling protein aggregation to become toxic late in life. The insulin/IGF signaling pathway (IIS) is a major ageing, stress resistance and lifespan regulator in worms and mice. We found that IIS reduction protects worms and mice from toxicity associated with the AD linked peptide, Aβ. These findings point to the alteration of ageing by IIS reduction as a promising research avenue towards the development of neurodegeneration therapies. In the nematode C. elegans, both effects of IIS reduction; longevity and protection from proteotoxicity are dependent on the activity of the FOXO transcription factor DAF-16. However, these functions of DAF-16/FOXO differ temporally; in worms the mediation of longevity by DAF-16 is restricted to reproductive adulthood while protection from proteotoxicity extends also to late adulthood. This differential temporal activity pattern suggests that different DAF-16 co-factors and target genes play roles in the mediation of longevity and in protection from proteotoxicity. Thus, a careful characterization of the late life DAF-16 regulated protective mechanism is required to evaluate the therapeutic potential of IIS reduction as a future treatment for neurodegenerative disorders. Here I propose to use nematodes and mice to explore the DAF-16/FOXO co-factors and target genes that mediate stress resistance and protection from proteotoxicity in the aged organism. Dual experimental approach will be utilized to achieve this goal; a directed genetic screen for the identification of co-factors and temporally differential set of DNA microarrays for the recognition of late life DAF-16/FOX target genes. This project is expected to yield new insight and to serve as a platform for future studies.
Summary
Aberrant protein aggregation (proteotoxicity) is an underlying mechanistic event common to numerous late-onset human neurodegenerative maladies including Alzheimer’s (AD) disease. Recent studies indicated that the ageing process plays key roles in enabling protein aggregation to become toxic late in life. The insulin/IGF signaling pathway (IIS) is a major ageing, stress resistance and lifespan regulator in worms and mice. We found that IIS reduction protects worms and mice from toxicity associated with the AD linked peptide, Aβ. These findings point to the alteration of ageing by IIS reduction as a promising research avenue towards the development of neurodegeneration therapies. In the nematode C. elegans, both effects of IIS reduction; longevity and protection from proteotoxicity are dependent on the activity of the FOXO transcription factor DAF-16. However, these functions of DAF-16/FOXO differ temporally; in worms the mediation of longevity by DAF-16 is restricted to reproductive adulthood while protection from proteotoxicity extends also to late adulthood. This differential temporal activity pattern suggests that different DAF-16 co-factors and target genes play roles in the mediation of longevity and in protection from proteotoxicity. Thus, a careful characterization of the late life DAF-16 regulated protective mechanism is required to evaluate the therapeutic potential of IIS reduction as a future treatment for neurodegenerative disorders. Here I propose to use nematodes and mice to explore the DAF-16/FOXO co-factors and target genes that mediate stress resistance and protection from proteotoxicity in the aged organism. Dual experimental approach will be utilized to achieve this goal; a directed genetic screen for the identification of co-factors and temporally differential set of DNA microarrays for the recognition of late life DAF-16/FOX target genes. This project is expected to yield new insight and to serve as a platform for future studies.
Max ERC Funding
1 438 899 €
Duration
Start date: 2011-10-01, End date: 2017-09-30
Project acronym LIVERMIRCOENV
Project Heterotypic Cell Interactions in Hepatitis induced Liver Cancer
Researcher (PI) Eli Pikarsky
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS6, ERC-2011-StG_20101109
Summary The link between inflammation and cancer is now established, yet the underlying molecular mechanisms are unresolved. As tumors progress, they modulate inflammatory cells towards a pro-tumorigenic phenotype. We have shown that inflammatory cells reciprocate by sculpting the parenchymal epithelial cells. I hypothesize that these reciprocal interactions lie at the heart of the link between inflammation and cancer.
Hepatocellular carcinoma (HCC), one of the deadliest tumors, is a prototype of inflammation induced cancer. My team will employ a twofold strategy to analyze the changes occuring in inflammatory cells before and after tumors emerge, based on preliminary findings showing that changes in inflammatory cells precede tumorigenesis. First, we will perform comprehensive mapping of the changing inflammatory microenvironment in a mouse model of inflammation induced HCC. We will employ genetic manipulation strategies, coupled to cell isolation techniques to delineate the molecular cues that mediate these changes and then will analyze the functional role of key mediators of these processes in HCC. Microfluidics approaches will give us a highthroughput quantitative view of these heterotypic interactions. The same approaches will be harnessed to identify the interactions that form the liver stem cell niche which dramatically expands in states of chronic inflammation. Second, drawing on our finding that a recurring tumor amplicon drives HCC progression by modulating the microenvironment, we will work towards identifying additional similar amplicons to define additional key effectors of the microenvironment.
Of special importance, heterotypic cell interactions that play key roles in both cancer initiation and progression, present ideal therapeutic targets, which are easily accessible and less amenable to mutational selection. Furthermore, the results of our experiments could also have far reaching implications in other inflammatory states and different types of cancer.
Summary
The link between inflammation and cancer is now established, yet the underlying molecular mechanisms are unresolved. As tumors progress, they modulate inflammatory cells towards a pro-tumorigenic phenotype. We have shown that inflammatory cells reciprocate by sculpting the parenchymal epithelial cells. I hypothesize that these reciprocal interactions lie at the heart of the link between inflammation and cancer.
Hepatocellular carcinoma (HCC), one of the deadliest tumors, is a prototype of inflammation induced cancer. My team will employ a twofold strategy to analyze the changes occuring in inflammatory cells before and after tumors emerge, based on preliminary findings showing that changes in inflammatory cells precede tumorigenesis. First, we will perform comprehensive mapping of the changing inflammatory microenvironment in a mouse model of inflammation induced HCC. We will employ genetic manipulation strategies, coupled to cell isolation techniques to delineate the molecular cues that mediate these changes and then will analyze the functional role of key mediators of these processes in HCC. Microfluidics approaches will give us a highthroughput quantitative view of these heterotypic interactions. The same approaches will be harnessed to identify the interactions that form the liver stem cell niche which dramatically expands in states of chronic inflammation. Second, drawing on our finding that a recurring tumor amplicon drives HCC progression by modulating the microenvironment, we will work towards identifying additional similar amplicons to define additional key effectors of the microenvironment.
Of special importance, heterotypic cell interactions that play key roles in both cancer initiation and progression, present ideal therapeutic targets, which are easily accessible and less amenable to mutational selection. Furthermore, the results of our experiments could also have far reaching implications in other inflammatory states and different types of cancer.
Max ERC Funding
1 499 940 €
Duration
Start date: 2011-10-01, End date: 2017-09-30
Project acronym NEURALCODES_EMO
Project Deciphering neural codes of valence-based emotional memories
Researcher (PI) Ron Paz
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS5, ERC-2011-StG_20101109
Summary Pioneering studies from the last decades have shown that we can use animal models to study the neural correlates of learning and emotional memories. These studies have identified a network of brain regions that co-orchestrate the dynamic processes that occur during acquisition, retrieval and update of memories with valence. In primates, these areas and their connectivity have evolved to allow a complex interaction between cognitive abilities and emotions. Although we know that emotional memories differ from neutral memories, the actual neural codes that underlie them are largely unknown. Understanding emotional memories at a detailed level in a primate model is a crucial step in our goal to understand ourselves, and would allow specific therapeutic interventions for a spectrum of psychiatric disorders.
Our primary objectives are: a. to develop a comprehensive battery of behavioral paradigms that targets emotional learning and memory in non-human primates; b. to understand the neural mechanisms that underlie long-term evolvement of these memories; c. to develop a computational and experimental framework to understand coding in these high-dimensionality networks. We combine large-scale inter- and intra-regional simultaneous electrophysiological recordings in the primate amygdala and prefrontal-cortices, with a set of focused paradigms that use behavioral generalization as a tool to probe the underlying neural building blocks. Generalization is a highly-evolved cognitive ability of primates and a desired feature of any learning algorithm. Importantly, it is a tool to modify the brain and unveil the structure of its internal representations i.e. the neural code. Together, it will form a unified framework to understand neural codes of learning and memory formation in valence-based situations. Our research will help establishing an emerging field of computational affective neuroscience, and our vision holds that Emotion is finally within the reach of neurophysiology.
Summary
Pioneering studies from the last decades have shown that we can use animal models to study the neural correlates of learning and emotional memories. These studies have identified a network of brain regions that co-orchestrate the dynamic processes that occur during acquisition, retrieval and update of memories with valence. In primates, these areas and their connectivity have evolved to allow a complex interaction between cognitive abilities and emotions. Although we know that emotional memories differ from neutral memories, the actual neural codes that underlie them are largely unknown. Understanding emotional memories at a detailed level in a primate model is a crucial step in our goal to understand ourselves, and would allow specific therapeutic interventions for a spectrum of psychiatric disorders.
Our primary objectives are: a. to develop a comprehensive battery of behavioral paradigms that targets emotional learning and memory in non-human primates; b. to understand the neural mechanisms that underlie long-term evolvement of these memories; c. to develop a computational and experimental framework to understand coding in these high-dimensionality networks. We combine large-scale inter- and intra-regional simultaneous electrophysiological recordings in the primate amygdala and prefrontal-cortices, with a set of focused paradigms that use behavioral generalization as a tool to probe the underlying neural building blocks. Generalization is a highly-evolved cognitive ability of primates and a desired feature of any learning algorithm. Importantly, it is a tool to modify the brain and unveil the structure of its internal representations i.e. the neural code. Together, it will form a unified framework to understand neural codes of learning and memory formation in valence-based situations. Our research will help establishing an emerging field of computational affective neuroscience, and our vision holds that Emotion is finally within the reach of neurophysiology.
Max ERC Funding
1 671 620 €
Duration
Start date: 2012-01-01, End date: 2017-08-31
Project acronym NEUROBAT
Project Neural codes for space in complex multi-scale environments: Insights from the bat
Researcher (PI) Nachum Ulanovsky
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS5, ERC-2011-StG_20101109
Summary Studies of spatial navigation and neural codes for space have followed two parallel tracks over the last 100 years: One research approach was to study animal navigation in the wild over large spatial scales (kilometers); this approach focused on non-mammalian species and on behavioral studies, with hardly any research on the underlying brain mechanisms. The other approach was to study the navigation of mammals (mostly rats) in mazes and small arenas; this approach revealed 'place cells' in the hippocampus, neurons that become active at specific locations; and 'grid cells' in entorhinal cortex – neurons that respond when the animal passes through the vertices of a hexagonal grid spanning the entire environment. However, it is unknown whether place- and grid-cells are relevant at all to large-scale navigation over kilometers. Thus, there is a large gap between the two parallel approaches to studying spatial memory and navigation – both a conceptual gap, and a gap in spatial scale. Here, we propose to bridge this gap, by recording from place cells and grid cells in a flying mammal – the bat – while it moves in 4 different environments of varying sizes, from centimeters to kilometers. We will conduct both standard (tethered) and wireless neural recordings, and will also pioneer the development of a novel sonar-based virtual reality system for studying large-scale navigation. The same neurons will be recorded across different spatial scales, which will allow comparing various neural-coding schemes. These new setups will allow the first testing for the existence of kilometer-sized hippocampal place-fields and entorhinal grids, in bats navigating through naturalistic virtual landscapes; they will also provide rich information on neural codes for 2-D and 3-D space in the mammalian brain. Our innovative project is expected to provide – for the first time – a true understanding of the brain mechanisms of large-scale, realistic navigation in complex 3-D environments.
Summary
Studies of spatial navigation and neural codes for space have followed two parallel tracks over the last 100 years: One research approach was to study animal navigation in the wild over large spatial scales (kilometers); this approach focused on non-mammalian species and on behavioral studies, with hardly any research on the underlying brain mechanisms. The other approach was to study the navigation of mammals (mostly rats) in mazes and small arenas; this approach revealed 'place cells' in the hippocampus, neurons that become active at specific locations; and 'grid cells' in entorhinal cortex – neurons that respond when the animal passes through the vertices of a hexagonal grid spanning the entire environment. However, it is unknown whether place- and grid-cells are relevant at all to large-scale navigation over kilometers. Thus, there is a large gap between the two parallel approaches to studying spatial memory and navigation – both a conceptual gap, and a gap in spatial scale. Here, we propose to bridge this gap, by recording from place cells and grid cells in a flying mammal – the bat – while it moves in 4 different environments of varying sizes, from centimeters to kilometers. We will conduct both standard (tethered) and wireless neural recordings, and will also pioneer the development of a novel sonar-based virtual reality system for studying large-scale navigation. The same neurons will be recorded across different spatial scales, which will allow comparing various neural-coding schemes. These new setups will allow the first testing for the existence of kilometer-sized hippocampal place-fields and entorhinal grids, in bats navigating through naturalistic virtual landscapes; they will also provide rich information on neural codes for 2-D and 3-D space in the mammalian brain. Our innovative project is expected to provide – for the first time – a true understanding of the brain mechanisms of large-scale, realistic navigation in complex 3-D environments.
Max ERC Funding
1 499 999 €
Duration
Start date: 2011-11-01, End date: 2016-10-31
Project acronym NEXTGENMOLECOL
Project Next Generation Molecular Ecology
Researcher (PI) Hans Ellegren
Host Institution (HI) Uppsala University
Call Details Advanced Grant (AdG), LS8, ERC-2009-AdG
Summary There is an immediate need to increase our understanding of the genetic basis for fitness differences in natural populations (Ellegren and Sheldon Nature 452:169-175, 2008). Fortunately, technological developments within genome research, notably the recent ability to retrieve massive amounts of DNA sequence data based on next generation sequencing , will make possible completely novel investigations of the link between genotypes and phenotypes in non-model organisms. With our background as major players in molecular ecology and evolutionary genomics of non-models for the last 15-20 years, we are excellently placed to take on a leading role in this process, developing a Next Generation Molecular Ecology . This research program will combine studies of candidate genes with large-scale gene expression analysis, several mapping approaches and comparative genomics to study the genetic basis of trait evolution in wild bird populations. First, we will search for and analyse loci involved with reproductive isolation and adaptive population divergence in a well-known system for speciation research the pied flycatcher and the collared flycatcher. A milestone of this program will be genome sequencing of the two flycatcher species. Second, we will track the genetic basis of behaviour using a unique breeding population of zebra finches and benefitting from the recently obtained genome sequence of this species. Third, we will identify the targets for adaptive evolution during avian evolution using comparative genomics. Overall, the program will be able to reveal the molecular genetic architecture behind phenotypic variation. The potential for scientific break-through in this interdisciplinary program should be significant.
Summary
There is an immediate need to increase our understanding of the genetic basis for fitness differences in natural populations (Ellegren and Sheldon Nature 452:169-175, 2008). Fortunately, technological developments within genome research, notably the recent ability to retrieve massive amounts of DNA sequence data based on next generation sequencing , will make possible completely novel investigations of the link between genotypes and phenotypes in non-model organisms. With our background as major players in molecular ecology and evolutionary genomics of non-models for the last 15-20 years, we are excellently placed to take on a leading role in this process, developing a Next Generation Molecular Ecology . This research program will combine studies of candidate genes with large-scale gene expression analysis, several mapping approaches and comparative genomics to study the genetic basis of trait evolution in wild bird populations. First, we will search for and analyse loci involved with reproductive isolation and adaptive population divergence in a well-known system for speciation research the pied flycatcher and the collared flycatcher. A milestone of this program will be genome sequencing of the two flycatcher species. Second, we will track the genetic basis of behaviour using a unique breeding population of zebra finches and benefitting from the recently obtained genome sequence of this species. Third, we will identify the targets for adaptive evolution during avian evolution using comparative genomics. Overall, the program will be able to reveal the molecular genetic architecture behind phenotypic variation. The potential for scientific break-through in this interdisciplinary program should be significant.
Max ERC Funding
2 500 000 €
Duration
Start date: 2010-04-01, End date: 2015-09-30
Project acronym OAPROGRESS
Project Evaluation of Osteoarthritis Progression in a Patient-Specific Manner using Magnetic Resonance Imaging and Computational Modeling
Researcher (PI) Rami Kristian Korhonen
Host Institution (HI) ITA-SUOMEN YLIOPISTO
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Background
Osteoarthritis (OA) is one of the most prevalent disorders of the musculoskeletal system. In OA, articular cartilage degenerates and its structure and mechanical properties change, but monitoring or predicting the progression of OA has not been possible. Magnetic resonance imaging (MRI) is a potential tool for the imaging of joint tissues, estimating cartilage structure and diagnostics of OA, whereas joint loading and estimation of stresses/strains within joint tissues necessitates computational modeling. It would be a major breakthrough if one could develop a technique where, based on MRI and computational modeling, prediction and evaluation of OA progression of a patient under a certain loading condition would be possible.
Objectives
1) to combine MRI with computational modeling for the estimation of stresses and possible failure points within human knee joints, and 2) to develop second generation adaptive models of articular cartilage for the prediction of altered tissue structure and composition during OA progression. For the model validation, cartilage structure, composition and biomechanical properties as well as cell responses in situ are characterized. At the end of the project these main aims will be merged 3) to estimate the effect of loading on cartilage degeneration during the progression of OA in a patient-specific manner.
Significance
Combining MRI information of joint tissues with computational modeling, we develop a tool to evaluate the effect of different interventions on stresses in human joints. By combining this tool with an adaptive model that can estimate the effect of loading on cartilage composition and structure, we hope to be able to predict changes in cartilage properties during OA progression in a patient-specific manner several years ahead. This would help in decision making of clinical treatments and interventions (conservative or surgical) for the prevention or further progression of OA.
Summary
Background
Osteoarthritis (OA) is one of the most prevalent disorders of the musculoskeletal system. In OA, articular cartilage degenerates and its structure and mechanical properties change, but monitoring or predicting the progression of OA has not been possible. Magnetic resonance imaging (MRI) is a potential tool for the imaging of joint tissues, estimating cartilage structure and diagnostics of OA, whereas joint loading and estimation of stresses/strains within joint tissues necessitates computational modeling. It would be a major breakthrough if one could develop a technique where, based on MRI and computational modeling, prediction and evaluation of OA progression of a patient under a certain loading condition would be possible.
Objectives
1) to combine MRI with computational modeling for the estimation of stresses and possible failure points within human knee joints, and 2) to develop second generation adaptive models of articular cartilage for the prediction of altered tissue structure and composition during OA progression. For the model validation, cartilage structure, composition and biomechanical properties as well as cell responses in situ are characterized. At the end of the project these main aims will be merged 3) to estimate the effect of loading on cartilage degeneration during the progression of OA in a patient-specific manner.
Significance
Combining MRI information of joint tissues with computational modeling, we develop a tool to evaluate the effect of different interventions on stresses in human joints. By combining this tool with an adaptive model that can estimate the effect of loading on cartilage composition and structure, we hope to be able to predict changes in cartilage properties during OA progression in a patient-specific manner several years ahead. This would help in decision making of clinical treatments and interventions (conservative or surgical) for the prevention or further progression of OA.
Max ERC Funding
1 303 056 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym ORIGINOME
Project Mammalian Origin of replication –
Genome-wide Mapping and Regulation
Researcher (PI) Itamar Simon
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS2, ERC-2011-StG_20101109
Summary Very little is known about the global organization and regulation of the replication program. Recent advances in genomic methods allow the genome-wide description of the time of replication in mammals. Nevertheless, the spatial information about replication initiations (origin of replication) is still sparse. Such information is crucial for studying mechanistic aspects of replication regulation since the time of replication is determined mainly by the time each origin is activated. In Originome I propose to develop novel genome-wide approaches that will allow origin mapping both in the cell population and the single cell levels. Applying these methods to multiple tissues, growth conditions and organisms, will put me in an excellent position for studying the regulation of the replication machinery. I am planning to combine bioinformatics, comparative genomics and reverse genetics tools to decipher the cis-acting elements that participate in the regulation of the replication program. Successful mapping of origins and characterization of their mode of regulation have broad implications not only in the field of DNA replication. First, replication stress is one of the initial causes of cancer. Therefore, I am planning to decipher the changes in the replication program that occur in response to stress in order to better understand the transformation process. Second, better understanding of replication regulation will allow studying systematically the association between time of replication, transcription and chromatin structure. Taken together, by moving the field of DNA replication from a descriptive into a mechanistic stage, Originome will set the stage for a variety of experimental approaches for deciphering replication organization and its effects on transcription, chromatin structure and cancer transformation.
Summary
Very little is known about the global organization and regulation of the replication program. Recent advances in genomic methods allow the genome-wide description of the time of replication in mammals. Nevertheless, the spatial information about replication initiations (origin of replication) is still sparse. Such information is crucial for studying mechanistic aspects of replication regulation since the time of replication is determined mainly by the time each origin is activated. In Originome I propose to develop novel genome-wide approaches that will allow origin mapping both in the cell population and the single cell levels. Applying these methods to multiple tissues, growth conditions and organisms, will put me in an excellent position for studying the regulation of the replication machinery. I am planning to combine bioinformatics, comparative genomics and reverse genetics tools to decipher the cis-acting elements that participate in the regulation of the replication program. Successful mapping of origins and characterization of their mode of regulation have broad implications not only in the field of DNA replication. First, replication stress is one of the initial causes of cancer. Therefore, I am planning to decipher the changes in the replication program that occur in response to stress in order to better understand the transformation process. Second, better understanding of replication regulation will allow studying systematically the association between time of replication, transcription and chromatin structure. Taken together, by moving the field of DNA replication from a descriptive into a mechanistic stage, Originome will set the stage for a variety of experimental approaches for deciphering replication organization and its effects on transcription, chromatin structure and cancer transformation.
Max ERC Funding
1 499 523 €
Duration
Start date: 2011-12-01, End date: 2016-11-30
Project acronym PATHEVOL
Project Linking Pathogen Evolution and Epidemiology
Researcher (PI) Anna-Liisa Laine
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Starting Grant (StG), LS8, ERC-2011-StG_20101109
Summary The goal of the proposed research is a comprehensive understanding of how evolutionary potential of pathogen populations interacts with epidemiological dynamics in natural populations. The empirical work will be conducted on the specialist fungal pathogen Podosphaera plantaginis and its host plant Plantago lanceolata in a large network of host populations. I will address the key theories of pathogen evolution, involving life-history trade-offs, competition for resources under multiple infection, and sexual reproduction. This project takes advantage of the exceptional research opportunities offered by the focal study species to test models that have not been validated with respect to realized population dynamics and the persistence of pathogen populations.
I have studied the coevolutionary dynamics between P. plantaginis and P. lanceolata for several years. Unique epidemiological data have been collected annually on the occurrence of the pathogen in a network of 4000 host populations since 2001. Recently, I have generated an EST library for the pathogen that allows and facilitates genetic studies. In the planned research, I will combine laboratory experiments with large-scale population surveys, genetic studies and mathematical modelling to achieve the objectives of this proposal.
The proposed research has potential for groundbreaking results on pathogen evolution and epidemiology through: i) Simultaneous study of multiple forces driving pathogen evolution and their importance in natural populations, with direct connections to epidemiology. ii) Development of new methodology for the study of obligate parasites like P. plantaginis. iii) Construction of a stochastic, spatially explicit epidemiological model predicting pathogen occurrence from one season to the next with applicability to a wide range of pathogens. iv) Identifying critical life-history stages and mechanisms for virulence evolution yield much needed insights and tools into the battle against disease.
Summary
The goal of the proposed research is a comprehensive understanding of how evolutionary potential of pathogen populations interacts with epidemiological dynamics in natural populations. The empirical work will be conducted on the specialist fungal pathogen Podosphaera plantaginis and its host plant Plantago lanceolata in a large network of host populations. I will address the key theories of pathogen evolution, involving life-history trade-offs, competition for resources under multiple infection, and sexual reproduction. This project takes advantage of the exceptional research opportunities offered by the focal study species to test models that have not been validated with respect to realized population dynamics and the persistence of pathogen populations.
I have studied the coevolutionary dynamics between P. plantaginis and P. lanceolata for several years. Unique epidemiological data have been collected annually on the occurrence of the pathogen in a network of 4000 host populations since 2001. Recently, I have generated an EST library for the pathogen that allows and facilitates genetic studies. In the planned research, I will combine laboratory experiments with large-scale population surveys, genetic studies and mathematical modelling to achieve the objectives of this proposal.
The proposed research has potential for groundbreaking results on pathogen evolution and epidemiology through: i) Simultaneous study of multiple forces driving pathogen evolution and their importance in natural populations, with direct connections to epidemiology. ii) Development of new methodology for the study of obligate parasites like P. plantaginis. iii) Construction of a stochastic, spatially explicit epidemiological model predicting pathogen occurrence from one season to the next with applicability to a wide range of pathogens. iv) Identifying critical life-history stages and mechanisms for virulence evolution yield much needed insights and tools into the battle against disease.
Max ERC Funding
1 498 811 €
Duration
Start date: 2011-10-01, End date: 2016-09-30
Project acronym PICHO
Project p53 control of epithelial homeostasis
Researcher (PI) Yinon Ben Neriah
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS4, ERC-2011-ADG_20110310
Summary Background: Our recent studies implicate p53 in gut tissue homeostasis - suppressing epithelial invasion. This function is tightly linked to suppression of a gene cluster (PSIS- p53-Suppressed Invasiveness Signature), which requires Wnt activation and other cues, yet is only expressed upon loss of p53. The invasive signature explains a broad spectrum of the invasiveness property, from the loss of enterocyte polarity to matrix degradation, pointing to a concerted action. We documented a tight association between invasiveness and coexpression of several PSIS genes in different mouse models and showed that PSIS expression is essential in mediating epithelial cell invasiveness following p53 depletion.
Goal: Elucidate functions of p53 activation which are of particular importance for epithelial tissues. Understand how WT p53 contributes to preserving epithelial boundaries, prohibiting invasion and abnormal cell mixture and controlling stem cell dynamics under tissue stress.
Methodology: We will investigate the epithelial role of p53 and the invasive signature genes in several mouse models of inflammatory bowel diseases and intestinal cancer. These models will incorporate p53-modulating switchable genetic elements and cell-tracking genetic markers for monitoring tissue dynamics. Analyses of relevant human pathology samples will complement the mouse studies.
Significance: Invasion is a defining hallmark of malignancy and understanding early invasion of tumor cells is of fundamental importance in designing future therapies for cancer - targeting PSIS is an example. PSIS database may also be used to develop biomarkers for distinguishing malignant tumors from benign ones, a critical determinant of therapeutic options in several types of cancers, currently solely based on morphologic assessment. A molecular definition of early invasive lesions may allow early implementation of curative treatments while withholding patient overtreatment which often results in serious morbidity.
Summary
Background: Our recent studies implicate p53 in gut tissue homeostasis - suppressing epithelial invasion. This function is tightly linked to suppression of a gene cluster (PSIS- p53-Suppressed Invasiveness Signature), which requires Wnt activation and other cues, yet is only expressed upon loss of p53. The invasive signature explains a broad spectrum of the invasiveness property, from the loss of enterocyte polarity to matrix degradation, pointing to a concerted action. We documented a tight association between invasiveness and coexpression of several PSIS genes in different mouse models and showed that PSIS expression is essential in mediating epithelial cell invasiveness following p53 depletion.
Goal: Elucidate functions of p53 activation which are of particular importance for epithelial tissues. Understand how WT p53 contributes to preserving epithelial boundaries, prohibiting invasion and abnormal cell mixture and controlling stem cell dynamics under tissue stress.
Methodology: We will investigate the epithelial role of p53 and the invasive signature genes in several mouse models of inflammatory bowel diseases and intestinal cancer. These models will incorporate p53-modulating switchable genetic elements and cell-tracking genetic markers for monitoring tissue dynamics. Analyses of relevant human pathology samples will complement the mouse studies.
Significance: Invasion is a defining hallmark of malignancy and understanding early invasion of tumor cells is of fundamental importance in designing future therapies for cancer - targeting PSIS is an example. PSIS database may also be used to develop biomarkers for distinguishing malignant tumors from benign ones, a critical determinant of therapeutic options in several types of cancers, currently solely based on morphologic assessment. A molecular definition of early invasive lesions may allow early implementation of curative treatments while withholding patient overtreatment which often results in serious morbidity.
Max ERC Funding
2 500 000 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym PREVENTRA
Project Genes, environment and immunity in the pathogenesis of rheumatoid arthritis
Researcher (PI) Lars Göran Yngve Klareskog
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary Aims and significance: The aims of this program are (1) To obtain new understanding of how environmental and life style factors interact with genes to induce immune reactions able to cause the different forms of arthritis that are defined as RA; (2) To use this understanding to develop prevention and targeted therapy for different forms of RA, and to enable efficient and eventually curative therapy. Background: We build on new understanding or RA etiology that has followed from studies on interactions between genes, environment and immunity in different subsets of RA. This has been provided for new detailed studies on specific and eventually disease-inducing autoimmunity in RA. Research program: We will use our infrastructure (longitudinal large cohorts, biobanks genetic information and our molecular immunology laboratory) to investigate (1) how genes and environment interact in causing different forms of RA; (2) how specific immune reactions against post-translationally modified (mainly citrullinated) autoantigens are triggered by environmental agents in specific genetic contexts in different individuals; (3) How these immune reactions target different organs (joints, lungs etc) and eventually cause arthritis in model systems; (4) how the combination of genes, environment and immunity may determine disease course and response to various therapies. Novelty and opportunities to take knowledge of autoimmune disease and RA to a new level: The recent advances in understanding interactions between genes, environment and immunity in RA, provides a striking new opportunity to understand basic features of autoimmunity and autoimmune disease, as well as potentials to prevent and treat RA very early. I believe that the presented program is well positioned to use this opportunity and contribute to a new paradigm for understanding and preventing RA.
Summary
Aims and significance: The aims of this program are (1) To obtain new understanding of how environmental and life style factors interact with genes to induce immune reactions able to cause the different forms of arthritis that are defined as RA; (2) To use this understanding to develop prevention and targeted therapy for different forms of RA, and to enable efficient and eventually curative therapy. Background: We build on new understanding or RA etiology that has followed from studies on interactions between genes, environment and immunity in different subsets of RA. This has been provided for new detailed studies on specific and eventually disease-inducing autoimmunity in RA. Research program: We will use our infrastructure (longitudinal large cohorts, biobanks genetic information and our molecular immunology laboratory) to investigate (1) how genes and environment interact in causing different forms of RA; (2) how specific immune reactions against post-translationally modified (mainly citrullinated) autoantigens are triggered by environmental agents in specific genetic contexts in different individuals; (3) How these immune reactions target different organs (joints, lungs etc) and eventually cause arthritis in model systems; (4) how the combination of genes, environment and immunity may determine disease course and response to various therapies. Novelty and opportunities to take knowledge of autoimmune disease and RA to a new level: The recent advances in understanding interactions between genes, environment and immunity in RA, provides a striking new opportunity to understand basic features of autoimmunity and autoimmune disease, as well as potentials to prevent and treat RA very early. I believe that the presented program is well positioned to use this opportunity and contribute to a new paradigm for understanding and preventing RA.
Max ERC Funding
2 000 000 €
Duration
Start date: 2010-04-01, End date: 2016-03-31
Project acronym PROTEINSEQ
Project Sensitive, specific high-throughput plasma proteome analysis via ProteinSeq
Researcher (PI) Dag Ake Ulf Landegren
Host Institution (HI) UPPSALA UNIVERSITET
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Despite increasing interest in biomarkers to diagnose and distinguish diseases and select treatment few new protein biomarkers are being successfully validated. Important explanations for the limited success are poor specificity and sensitivity of detection of plasma proteins using current assays, and inability to analyze large numbers of markers and samples. Improved assays should provide access to plasma protein biomarkers at levels below current detection thresholds, potentially reflecting disease processes anywhere in the body at early stages. They should also distinguish closely similar protein variants. The assays should furthermore assess numerous markers in parallel with limited consumption of biobank samples.
The aim of this project is to enable investigations of protein biomarkers at entirely new levels of performance. The basis of the proposal is our expertise in molecular tools and specifically an approach to protein analysis that addresses the above requirements.
This technology is now ready to be scaled for large-volume, high-performance assays with the following characteristics: 1) Unsurpassed specificity via simultaneous detection of three epitopes on any target protein. 2) Amplifiable DNA strands form upon specific detection, permitting ultrasensitive detection. 3) Only appropriate reagent combinations result in amplifiable reporter strands, overcoming obstacles to multiplex protein detection. 4) Tag sequences in the amplified DNA strands identify the detected proteins and the investigated samples, allowing digital information of protein abundance to be retrieved via next generation DNA sequencing of multiplex reactions for large numbers of patients in single runs.
I propose to set up and apply assays of a thousand proteins in parallel in small plasma samples, and in a subproject I will target proteins on a novel and promising class of cancer biomarkers – exosomes.
Summary
Despite increasing interest in biomarkers to diagnose and distinguish diseases and select treatment few new protein biomarkers are being successfully validated. Important explanations for the limited success are poor specificity and sensitivity of detection of plasma proteins using current assays, and inability to analyze large numbers of markers and samples. Improved assays should provide access to plasma protein biomarkers at levels below current detection thresholds, potentially reflecting disease processes anywhere in the body at early stages. They should also distinguish closely similar protein variants. The assays should furthermore assess numerous markers in parallel with limited consumption of biobank samples.
The aim of this project is to enable investigations of protein biomarkers at entirely new levels of performance. The basis of the proposal is our expertise in molecular tools and specifically an approach to protein analysis that addresses the above requirements.
This technology is now ready to be scaled for large-volume, high-performance assays with the following characteristics: 1) Unsurpassed specificity via simultaneous detection of three epitopes on any target protein. 2) Amplifiable DNA strands form upon specific detection, permitting ultrasensitive detection. 3) Only appropriate reagent combinations result in amplifiable reporter strands, overcoming obstacles to multiplex protein detection. 4) Tag sequences in the amplified DNA strands identify the detected proteins and the investigated samples, allowing digital information of protein abundance to be retrieved via next generation DNA sequencing of multiplex reactions for large numbers of patients in single runs.
I propose to set up and apply assays of a thousand proteins in parallel in small plasma samples, and in a subproject I will target proteins on a novel and promising class of cancer biomarkers – exosomes.
Max ERC Funding
2 499 600 €
Duration
Start date: 2012-03-01, End date: 2017-02-28
Project acronym PSARPS
Project Practical statistical approaches for addressing replicability problems in life sciences
Researcher (PI) Yoav Benjamini
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Advanced Grant (AdG), LS2, ERC-2011-ADG_20110310
Summary Lack of replicability of scientific discoveries has surfaced too often in recent years, and even reached the attention of the general public. An ignored cause is the inappropriate statistical treatment of two statistical problems: (1) selective inference, manifested in selecting few promising leads following the statistical analysis of the potential many, where ignoring the selection process on estimates, confidence intervals and observed significance; (2) using too optimistic a yardstick of variation with which confidence intervals set and statistical significance of the potential discovery is judged, as a result of ignoring the variability between laboratories and subjects. The first problem becomes more serious as the pool of potential discoveries increases, the second paradoxically becomes more serious as measuring ability improves, which explain why the two problems are more prominent in recent years. Both problems have statistical solutions, but the solutions are not practical as they burden the analysis to a point where the power to discover new findings is exceedingly low. Therefore, unless required by regulating agencies, scientists tend to avoid using these solutions.
I propose to develop methods that address such replicablity problems specific to medical research, epidemiology, genomics, brain research, and behavioral neuroscience. The methods include (a) new hierarchical weighted procedures, and model selection methods, that control the false discovery rate in testing; (b) shorter confidence intervals that offer false coverage-statement rate for the selected, both addressing the concern about selective inference; and (c) a compromise between using random effects models for the laboratories and subjects and treating them as fixed, to be aided by multiple laboratory database in behavior genetics and neuroscience. By serving the exact needs of scientists, while avoiding excessive protection, I expect the offered methodologies to become widely adapted.
Summary
Lack of replicability of scientific discoveries has surfaced too often in recent years, and even reached the attention of the general public. An ignored cause is the inappropriate statistical treatment of two statistical problems: (1) selective inference, manifested in selecting few promising leads following the statistical analysis of the potential many, where ignoring the selection process on estimates, confidence intervals and observed significance; (2) using too optimistic a yardstick of variation with which confidence intervals set and statistical significance of the potential discovery is judged, as a result of ignoring the variability between laboratories and subjects. The first problem becomes more serious as the pool of potential discoveries increases, the second paradoxically becomes more serious as measuring ability improves, which explain why the two problems are more prominent in recent years. Both problems have statistical solutions, but the solutions are not practical as they burden the analysis to a point where the power to discover new findings is exceedingly low. Therefore, unless required by regulating agencies, scientists tend to avoid using these solutions.
I propose to develop methods that address such replicablity problems specific to medical research, epidemiology, genomics, brain research, and behavioral neuroscience. The methods include (a) new hierarchical weighted procedures, and model selection methods, that control the false discovery rate in testing; (b) shorter confidence intervals that offer false coverage-statement rate for the selected, both addressing the concern about selective inference; and (c) a compromise between using random effects models for the laboratories and subjects and treating them as fixed, to be aided by multiple laboratory database in behavior genetics and neuroscience. By serving the exact needs of scientists, while avoiding excessive protection, I expect the offered methodologies to become widely adapted.
Max ERC Funding
1 933 200 €
Duration
Start date: 2012-03-01, End date: 2018-02-28
Project acronym risk factors cancer
Project Genetic and environmental risk factors for common malignant tumours especially breast cancer and melanoma
Researcher (PI) Lars Håkan Olsson
Host Institution (HI) LUNDS UNIVERSITET
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary A prerequisite for prevention or early diagnosis of a tumour disease is that environmental and/or genetic risk factors are characterized in order to better define risk groups. The present research proposal focus on common malignant tumours, especially breast cancer and malignant melanoma . By combining risk factor studies on endogenous and exogenous environmental, and genetic risk factors and its interaction, the aim is to better characterize strong determinants of risk. The project also aims at better understanding the mechanisms of disease and for different exposures, such as sun exposure and different hormonal exposures, obtain a global assessment of possible positive and negative health and disease effects. The infrastructure includes large population based cohort and case-control studies, pathological and clinical patient information, biobanks from cancer patients and controls, availability of excellent genomic resources and an extensive network of national and international collaborations. Very often it has been possible to work out from a population based perspective. Both for breast cancer and melanoma the research group has identified important genes or modifiers of dominant predisposing genes as well as environmental or constitutional risk factors. Through an extensive international collaboration gene and gene-environemnt interaction studies are undertaken. The ulitmate goal for the new knowledge is to prevent or early diagnose the malignancy. In hereditary cancer and for some of the hormonal exposures successful results already are seen. The applicant has been involved more than 30 years in epidemiological research of cancer, and published more than 400 publications in international journals and fostered a large number of PhD students.
Summary
A prerequisite for prevention or early diagnosis of a tumour disease is that environmental and/or genetic risk factors are characterized in order to better define risk groups. The present research proposal focus on common malignant tumours, especially breast cancer and malignant melanoma . By combining risk factor studies on endogenous and exogenous environmental, and genetic risk factors and its interaction, the aim is to better characterize strong determinants of risk. The project also aims at better understanding the mechanisms of disease and for different exposures, such as sun exposure and different hormonal exposures, obtain a global assessment of possible positive and negative health and disease effects. The infrastructure includes large population based cohort and case-control studies, pathological and clinical patient information, biobanks from cancer patients and controls, availability of excellent genomic resources and an extensive network of national and international collaborations. Very often it has been possible to work out from a population based perspective. Both for breast cancer and melanoma the research group has identified important genes or modifiers of dominant predisposing genes as well as environmental or constitutional risk factors. Through an extensive international collaboration gene and gene-environemnt interaction studies are undertaken. The ulitmate goal for the new knowledge is to prevent or early diagnose the malignancy. In hereditary cancer and for some of the hormonal exposures successful results already are seen. The applicant has been involved more than 30 years in epidemiological research of cancer, and published more than 400 publications in international journals and fostered a large number of PhD students.
Max ERC Funding
1 200 000 €
Duration
Start date: 2012-04-01, End date: 2017-03-31
Project acronym RUBICAN
Project RNF20 and H2B ubiquitination: linking chromatin dynamics, transcriptional control and cancer
Researcher (PI) Moshe Oren
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS4, ERC-2011-ADG_20110310
Summary Chromatin modifications play a major role in regulating genome function. Perturbations in such modifications can contribute to neoplastic processes. We will focus on a specific chromatin modification: histone H2B monoubiquitylation. The significance of monoubiquitylated H2B (H2Bub) will be studied by manipulating RNF20, the major E3 ubiquitin ligase responsible for H2B ubiquitylation as part of a heteromeric complex with RNF40.
In one major line of research, we will assess the biochemistry of RNF20/H2Bub. The effects of RNF20/H2B on gene expression will be explored through identification of proteins that interact with H2Bub and through in vitro incorporation of H2Bub into nucleosomes. Effects of H2Bub on transcription elongation will be studied by a new high resolution ChIP-seq method (NET-seq). Based on recent ChIP-seq data, we will also explore links between H2B and regulation of splicing. Furthermore, we will investigate the regulatory crosstalk between H2Bub and microRNAs.
The other major line of research will explore the biology of RNF20/H2Bub, with particular emphasis on cancer-related processes. This will be addressed through a combination of cell culture models and mouse models, including constitutive and conditional RNF20 knockout mice. The contribution of RNF20/H2Bub to various differentiation programs, with particular emphasis on embryonic stem cell differentiation, will also be investigated. In addition, we will study the impact of RNF20/H2Bub on NF-kB activity and on inflammatory responses; this will combine in vitro and in vivo systems, with emphasis on inflammation-related cancer. Finally, we will monitor changes in RNF20, RNF40 and H2Bub status in human tumors and investigate underlying mechanisms.
Summary
Chromatin modifications play a major role in regulating genome function. Perturbations in such modifications can contribute to neoplastic processes. We will focus on a specific chromatin modification: histone H2B monoubiquitylation. The significance of monoubiquitylated H2B (H2Bub) will be studied by manipulating RNF20, the major E3 ubiquitin ligase responsible for H2B ubiquitylation as part of a heteromeric complex with RNF40.
In one major line of research, we will assess the biochemistry of RNF20/H2Bub. The effects of RNF20/H2B on gene expression will be explored through identification of proteins that interact with H2Bub and through in vitro incorporation of H2Bub into nucleosomes. Effects of H2Bub on transcription elongation will be studied by a new high resolution ChIP-seq method (NET-seq). Based on recent ChIP-seq data, we will also explore links between H2B and regulation of splicing. Furthermore, we will investigate the regulatory crosstalk between H2Bub and microRNAs.
The other major line of research will explore the biology of RNF20/H2Bub, with particular emphasis on cancer-related processes. This will be addressed through a combination of cell culture models and mouse models, including constitutive and conditional RNF20 knockout mice. The contribution of RNF20/H2Bub to various differentiation programs, with particular emphasis on embryonic stem cell differentiation, will also be investigated. In addition, we will study the impact of RNF20/H2Bub on NF-kB activity and on inflammatory responses; this will combine in vitro and in vivo systems, with emphasis on inflammation-related cancer. Finally, we will monitor changes in RNF20, RNF40 and H2Bub status in human tumors and investigate underlying mechanisms.
Max ERC Funding
2 500 000 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym SENSTRIATUM
Project Sensory Integration in the Striatal Microcircuit
Researcher (PI) Gilad Daniel Silberberg
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Starting Grant (StG), LS5, ERC-2011-StG_20101109
Summary Motor behaviour requires the meaningful integration of a multitude of sensory information. The basal ganglia are essential for such sensory-motor processing and underlie motor planning, performance, and learning. The striatum is the input layer of the basal ganglia, acting as a “hub” that receives synaptic inputs from different brain regions, interacting primarily with the GABAergic striatal microcircuit. Sensory excitatory inputs to striatum arise from the thalamus and neocortex, targeting striatal projection neurons and interneurons.
Previous work in basal ganglia focused mainly on their role in motor and reward related functions, but the functional role of striatum in sensory processing is largely unknown. In this study I will elucidate the principles of sensory processing performed at the striatal microcircuit. In particular, I aim to answer these fundamental questions:
- How do striatal neurons integrate sensory input? How is sensory input from different sensory modalities integrated in the striatum? How are ipsi- and contralateral inputs integrated?
- What are the respective roles of cortical and thalamic sensory inputs in striatal function?
- How is the intra-striatal microcircuitry organized to support sensory integration?
To address these questions I will use a combination of electrophysiological, optical, and anatomical methods, including:
- In vivo whole-cell recordings from striatal neurons during visual and tactile stimulation.
- Multi-neuron whole-cell recordings in corticostriatal and thalamostriatal slices.
- Optical manipulation of identified neuronal subpopulations in slice and in vivo.
The proposed study will provide a new understanding of sensory processing at the level of basal ganglia and may provide insights regarding basal ganglia dysfunction.
Summary
Motor behaviour requires the meaningful integration of a multitude of sensory information. The basal ganglia are essential for such sensory-motor processing and underlie motor planning, performance, and learning. The striatum is the input layer of the basal ganglia, acting as a “hub” that receives synaptic inputs from different brain regions, interacting primarily with the GABAergic striatal microcircuit. Sensory excitatory inputs to striatum arise from the thalamus and neocortex, targeting striatal projection neurons and interneurons.
Previous work in basal ganglia focused mainly on their role in motor and reward related functions, but the functional role of striatum in sensory processing is largely unknown. In this study I will elucidate the principles of sensory processing performed at the striatal microcircuit. In particular, I aim to answer these fundamental questions:
- How do striatal neurons integrate sensory input? How is sensory input from different sensory modalities integrated in the striatum? How are ipsi- and contralateral inputs integrated?
- What are the respective roles of cortical and thalamic sensory inputs in striatal function?
- How is the intra-striatal microcircuitry organized to support sensory integration?
To address these questions I will use a combination of electrophysiological, optical, and anatomical methods, including:
- In vivo whole-cell recordings from striatal neurons during visual and tactile stimulation.
- Multi-neuron whole-cell recordings in corticostriatal and thalamostriatal slices.
- Optical manipulation of identified neuronal subpopulations in slice and in vivo.
The proposed study will provide a new understanding of sensory processing at the level of basal ganglia and may provide insights regarding basal ganglia dysfunction.
Max ERC Funding
1 494 445 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym SINGLE-CELL GENOMICS
Project Single-cell Gene Regulation in Differentiation and Pluripotency
Researcher (PI) Thore Rickard Hakan Sandberg
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Starting Grant (StG), LS2, ERC-2009-StG
Summary We aim to study transcriptomes with single-cell resolution, a long-standing goal in biology, to answer fundamental questions about gene regulation. The main objective concerns gene regulation during in vivo differentiation and in pluripotent cells by studying single-cells from murine preimplantation embryos, a model system with natural single-cell resolution, important biology and medical potential. This would also allow us to explore general regulatory principles of gene expression programs of individual cells. This research program will be accomplished by novel deep sequencing technology of mRNAs (mRNA-Seq) to obtain quantitative, unbiased and genome-wide gene and isoform expression measurements. We are therefore developing new experimental and computational methods for genome-wide analyses of transcriptomes at single-cell resolution. The biological significances of the proposed research are unique insights into early embryonic development. Deep sequencing of transcriptomes will also reveal post-transcriptional gene regulation important for pluripotent cells and identified pluripotency-specific gene and isoform expressions will be important for future stem cell based therapies. The inherit single-cell nature of the model system together with its important biology makes it a model systems exceptionally well suited for a systems biology approach aiming to characterize gene regulation at single-cell resolution. The novel methodology has tremendous potential to enable complete mRNA characterization of individual cells. The deep sequencing approach with state-of-the-art computational analyses is both more quantitative than previous methods and it will give readouts on alternative isoforms generated by alternative promoters, splicing and polyadenylation.
Summary
We aim to study transcriptomes with single-cell resolution, a long-standing goal in biology, to answer fundamental questions about gene regulation. The main objective concerns gene regulation during in vivo differentiation and in pluripotent cells by studying single-cells from murine preimplantation embryos, a model system with natural single-cell resolution, important biology and medical potential. This would also allow us to explore general regulatory principles of gene expression programs of individual cells. This research program will be accomplished by novel deep sequencing technology of mRNAs (mRNA-Seq) to obtain quantitative, unbiased and genome-wide gene and isoform expression measurements. We are therefore developing new experimental and computational methods for genome-wide analyses of transcriptomes at single-cell resolution. The biological significances of the proposed research are unique insights into early embryonic development. Deep sequencing of transcriptomes will also reveal post-transcriptional gene regulation important for pluripotent cells and identified pluripotency-specific gene and isoform expressions will be important for future stem cell based therapies. The inherit single-cell nature of the model system together with its important biology makes it a model systems exceptionally well suited for a systems biology approach aiming to characterize gene regulation at single-cell resolution. The novel methodology has tremendous potential to enable complete mRNA characterization of individual cells. The deep sequencing approach with state-of-the-art computational analyses is both more quantitative than previous methods and it will give readouts on alternative isoforms generated by alternative promoters, splicing and polyadenylation.
Max ERC Funding
1 654 384 €
Duration
Start date: 2010-02-01, End date: 2015-01-31
Project acronym SIRAID
Project SIRT6 activation for countering age-related metabolic diseases
Researcher (PI) Haim Cohen
Host Institution (HI) BAR ILAN UNIVERSITY
Call Details Starting Grant (StG), LS4, ERC-2009-StG
Summary The significant increase in the human lifespan during the last century confronts us with great medical challenges. To answer them, one must understand and control the mechanisms that determine the rate of ageing. The sirtuins, and in particular the mammalian member SIRT6, are a family of NAD+ dependent deacetylases that were implicated in ageing and the regulation of metabolism. Much evidence correlates SIRT6 with the regulation of ageing, primarily the manifestation of ageing related pathologies in SIRT6 deficient mice, and the induction of SIRT6 by calorie-restricted diet that delays ageing and reduces its related diseases. Nonetheless, the role of SIRT6 in ageing and the mechanisms by which it might act are still elusive. To explore it at the molecular mechanistic level, SIRAID aims to i) study the role of SIRT6 in glucose and fat metabolism under high fat diet; ii) to determine whether SIRT6 is involved in regulating life span, and to characterise how SIRT6 is activated by calorie restriction; and iii) to perform large scale SILAC-based proteomics screening for SIRT6 substrates. These results will then be used for the development of small activator molecules of SIRT6 that may be used therapeutically for age related metabolic diseases. Taken together, we suggest a multifaceted approach that will allow us to explore the role of SIRT6 in ageing and metabolism, and to translate this knowledge to counter and prevent the medical problems associated with human longevity.
Summary
The significant increase in the human lifespan during the last century confronts us with great medical challenges. To answer them, one must understand and control the mechanisms that determine the rate of ageing. The sirtuins, and in particular the mammalian member SIRT6, are a family of NAD+ dependent deacetylases that were implicated in ageing and the regulation of metabolism. Much evidence correlates SIRT6 with the regulation of ageing, primarily the manifestation of ageing related pathologies in SIRT6 deficient mice, and the induction of SIRT6 by calorie-restricted diet that delays ageing and reduces its related diseases. Nonetheless, the role of SIRT6 in ageing and the mechanisms by which it might act are still elusive. To explore it at the molecular mechanistic level, SIRAID aims to i) study the role of SIRT6 in glucose and fat metabolism under high fat diet; ii) to determine whether SIRT6 is involved in regulating life span, and to characterise how SIRT6 is activated by calorie restriction; and iii) to perform large scale SILAC-based proteomics screening for SIRT6 substrates. These results will then be used for the development of small activator molecules of SIRT6 that may be used therapeutically for age related metabolic diseases. Taken together, we suggest a multifaceted approach that will allow us to explore the role of SIRT6 in ageing and metabolism, and to translate this knowledge to counter and prevent the medical problems associated with human longevity.
Max ERC Funding
1 510 968 €
Duration
Start date: 2009-10-01, End date: 2014-09-30
Project acronym TARGETING_CANCER
Project Eradication of tumors by targeting dsRNA selectively to cancer cells and recruitment of the innate immune system
Researcher (PI) Alexander Levitzki
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary We have recently shown that EGFR over-expressing tumors can be eradicated by an EGFR homing chemical vector, carrying dsRNA. The vector is PolyInosine/Cytosine (PolyIC) bound to Polyethleneimine-Polyethyleneglycol-EGF (PEI-PEG-EGF, PPE). We have shown that even tumors in which up to 50% of cells do not express EGFR are eradicated, due to the strong tumor-localized bystander effects, which involve the innate immune system. Using this EGFR homing vector we have been able to eradicate EGFR overexpressing tumors by either local or systemic application. Since the success of this strategy seems to be due to the strong bystander effects induced by the internalized PolyIC it is likely that heterogeneous tumors, in which only a portion of the cells harbor the targeted receptor, will be eradicated too, as shown in our preliminary studies (PloS Med, 2006). This strategy actually targets the innate immune system to the tumor. We propose to establish tumors in which decreasing portions of cells over-express EGFR and determine the lowest number of EGFR over-expressing cells that can yield tumor eradication by the lowest dose of PolyIC/PPE. The principle behind the success of the Trojan horse approach is that the targeting moiety, EGF, is tethered to the other components of the vector in such a way that it retains its native EGFR binding properties and its ability to internalize with the receptor. The composition of the vector is such that the ligand EGF can be replaced by any other ligand, if the appropriate coupling conditions are used, retaining the ability of the ligand to bind to the target protein and internalize with it. We propose to replace EGF by a number of other ligands, such PSMA binding ligand (targeting prostate cancer) and Her-2 affibodies. Although only a fraction of women who over-express Her-2 respond to Herceptin, it is likely that they will respond to PolyIC/PP-Her-2 affibody.
Summary
We have recently shown that EGFR over-expressing tumors can be eradicated by an EGFR homing chemical vector, carrying dsRNA. The vector is PolyInosine/Cytosine (PolyIC) bound to Polyethleneimine-Polyethyleneglycol-EGF (PEI-PEG-EGF, PPE). We have shown that even tumors in which up to 50% of cells do not express EGFR are eradicated, due to the strong tumor-localized bystander effects, which involve the innate immune system. Using this EGFR homing vector we have been able to eradicate EGFR overexpressing tumors by either local or systemic application. Since the success of this strategy seems to be due to the strong bystander effects induced by the internalized PolyIC it is likely that heterogeneous tumors, in which only a portion of the cells harbor the targeted receptor, will be eradicated too, as shown in our preliminary studies (PloS Med, 2006). This strategy actually targets the innate immune system to the tumor. We propose to establish tumors in which decreasing portions of cells over-express EGFR and determine the lowest number of EGFR over-expressing cells that can yield tumor eradication by the lowest dose of PolyIC/PPE. The principle behind the success of the Trojan horse approach is that the targeting moiety, EGF, is tethered to the other components of the vector in such a way that it retains its native EGFR binding properties and its ability to internalize with the receptor. The composition of the vector is such that the ligand EGF can be replaced by any other ligand, if the appropriate coupling conditions are used, retaining the ability of the ligand to bind to the target protein and internalize with it. We propose to replace EGF by a number of other ligands, such PSMA binding ligand (targeting prostate cancer) and Her-2 affibodies. Although only a fraction of women who over-express Her-2 respond to Herceptin, it is likely that they will respond to PolyIC/PP-Her-2 affibody.
Max ERC Funding
2 054 340 €
Duration
Start date: 2010-02-01, End date: 2015-01-31
Project acronym TMIHCV
Project Microfabrication-Based Rational Design of Transcriptional-Metabolic Intervention for the Treatment of Hepatitis C Virus (HCV) Infection
Researcher (PI) Yaakov Nahmias
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS9, ERC-2009-StG
Summary Hepatitis C Virus (HCV) infection affects over 3% of the world population and is the leading cause of chronic liver disease worldwide. Current treatments are effective in only 50% of the cases and associated with significant side effects. Therefore, there is a pressing need for the development of alternative treatments. Recently, our group and others demonstrated that the HCV lifecycle is critically dependent on host lipid metabolism. In this context, we demonstrated that the grapefruit flavonoid naringenin blocks HCV production through PPAR± and LXR±, transcriptional regulators of hepatic lipid metabolism. While these results are promising, our ability to rationally control metabolic pathways in infected cells is limited due to an incomplete understanding of the regulation of hepatic metabolism by its underlying transcriptional network. This project aims to develop a comprehensive model of hepatic metabolism by integrating metabolic fluxes with transcriptional regulation enabling the rational design of transcriptional-interventions which will minimize HCV replication and release. Our approach is to develop two microfabricated platforms that will enable high-throughput data acquisition and a human-relevant screening. One component is the Transcriptional Activity Array (TAA), a microdevice for the high-throughput temporal acquisition of transcriptional activity data. The second is the Portal Circulation Platform (PCP) which integrates intestinal absorption module with a liver metabolism compartment enabling the high-throughput human-relevant screening of treatments as a substitute to animal experiments. This work will lead to the development of novel drug combinations for the treatment of HCV infection and impact the treatment of diabetes, obesity, and dyslipidemia.
Summary
Hepatitis C Virus (HCV) infection affects over 3% of the world population and is the leading cause of chronic liver disease worldwide. Current treatments are effective in only 50% of the cases and associated with significant side effects. Therefore, there is a pressing need for the development of alternative treatments. Recently, our group and others demonstrated that the HCV lifecycle is critically dependent on host lipid metabolism. In this context, we demonstrated that the grapefruit flavonoid naringenin blocks HCV production through PPAR± and LXR±, transcriptional regulators of hepatic lipid metabolism. While these results are promising, our ability to rationally control metabolic pathways in infected cells is limited due to an incomplete understanding of the regulation of hepatic metabolism by its underlying transcriptional network. This project aims to develop a comprehensive model of hepatic metabolism by integrating metabolic fluxes with transcriptional regulation enabling the rational design of transcriptional-interventions which will minimize HCV replication and release. Our approach is to develop two microfabricated platforms that will enable high-throughput data acquisition and a human-relevant screening. One component is the Transcriptional Activity Array (TAA), a microdevice for the high-throughput temporal acquisition of transcriptional activity data. The second is the Portal Circulation Platform (PCP) which integrates intestinal absorption module with a liver metabolism compartment enabling the high-throughput human-relevant screening of treatments as a substitute to animal experiments. This work will lead to the development of novel drug combinations for the treatment of HCV infection and impact the treatment of diabetes, obesity, and dyslipidemia.
Max ERC Funding
1 994 395 €
Duration
Start date: 2010-04-01, End date: 2015-03-31
Project acronym TransporterPGx
Project Transporter pharmacogenomics – the contribution of transporters to variability in drug response
Researcher (PI) Mikko Olavi Niemi
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary The response to drug therapy varies widely between individuals. Proteins involved in the absorption, distribution, metabolism and excretion of drugs play a central role in determining the concentration of a drug at the target site, and thus drug efficacy and toxicity. Transporters are membrane proteins that mediate the translocation of chemicals into and out of cells using active and passive mechanisms. We have identified a single nucleotide variant in the SLCO1B1 gene encoding the organic anion transporting polypeptide 1B1 (OATP1B1), which severely impairs the hepatic uptake of the cholesterol-lowering drug simvastatin leading to an increased systemic exposure to the drug, and a markedly increased risk of simvastatin-induced muscle toxicity. The effects of this variant differ significantly between statins, forming a rational basis for individualized lipid-lowering therapy. In addition to OATP1B1, also OATP1A2, OATP1B3, and OATP2B1 are known to transport several drugs in vitro (e.g., anticancer, cardiovascular and anti-infective drugs). However, the roles of these transporters in the pharmacokinetics of drugs in vivo in humans are unknown. The aim of this project is to systematically search for genetic variants of SLCO1A2, SLCO1B3 and SLCO2B1, which have functional significance in vivo in humans. This project will enable studies to determine the roles of these transporters in the pharmacokinetics of drugs and in the disposition of endogenous compounds in vivo, with implications for drug development and drug safety. Moreover, functionally significant variants in these genes may be used to personalize drug therapies. Overall, the project can significantly facilitate the development of new drugs and can improve the safe and effective use of drugs already in clinical use, thus increasing the health and well-being of mankind and reducing the overall costs of healthcare and drug development.
Summary
The response to drug therapy varies widely between individuals. Proteins involved in the absorption, distribution, metabolism and excretion of drugs play a central role in determining the concentration of a drug at the target site, and thus drug efficacy and toxicity. Transporters are membrane proteins that mediate the translocation of chemicals into and out of cells using active and passive mechanisms. We have identified a single nucleotide variant in the SLCO1B1 gene encoding the organic anion transporting polypeptide 1B1 (OATP1B1), which severely impairs the hepatic uptake of the cholesterol-lowering drug simvastatin leading to an increased systemic exposure to the drug, and a markedly increased risk of simvastatin-induced muscle toxicity. The effects of this variant differ significantly between statins, forming a rational basis for individualized lipid-lowering therapy. In addition to OATP1B1, also OATP1A2, OATP1B3, and OATP2B1 are known to transport several drugs in vitro (e.g., anticancer, cardiovascular and anti-infective drugs). However, the roles of these transporters in the pharmacokinetics of drugs in vivo in humans are unknown. The aim of this project is to systematically search for genetic variants of SLCO1A2, SLCO1B3 and SLCO2B1, which have functional significance in vivo in humans. This project will enable studies to determine the roles of these transporters in the pharmacokinetics of drugs and in the disposition of endogenous compounds in vivo, with implications for drug development and drug safety. Moreover, functionally significant variants in these genes may be used to personalize drug therapies. Overall, the project can significantly facilitate the development of new drugs and can improve the safe and effective use of drugs already in clinical use, thus increasing the health and well-being of mankind and reducing the overall costs of healthcare and drug development.
Max ERC Funding
1 882 212 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym TREATPD
Project Cell and gene therapy based approaches for treatment of Parkinson's disease: from models to clinics
Researcher (PI) Deniz Kirik
Host Institution (HI) LUNDS UNIVERSITET
Call Details Starting Grant (StG), LS5, ERC-2009-StG
Summary Parkinson s disease is one of the common causes of disability in the aging population, representing a major health problem for the affected individuals and a socioeconomic burden to the society. In the present proposal, the applicant puts forward an ambitious but feasible program to tackle a number of significant issues that remain unsolved in the field. He combines his strong track record in animal models of Parkinson s disease and novel cell and gene therapy-based therapeutic strategies with powerful bio-imaging techniques in order to make bold steps towards translation of new and better treatments to patients suffering from this illness. He does so in a manner that combines, on one hand, the strength of clearly-defined hypotheses and well-established tools for results towards clinical translation, with high-risk high-reward projects that hold the potential to yield ground-breaking discoveries in implementation of novel imaging techniques, on the other.
Summary
Parkinson s disease is one of the common causes of disability in the aging population, representing a major health problem for the affected individuals and a socioeconomic burden to the society. In the present proposal, the applicant puts forward an ambitious but feasible program to tackle a number of significant issues that remain unsolved in the field. He combines his strong track record in animal models of Parkinson s disease and novel cell and gene therapy-based therapeutic strategies with powerful bio-imaging techniques in order to make bold steps towards translation of new and better treatments to patients suffering from this illness. He does so in a manner that combines, on one hand, the strength of clearly-defined hypotheses and well-established tools for results towards clinical translation, with high-risk high-reward projects that hold the potential to yield ground-breaking discoveries in implementation of novel imaging techniques, on the other.
Max ERC Funding
1 508 940 €
Duration
Start date: 2009-11-01, End date: 2014-10-31
Project acronym Triploid Block
Project Mechanisms of polyploidy-mediated postzygotic reproductive isolation
Researcher (PI) Claudia Köhler
Host Institution (HI) SVERIGES LANTBRUKSUNIVERSITET
Call Details Starting Grant (StG), LS3, ERC-2011-StG_20101109
Summary Polyploidization is a widespread phenomenon among plants and is considered a major speciation mechanism. Before becoming evolutionary successful, newly formed polyploids often have to overcome fertility bottlenecks, because mating with partners of lower ploidy causes incompatibilities in the endosperm leading to the formation of mainly non-viable progeny. This reproductive barrier is called the triploid block. Nevertheless, the most frequent route to polyploid formation is probably through unreduced gametes, suggesting that the triploid block can be overcome. Recent work from our laboratory uncovered a genetic pathway leading to unreduced gamete formation at high frequency and revealed that the triploid block is mainly caused by malfunction of Polycomb group (PcG) proteins. PcG proteins are evolutionary conserved proteins, which assemble into multimeric complexes with chromatin-modifying enzymatic activity, implicating epigenetic regulatory mechanisms as an important element of speciation. Here, I propose to unravel the underlying molecular mechanism(s) of the triploid block by identifying the responsible genes causing endosperm failure upon deregulation and their mechanism of regulation in response to interploidy crosses. I also plan to investigate whether genes that contribute to the triploid block are as well responsible for establishing interspecies incompatibilities within the Arabidopsis genus. This project will combine genetics, genomics and epigenomics and will make extensive use of knowledge and tools that we have been established in my laboratory over the recent years, making it likely that the proposed objectives can be achieved. The results of this project will be of interest to a broad scientific community, including biologists with a strong interest in epigenetic mechanisms as well as ecologists interested to understand mechanisms of plant speciation.
Summary
Polyploidization is a widespread phenomenon among plants and is considered a major speciation mechanism. Before becoming evolutionary successful, newly formed polyploids often have to overcome fertility bottlenecks, because mating with partners of lower ploidy causes incompatibilities in the endosperm leading to the formation of mainly non-viable progeny. This reproductive barrier is called the triploid block. Nevertheless, the most frequent route to polyploid formation is probably through unreduced gametes, suggesting that the triploid block can be overcome. Recent work from our laboratory uncovered a genetic pathway leading to unreduced gamete formation at high frequency and revealed that the triploid block is mainly caused by malfunction of Polycomb group (PcG) proteins. PcG proteins are evolutionary conserved proteins, which assemble into multimeric complexes with chromatin-modifying enzymatic activity, implicating epigenetic regulatory mechanisms as an important element of speciation. Here, I propose to unravel the underlying molecular mechanism(s) of the triploid block by identifying the responsible genes causing endosperm failure upon deregulation and their mechanism of regulation in response to interploidy crosses. I also plan to investigate whether genes that contribute to the triploid block are as well responsible for establishing interspecies incompatibilities within the Arabidopsis genus. This project will combine genetics, genomics and epigenomics and will make extensive use of knowledge and tools that we have been established in my laboratory over the recent years, making it likely that the proposed objectives can be achieved. The results of this project will be of interest to a broad scientific community, including biologists with a strong interest in epigenetic mechanisms as well as ecologists interested to understand mechanisms of plant speciation.
Max ERC Funding
1 447 596 €
Duration
Start date: 2011-10-01, End date: 2016-09-30
Project acronym WTBLDOHRNCE
Project Walking the tightrope between life and death: Oxygen homeostasis regulation in the nematode Caenorhabditis elegans
Researcher (PI) Einav Gross
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary Oxygen (O2) is vital for the life of all aerobic animals. However, fine-tuned regulation of O2 levels is crucial since both shortage (hypoxia) and excess (via the production of reactive oxygen species, ROS) may be harmful. Indeed, both hypoxia and ROS may underlie the pathophysiology of many diseases such as atherosclerosis and Alzheimer’s. To understand how this fine-tuned O2 regulation is achieved at both the molecular and organismal levels my research proposal aims to explore the following integrated questions, using the nematode C. elegans as a model organism.
1) How do animal sense O2? What are the molecular sensors and how do they act together to fine-tune O2 responses?
2) How does O2 regulate food intake, and repress appetite in hypoxia?
3) How do animals survive and behaviorally adapt to hypoxia without HIF-1?
4) How hydrogen sulfide (H2S) regulates O2 responses and aging?
5) How do animals protect against mRNA oxidation damage?
I have focused my research on the globins. GLB-5 is a C. elegans hexacoordinated globin that regulates foraging behavior in response to subtle changes in O2 concentration. Like neuroglobin and cytoglobin in our brain, GLB-5 is expressed in neurons. Recently I discovered that GLB-5 regulates the re-adaptation of animals to 21% O2 after hypoxia. To understand how GLB-5 regulates hypoxia-reoxygenation responses I made a mutagenesis screen and isolated four classes of GLB-5 suppressors, and mapped them using single-nucleotide polymorphisms (SNP’s) to about a 1 Mbp genomic interval. Using a novel non-PCR based libraries preparation and Next Generation whole-genome sequencing, I have already sequenced four independent mutations and cloned one of the GLB-5 suppressors. In the future, I intend to clone more suppressor genes, and use this methodology in other parts of my project. By doing so, I aim to understand O2 homeostasis regulation at all levels; from the molecular signaling network to the physiology and behavior of the whole animal.
Summary
Oxygen (O2) is vital for the life of all aerobic animals. However, fine-tuned regulation of O2 levels is crucial since both shortage (hypoxia) and excess (via the production of reactive oxygen species, ROS) may be harmful. Indeed, both hypoxia and ROS may underlie the pathophysiology of many diseases such as atherosclerosis and Alzheimer’s. To understand how this fine-tuned O2 regulation is achieved at both the molecular and organismal levels my research proposal aims to explore the following integrated questions, using the nematode C. elegans as a model organism.
1) How do animal sense O2? What are the molecular sensors and how do they act together to fine-tune O2 responses?
2) How does O2 regulate food intake, and repress appetite in hypoxia?
3) How do animals survive and behaviorally adapt to hypoxia without HIF-1?
4) How hydrogen sulfide (H2S) regulates O2 responses and aging?
5) How do animals protect against mRNA oxidation damage?
I have focused my research on the globins. GLB-5 is a C. elegans hexacoordinated globin that regulates foraging behavior in response to subtle changes in O2 concentration. Like neuroglobin and cytoglobin in our brain, GLB-5 is expressed in neurons. Recently I discovered that GLB-5 regulates the re-adaptation of animals to 21% O2 after hypoxia. To understand how GLB-5 regulates hypoxia-reoxygenation responses I made a mutagenesis screen and isolated four classes of GLB-5 suppressors, and mapped them using single-nucleotide polymorphisms (SNP’s) to about a 1 Mbp genomic interval. Using a novel non-PCR based libraries preparation and Next Generation whole-genome sequencing, I have already sequenced four independent mutations and cloned one of the GLB-5 suppressors. In the future, I intend to clone more suppressor genes, and use this methodology in other parts of my project. By doing so, I aim to understand O2 homeostasis regulation at all levels; from the molecular signaling network to the physiology and behavior of the whole animal.
Max ERC Funding
1 495 922 €
Duration
Start date: 2011-11-01, End date: 2017-10-31
Project acronym YIELD
Project Is there a limit to yield?
Researcher (PI) Daniel Zamir
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS9, ERC-2011-ADG_20110310
Summary Plant breeders are challenged with sustaining global crop improvements. Is there a limit to crop yield? This project will address this central question using processing tomatoes as a model for a mechanized crop. By integrating in a single web-based platform of ‘Phenom Networks’ a broad germplasm base, deep phenotypes, and multiple genome sequences of tomato species, we will identify the genes and mechanisms that dictate crop productivity and implement them in the creation of next generation F1 hybrids. Our work is founded on our years of efforts to establish the following integrated genetic pillars: 1) The tomato genome revealed SNPs for broader marker-assisted selection between cultivated parents and untapped diversity from closely-related red-fruited ancestors. We will develop new elite parental lines into which all discovered traits will be introduced. 2) We will enrich the narrow genetic base of modern processing tomato by pyramiding recessive, additive, dominant and overdominant QTL from six wild species introgression lines (ILs) and field-test them in diverse environments. 3) By producing hybrids with ‘recessive’ deleterious mutants we have identified heterosis genes that increase yield by ~50%; these will be combined with the IL QTL. 4) Finally, we will focus on newly discovered “stability QTL” that significantly improve the reproducibility of yield effects by canalizing this phenotype in spite of environmental perturbations. This multi-faceted integrated tomato breeding effort will unite classical and genomics assisted methods with statistical genetics to demonstrate that yield barriers of the leading commercial hybrids are only there to be broken. We will clone the genes responsible for yield, domestication, heterosis, epistasis and canalization and explore their molecular action. I expect that the breeding concepts and methods developed through this project will lead the way to increased productivity of crops that are important for global food security.
Summary
Plant breeders are challenged with sustaining global crop improvements. Is there a limit to crop yield? This project will address this central question using processing tomatoes as a model for a mechanized crop. By integrating in a single web-based platform of ‘Phenom Networks’ a broad germplasm base, deep phenotypes, and multiple genome sequences of tomato species, we will identify the genes and mechanisms that dictate crop productivity and implement them in the creation of next generation F1 hybrids. Our work is founded on our years of efforts to establish the following integrated genetic pillars: 1) The tomato genome revealed SNPs for broader marker-assisted selection between cultivated parents and untapped diversity from closely-related red-fruited ancestors. We will develop new elite parental lines into which all discovered traits will be introduced. 2) We will enrich the narrow genetic base of modern processing tomato by pyramiding recessive, additive, dominant and overdominant QTL from six wild species introgression lines (ILs) and field-test them in diverse environments. 3) By producing hybrids with ‘recessive’ deleterious mutants we have identified heterosis genes that increase yield by ~50%; these will be combined with the IL QTL. 4) Finally, we will focus on newly discovered “stability QTL” that significantly improve the reproducibility of yield effects by canalizing this phenotype in spite of environmental perturbations. This multi-faceted integrated tomato breeding effort will unite classical and genomics assisted methods with statistical genetics to demonstrate that yield barriers of the leading commercial hybrids are only there to be broken. We will clone the genes responsible for yield, domestication, heterosis, epistasis and canalization and explore their molecular action. I expect that the breeding concepts and methods developed through this project will lead the way to increased productivity of crops that are important for global food security.
Max ERC Funding
2 500 000 €
Duration
Start date: 2012-01-01, End date: 2016-12-31