Project acronym AEROBIC
Project Assessing the Effects of Rising O2 on Biogeochemical Cycles: Integrated Laboratory Experiments and Numerical Simulations
Researcher (PI) Itay Halevy
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), PE10, ERC-2013-StG
Summary The rise of atmospheric O2 ~2,500 million years ago is one of the most profound transitions in Earth's history. Yet, despite its central role in shaping Earth's surface environment, the cause for the rise of O2 remains poorly understood. Tight coupling between the O2 cycle and the biogeochemical cycles of redox-active elements, such as C, Fe and S, implies radical changes in these cycles before, during and after the rise of O2. These changes, too, are incompletely understood, but have left valuable information encoded in the geological record. This information has been qualitatively interpreted, leaving many aspects of the rise of O2, including its causes and constraints on ocean chemistry before and after it, topics of ongoing research and debate. Here, I outline a research program to address this fundamental question in geochemical Earth systems evolution. The inherently interdisciplinary program uniquely integrates laboratory experiments, numerical models, geological observations, and geochemical analyses. Laboratory experiments and geological observations will constrain unknown parameters of the early biogeochemical cycles, and, in combination with field studies, will validate and refine the use of paleoenvironmental proxies. The insight gained will be used to develop detailed models of the coupled biogeochemical cycles, which will themselves be used to quantitatively understand the events surrounding the rise of O2, and to illuminate the dynamics of elemental cycles in the early oceans.
This program is expected to yield novel, quantitative insight into these important events in Earth history and to have a major impact on our understanding of early ocean chemistry and the rise of O2. An ERC Starting Grant will enable me to use the excellent experimental and computational facilities at my disposal, to access the outstanding human resource at the Weizmann Institute of Science, and to address one of the major open questions in modern geochemistry.
Summary
The rise of atmospheric O2 ~2,500 million years ago is one of the most profound transitions in Earth's history. Yet, despite its central role in shaping Earth's surface environment, the cause for the rise of O2 remains poorly understood. Tight coupling between the O2 cycle and the biogeochemical cycles of redox-active elements, such as C, Fe and S, implies radical changes in these cycles before, during and after the rise of O2. These changes, too, are incompletely understood, but have left valuable information encoded in the geological record. This information has been qualitatively interpreted, leaving many aspects of the rise of O2, including its causes and constraints on ocean chemistry before and after it, topics of ongoing research and debate. Here, I outline a research program to address this fundamental question in geochemical Earth systems evolution. The inherently interdisciplinary program uniquely integrates laboratory experiments, numerical models, geological observations, and geochemical analyses. Laboratory experiments and geological observations will constrain unknown parameters of the early biogeochemical cycles, and, in combination with field studies, will validate and refine the use of paleoenvironmental proxies. The insight gained will be used to develop detailed models of the coupled biogeochemical cycles, which will themselves be used to quantitatively understand the events surrounding the rise of O2, and to illuminate the dynamics of elemental cycles in the early oceans.
This program is expected to yield novel, quantitative insight into these important events in Earth history and to have a major impact on our understanding of early ocean chemistry and the rise of O2. An ERC Starting Grant will enable me to use the excellent experimental and computational facilities at my disposal, to access the outstanding human resource at the Weizmann Institute of Science, and to address one of the major open questions in modern geochemistry.
Max ERC Funding
1 472 690 €
Duration
Start date: 2013-09-01, End date: 2018-08-31
Project acronym Agglomerates
Project Infinite Protein Self-Assembly in Health and Disease
Researcher (PI) Emmanuel Doram LEVY
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS2, ERC-2018-COG
Summary Understanding how proteins respond to mutations is of paramount importance to biology and disease. While protein stability and misfolding have been instrumental in rationalizing the impact of mutations, we recently discovered that an alternative route is also frequent, where mutations at the surface of symmetric proteins trigger novel self-interactions that lead to infinite self-assembly. This mechanism can be involved in disease, as in sickle-cell anemia, but may also serve in adaptation. Importantly, it differs fundamentally from aggregation, because misfolding does not drive it. Thus, we term it “agglomeration”. The ease with which agglomeration can occur, even by single point mutations, shifts the paradigm of how quickly new protein assemblies can emerge, both in health and disease. This prompts us to determine the basic principles of protein agglomeration and explore its implications in cell physiology and human disease.
We propose an interdisciplinary research program bridging atomic and cellular scales to explore agglomeration in three aims: (i) Map the landscape of protein agglomeration in response to mutation in endogenous yeast proteins; (ii) Characterize how yeast physiology impacts agglomeration by changes in gene expression or cell state, and, conversely, how protein agglomerates impact yeast fitness. (iii) Analyze agglomeration in relation to human disease via two approaches. First, by predicting single nucleotide polymorphisms that trigger agglomeration, prioritizing them using knowledge from Aims 1 & 2, and characterizing them experimentally. Second, by providing a proof-of-concept that agglomeration can be exploited in drug design, whereby drugs induce its formation, like mutations can do.
Overall, through this research, we aim to establish agglomeration as a paradigm for protein assembly, with implications for our understanding of evolution, physiology, and disease.
Summary
Understanding how proteins respond to mutations is of paramount importance to biology and disease. While protein stability and misfolding have been instrumental in rationalizing the impact of mutations, we recently discovered that an alternative route is also frequent, where mutations at the surface of symmetric proteins trigger novel self-interactions that lead to infinite self-assembly. This mechanism can be involved in disease, as in sickle-cell anemia, but may also serve in adaptation. Importantly, it differs fundamentally from aggregation, because misfolding does not drive it. Thus, we term it “agglomeration”. The ease with which agglomeration can occur, even by single point mutations, shifts the paradigm of how quickly new protein assemblies can emerge, both in health and disease. This prompts us to determine the basic principles of protein agglomeration and explore its implications in cell physiology and human disease.
We propose an interdisciplinary research program bridging atomic and cellular scales to explore agglomeration in three aims: (i) Map the landscape of protein agglomeration in response to mutation in endogenous yeast proteins; (ii) Characterize how yeast physiology impacts agglomeration by changes in gene expression or cell state, and, conversely, how protein agglomerates impact yeast fitness. (iii) Analyze agglomeration in relation to human disease via two approaches. First, by predicting single nucleotide polymorphisms that trigger agglomeration, prioritizing them using knowledge from Aims 1 & 2, and characterizing them experimentally. Second, by providing a proof-of-concept that agglomeration can be exploited in drug design, whereby drugs induce its formation, like mutations can do.
Overall, through this research, we aim to establish agglomeration as a paradigm for protein assembly, with implications for our understanding of evolution, physiology, and disease.
Max ERC Funding
2 574 819 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym ANTHROPOID
Project Great ape organoids to reconstruct uniquely human development
Researcher (PI) Jarrett CAMP
Host Institution (HI) INSTITUT FUR MOLEKULARE UND KLINISCHE OPHTHALMOLOGIE BASEL
Call Details Starting Grant (StG), LS2, ERC-2018-STG
Summary Humans diverged from our closest living relatives, chimpanzees and other great apes, 6-10 million years ago. Since this divergence, our ancestors acquired genetic changes that enhanced cognition, altered metabolism, and endowed our species with an adaptive capacity to colonize the entire planet and reshape the biosphere. Through genome comparisons between modern humans, Neandertals, chimpanzees and other apes we have identified genetic changes that likely contribute to innovations in human metabolic and cognitive physiology. However, it has been difficult to assess the functional effects of these genetic changes due to the lack of cell culture systems that recapitulate great ape organ complexity. Human and chimpanzee pluripotent stem cells (PSCs) can self-organize into three-dimensional (3D) tissues that recapitulate the morphology, function, and genetic programs controlling organ development. Our vision is to use organoids to study the changes that set modern humans apart from our closest evolutionary relatives as well as all other organisms on the planet. In ANTHROPOID we will generate a great ape developmental cell atlas using cortex, liver, and small intestine organoids. We will use single-cell transcriptomics and chromatin accessibility to identify cell type-specific features of transcriptome divergence at cellular resolution. We will dissect enhancer evolution using single-cell genomic screens and ancestralize human cells to resurrect pre-human cellular phenotypes. ANTHROPOID utilizes quantitative and state-of-the-art methods to explore exciting high-risk questions at multiple branches of the modern human lineage. This project is a ground breaking starting point to replay evolution and tackle the ancient question of what makes us uniquely human?
Summary
Humans diverged from our closest living relatives, chimpanzees and other great apes, 6-10 million years ago. Since this divergence, our ancestors acquired genetic changes that enhanced cognition, altered metabolism, and endowed our species with an adaptive capacity to colonize the entire planet and reshape the biosphere. Through genome comparisons between modern humans, Neandertals, chimpanzees and other apes we have identified genetic changes that likely contribute to innovations in human metabolic and cognitive physiology. However, it has been difficult to assess the functional effects of these genetic changes due to the lack of cell culture systems that recapitulate great ape organ complexity. Human and chimpanzee pluripotent stem cells (PSCs) can self-organize into three-dimensional (3D) tissues that recapitulate the morphology, function, and genetic programs controlling organ development. Our vision is to use organoids to study the changes that set modern humans apart from our closest evolutionary relatives as well as all other organisms on the planet. In ANTHROPOID we will generate a great ape developmental cell atlas using cortex, liver, and small intestine organoids. We will use single-cell transcriptomics and chromatin accessibility to identify cell type-specific features of transcriptome divergence at cellular resolution. We will dissect enhancer evolution using single-cell genomic screens and ancestralize human cells to resurrect pre-human cellular phenotypes. ANTHROPOID utilizes quantitative and state-of-the-art methods to explore exciting high-risk questions at multiple branches of the modern human lineage. This project is a ground breaking starting point to replay evolution and tackle the ancient question of what makes us uniquely human?
Max ERC Funding
1 500 000 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym BEFINE
Project mechanical BEhavior of Fluid-INduced Earthquakes
Researcher (PI) Marie, Estelle, Solange VIOLAY
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Starting Grant (StG), PE10, ERC-2017-STG
Summary Fluids play an important role in fault zone and in earthquakes generation. Fluid pressure reduces the normal effective stress, lowering the frictional strength of the fault, potentially triggering earthquake ruptures. Fluid injection induced earthquakes (FIE) are direct evidence of the effect of fluid pressure on the fault strength. In addition, natural earthquake sequences are often associated with high fluid pressures at seismogenic depths. Although simple in theory, the mechanisms that govern the nucleation, propagation and recurrence of FIEs are poorly constrained, and our ability to assess the seismic hazard that is associated with natural and induced events remains limited. This project aims to enhance our knowledge of FIE mechanisms over entire seismic cycles through multidisciplinary approaches, including the following:
- Set-up and installation of a new and unique rock friction apparatus that is dedicated to the study of FIEs.
- Low strain rate friction experiments (coupled with electrical conductivity measurements) to investigate the influence of fluids on fault creep and earthquake recurrence.
- Intermediate strain rate friction experiments to investigate the effect of fluids on fault stability during earthquake nucleation.
- High strain rate friction experiments to investigate the effect of fluids on fault weakening during earthquake propagation.
- Post-mortem experimental fault analyses with state-of-art microstructural techniques.
- The theoretical friction law will be calibrated with friction experiments and faulted rock microstructural observations.
These steps will produce fundamental discoveries regarding natural earthquakes and tectonic processes and help scientists understand and eventually manage the occurrence of induced seismicity, an increasingly hot topic in geo-engineering. The sustainable exploitation of geo-resources is a key research and technology challenge at the European scale, with a substantial economical and societal impact.
Summary
Fluids play an important role in fault zone and in earthquakes generation. Fluid pressure reduces the normal effective stress, lowering the frictional strength of the fault, potentially triggering earthquake ruptures. Fluid injection induced earthquakes (FIE) are direct evidence of the effect of fluid pressure on the fault strength. In addition, natural earthquake sequences are often associated with high fluid pressures at seismogenic depths. Although simple in theory, the mechanisms that govern the nucleation, propagation and recurrence of FIEs are poorly constrained, and our ability to assess the seismic hazard that is associated with natural and induced events remains limited. This project aims to enhance our knowledge of FIE mechanisms over entire seismic cycles through multidisciplinary approaches, including the following:
- Set-up and installation of a new and unique rock friction apparatus that is dedicated to the study of FIEs.
- Low strain rate friction experiments (coupled with electrical conductivity measurements) to investigate the influence of fluids on fault creep and earthquake recurrence.
- Intermediate strain rate friction experiments to investigate the effect of fluids on fault stability during earthquake nucleation.
- High strain rate friction experiments to investigate the effect of fluids on fault weakening during earthquake propagation.
- Post-mortem experimental fault analyses with state-of-art microstructural techniques.
- The theoretical friction law will be calibrated with friction experiments and faulted rock microstructural observations.
These steps will produce fundamental discoveries regarding natural earthquakes and tectonic processes and help scientists understand and eventually manage the occurrence of induced seismicity, an increasingly hot topic in geo-engineering. The sustainable exploitation of geo-resources is a key research and technology challenge at the European scale, with a substantial economical and societal impact.
Max ERC Funding
1 982 925 €
Duration
Start date: 2018-03-01, End date: 2023-02-28
Project acronym BIOCARB
Project Carbonate Biomineralization in the Marine Environment: Paleo-climate proxies and the origin of vital effects
Researcher (PI) Anders Meibom
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), PE10, ERC-2009-AdG
Summary This interdisciplinary proposal has the objective to greatly enhance our understanding of fundamental biomineralization processes involved in the formation of calcium carbonates by marine organisms, such as corals, foraminifera and bivalves, in order to better understand vital effects. This is essential to the application of these carbonates as proxies for global (paleo-) environmental change. The core of the proposal is an experimental capability that I have pioneered during 2008: Dynamic stable isotopic labeling during formation of carbonate skeletons, tests, and shells, combined with NanoSIMS imaging. The NanoSIMS ion microprobe is a state-of-the-art analytical technology that allows precise elemental and isotopic imaging with a spatial resolution of ~100 nanometers. NanoSIMS imaging of the isotopic label(s) in the resulting biocarbonates and in associated cell-structures will be used to uncover cellular-level transport processes, timescales of formation of different biocarbonate components, as well as trace-elemental and isotopic fractionations. This will uncover the origin of vital effects. With this proposal, I establish a new scientific frontier and guarantee European leadership. The technical and scientific developments resulting from this work are broadly applicable and will radically change scientific ideas about marine carbonate biomineralization and compositional vital effects.
Summary
This interdisciplinary proposal has the objective to greatly enhance our understanding of fundamental biomineralization processes involved in the formation of calcium carbonates by marine organisms, such as corals, foraminifera and bivalves, in order to better understand vital effects. This is essential to the application of these carbonates as proxies for global (paleo-) environmental change. The core of the proposal is an experimental capability that I have pioneered during 2008: Dynamic stable isotopic labeling during formation of carbonate skeletons, tests, and shells, combined with NanoSIMS imaging. The NanoSIMS ion microprobe is a state-of-the-art analytical technology that allows precise elemental and isotopic imaging with a spatial resolution of ~100 nanometers. NanoSIMS imaging of the isotopic label(s) in the resulting biocarbonates and in associated cell-structures will be used to uncover cellular-level transport processes, timescales of formation of different biocarbonate components, as well as trace-elemental and isotopic fractionations. This will uncover the origin of vital effects. With this proposal, I establish a new scientific frontier and guarantee European leadership. The technical and scientific developments resulting from this work are broadly applicable and will radically change scientific ideas about marine carbonate biomineralization and compositional vital effects.
Max ERC Funding
2 182 000 €
Duration
Start date: 2010-07-01, End date: 2015-06-30
Project acronym BiomeRiskFactors
Project Discovering microbiome-based disease risk factors
Researcher (PI) Eran Segal
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS2, ERC-2017-ADG
Summary Identifying risk factors for diseases that can be prevented or delayed by early intervention is of major importance, and numerous genetic, lifestyle, anthropometric and clinical risk factors were found for many different diseases. Another source of potentially pertinent disease risk factors is the human microbiome - the collective genome of trillions of bacteria, viruses, fungi, and parasites that reside in the human gut. However, very few microbiome disease markers were found to date.
Here, we aim to develop risk prediction tools based on the human microbiome that predict the likelihood of an individual to develop a particular condition or disease within 5-10 years. We will use a cohort of >2200 individuals that my group previously assembled, for whom we have clinical profiles, gut microbiome data, and banked blood and stool samples. We will invite people 5-10 years after their initial recruitment time, profile disease status and blood markers, and develop algorithms for predicting 5-10 year onset of Type 2 diabetes, cardiovascular disease, and obesity, using microbiome data from recruitment time.
To increase the likelihood of finding microbiome markers predictive of disease onset, we will develop novel experimental and computational methods for in-depth characterization of microbial gene function, the metabolites produced by the microbiome, the underexplored fungal microbiome members, and the interactions between the gut microbiota and the host adaptive immune system. We will then apply these methods to >2200 banked samples from cohort recruitment time and use the resulting data in devising our microbiome-based risk prediction tools. In themselves, these novel assays and their application to >2200 samples should greatly advance the microbiome field.
If successful, our proposal will identify new disease risk factors and risk prediction tools based on the microbiome, paving the way towards using the microbiome in early disease detection and prevention.
Summary
Identifying risk factors for diseases that can be prevented or delayed by early intervention is of major importance, and numerous genetic, lifestyle, anthropometric and clinical risk factors were found for many different diseases. Another source of potentially pertinent disease risk factors is the human microbiome - the collective genome of trillions of bacteria, viruses, fungi, and parasites that reside in the human gut. However, very few microbiome disease markers were found to date.
Here, we aim to develop risk prediction tools based on the human microbiome that predict the likelihood of an individual to develop a particular condition or disease within 5-10 years. We will use a cohort of >2200 individuals that my group previously assembled, for whom we have clinical profiles, gut microbiome data, and banked blood and stool samples. We will invite people 5-10 years after their initial recruitment time, profile disease status and blood markers, and develop algorithms for predicting 5-10 year onset of Type 2 diabetes, cardiovascular disease, and obesity, using microbiome data from recruitment time.
To increase the likelihood of finding microbiome markers predictive of disease onset, we will develop novel experimental and computational methods for in-depth characterization of microbial gene function, the metabolites produced by the microbiome, the underexplored fungal microbiome members, and the interactions between the gut microbiota and the host adaptive immune system. We will then apply these methods to >2200 banked samples from cohort recruitment time and use the resulting data in devising our microbiome-based risk prediction tools. In themselves, these novel assays and their application to >2200 samples should greatly advance the microbiome field.
If successful, our proposal will identify new disease risk factors and risk prediction tools based on the microbiome, paving the way towards using the microbiome in early disease detection and prevention.
Max ERC Funding
2 500 000 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym BioMeTRe
Project Biophysical mechanisms of long-range transcriptional regulation
Researcher (PI) Luca GIORGETTI
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Call Details Starting Grant (StG), LS2, ERC-2017-STG
Summary In mammals, transcriptional control of many genes relies on cis-regulatory elements such as enhancers, which are often located tens to hundreds of kilobases away from their cognate promoters. Functional interactions between distal regulatory elements and target promoters require mutual physical proximity, which is linked to the three-dimensional structure of the chromatin fiber. Chromosome conformation capture studies revealed that chromosomes are partitioned into Topologically Associating Domains (TADs), sub-megabase domains of preferential physical interactions of the chromatin fiber. Genetic evidence showed that TAD boundaries restrict the genomic range of enhancer-promoter communication, and that interactions between regulatory sequences within TADs are further fine-tuned by smaller-scale structures. However, the mechanistic details of how physical interactions translate into transcriptional outputs are totally unknown. Here we propose to explore the biophysical mechanisms that link chromosome conformation and long-range transcriptional regulation using molecular biology, genetic engineering, single-cell experiments and physical modeling. We will measure chromosomal interactions in single cells and in time using a novel method that relies on an enzymatic process in vivo. Genetic engineering will be used to establish a cell system that allows quantitative measurement of how enhancer-promoter interactions relate to transcription at the population and single-cell levels, and to test the effects of perturbations without confounding effects. Finally, we will develop physical models of promoter operation in the presence of distal enhancers, which will be used to interpret the experimental data and formulate new testable predictions. With this integrated approach we aim at providing an entirely new layer of description of the general principles underlying transcriptional control, which could establish new paradigms for research in epigenetics and gene regulation.
Summary
In mammals, transcriptional control of many genes relies on cis-regulatory elements such as enhancers, which are often located tens to hundreds of kilobases away from their cognate promoters. Functional interactions between distal regulatory elements and target promoters require mutual physical proximity, which is linked to the three-dimensional structure of the chromatin fiber. Chromosome conformation capture studies revealed that chromosomes are partitioned into Topologically Associating Domains (TADs), sub-megabase domains of preferential physical interactions of the chromatin fiber. Genetic evidence showed that TAD boundaries restrict the genomic range of enhancer-promoter communication, and that interactions between regulatory sequences within TADs are further fine-tuned by smaller-scale structures. However, the mechanistic details of how physical interactions translate into transcriptional outputs are totally unknown. Here we propose to explore the biophysical mechanisms that link chromosome conformation and long-range transcriptional regulation using molecular biology, genetic engineering, single-cell experiments and physical modeling. We will measure chromosomal interactions in single cells and in time using a novel method that relies on an enzymatic process in vivo. Genetic engineering will be used to establish a cell system that allows quantitative measurement of how enhancer-promoter interactions relate to transcription at the population and single-cell levels, and to test the effects of perturbations without confounding effects. Finally, we will develop physical models of promoter operation in the presence of distal enhancers, which will be used to interpret the experimental data and formulate new testable predictions. With this integrated approach we aim at providing an entirely new layer of description of the general principles underlying transcriptional control, which could establish new paradigms for research in epigenetics and gene regulation.
Max ERC Funding
1 500 000 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym BLACARAT
Project "Black Carbon in the Atmosphere: Emissions, Aging and Cloud Interactions"
Researcher (PI) Martin Gysel Beer
Host Institution (HI) PAUL SCHERRER INSTITUT
Call Details Consolidator Grant (CoG), PE10, ERC-2013-CoG
Summary "Atmospheric aerosol particles have been shown to impact the earth's climate because they scatter and absorb solar radiation (direct effect) and because they can modify the microphysical properties of clouds by acting as cloud condensation nuclei or ice nuclei (indirect effects). Radiative forcing by anthropogenic aerosols remains poorly quantified, thus leading to considerable uncertainty in our understanding of the earth’s climate response to the radiative forcing by greenhouse gases. Black carbon (BC), mostly emitted by anthropogenic combustion processes and biomass burning, is an important component of atmospheric aerosols. Estimates show that BC may be the second strongest contributor (after CO2) to global warming. Adverse health effects due to particulate air pollution have also been associated with traffic-related BC particles. These climate and health effects brought BC emission reductions into the political focus of possible mitigation strategies with immediate and multiple benefits for human well-being.
Laboratory experiments aim at the physical and chemical characterisation of BC emissions from diesel engines and biomass burning under controlled conditions. A mobile laboratory equipped with state-of-the-art aerosol sensors will be used to determine the contribution of different BC sources to atmospheric BC loadings, and to investigate the evolution of the relevant BC properties with atmospheric aging during transport from sources to remote areas. The interactions of BC particles with clouds as a function of BC properties will be investigated with in-situ measurements by operating quantitative single particle instruments behind a novel sampling inlet, which makes selective sampling of interstitial, cloud droplet residual or ice crystal residual particles possible. Above experimental studies aim at improving our understanding of BC’s atmospheric life cycle and will be used in model simulations for quantitatively assessing the atmospheric impacts of BC."
Summary
"Atmospheric aerosol particles have been shown to impact the earth's climate because they scatter and absorb solar radiation (direct effect) and because they can modify the microphysical properties of clouds by acting as cloud condensation nuclei or ice nuclei (indirect effects). Radiative forcing by anthropogenic aerosols remains poorly quantified, thus leading to considerable uncertainty in our understanding of the earth’s climate response to the radiative forcing by greenhouse gases. Black carbon (BC), mostly emitted by anthropogenic combustion processes and biomass burning, is an important component of atmospheric aerosols. Estimates show that BC may be the second strongest contributor (after CO2) to global warming. Adverse health effects due to particulate air pollution have also been associated with traffic-related BC particles. These climate and health effects brought BC emission reductions into the political focus of possible mitigation strategies with immediate and multiple benefits for human well-being.
Laboratory experiments aim at the physical and chemical characterisation of BC emissions from diesel engines and biomass burning under controlled conditions. A mobile laboratory equipped with state-of-the-art aerosol sensors will be used to determine the contribution of different BC sources to atmospheric BC loadings, and to investigate the evolution of the relevant BC properties with atmospheric aging during transport from sources to remote areas. The interactions of BC particles with clouds as a function of BC properties will be investigated with in-situ measurements by operating quantitative single particle instruments behind a novel sampling inlet, which makes selective sampling of interstitial, cloud droplet residual or ice crystal residual particles possible. Above experimental studies aim at improving our understanding of BC’s atmospheric life cycle and will be used in model simulations for quantitatively assessing the atmospheric impacts of BC."
Max ERC Funding
1 992 015 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym C8
Project Consistent computation of the chemistry-cloud continuum and climate change in Cyprus
Researcher (PI) Johannes Lelieveld
Host Institution (HI) THE CYPRUS RESEARCH AND EDUCATIONAL FOUNDATION
Call Details Advanced Grant (AdG), PE10, ERC-2008-AdG
Summary We have developed a new numerical method to consistently compute atmospheric trace gas and aerosol chemistry and cloud processes. The method is computationally efficient so that it can be used in climate models. For the first time cloud droplet formation on multi-component particles can be represented based on first principles rather than parameterisations. This allows for a direct coupling in models between aerosol chemical composition and the continuum between hazes and clouds as a function of ambient relative humidity. We will apply the method in a new nested global-limited area model system to study atmospheric chemistry climate interactions and anthropogenic influences. We will focus on the Mediterranean region because it is a hot spot in climate change exposed to drying and air pollution. The limited area model will also be applied as cloud-resolving model to study aerosol influences on precipitation and storm development. By simulating realistic meteorological conditions at high spatial resolution our method can be straightforwardly tested against observations. Central questions are: - How does the simulated haze-cloud continuum compare with remote sensing measurements and what is the consequence of abandoning the traditional and artificial distinction between aerosols and clouds? - How are cloud and precipitation formation influenced by atmospheric chemical composition changes? - To what extent do haze and cloud formation in polluted air exert forcings of synoptic meteorological conditions and climate? - Can aerosol pollution in the Mediterranean region exacerbate the predicted and observed drying in a changing climate? The model system is user-friendly and will facilitate air quality and climate studies by regional scientists. The project will be part of the Energy, Environment and Water Centre of the newly founded Cyprus Institute, provide input to climate impact assessments and contribute to a regional outreach programme.
Summary
We have developed a new numerical method to consistently compute atmospheric trace gas and aerosol chemistry and cloud processes. The method is computationally efficient so that it can be used in climate models. For the first time cloud droplet formation on multi-component particles can be represented based on first principles rather than parameterisations. This allows for a direct coupling in models between aerosol chemical composition and the continuum between hazes and clouds as a function of ambient relative humidity. We will apply the method in a new nested global-limited area model system to study atmospheric chemistry climate interactions and anthropogenic influences. We will focus on the Mediterranean region because it is a hot spot in climate change exposed to drying and air pollution. The limited area model will also be applied as cloud-resolving model to study aerosol influences on precipitation and storm development. By simulating realistic meteorological conditions at high spatial resolution our method can be straightforwardly tested against observations. Central questions are: - How does the simulated haze-cloud continuum compare with remote sensing measurements and what is the consequence of abandoning the traditional and artificial distinction between aerosols and clouds? - How are cloud and precipitation formation influenced by atmospheric chemical composition changes? - To what extent do haze and cloud formation in polluted air exert forcings of synoptic meteorological conditions and climate? - Can aerosol pollution in the Mediterranean region exacerbate the predicted and observed drying in a changing climate? The model system is user-friendly and will facilitate air quality and climate studies by regional scientists. The project will be part of the Energy, Environment and Water Centre of the newly founded Cyprus Institute, provide input to climate impact assessments and contribute to a regional outreach programme.
Max ERC Funding
2 196 000 €
Duration
Start date: 2009-01-01, End date: 2014-12-31
Project acronym CancerFluxome
Project Cancer Cellular Metabolism across Space and Time
Researcher (PI) Tomer Shlomi
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS2, ERC-2016-STG
Summary The metabolism of cancer cells is altered to meet cellular requirements for growth, providing novel means to selectively target tumorigenesis. While extensively studied, our current view of cancer cellular metabolism is fundamentally limited by lack of information on variability in metabolic activity between distinct subcellular compartments and cells.
We propose to develop a spatio-temporal fluxomics approach for quantifying metabolic fluxes in the cytoplasm vs. mitochondria as well as their cell-cycle dynamics, combining mass-spectrometry based isotope tracing with cell synchronization, rapid cellular fractionation, and computational metabolic network modelling.
Spatio-temporal fluxomics will be used to revisit and challenge our current understanding of central metabolism and its induced adaptation to oncogenic events – an important endeavour considering that mitochondrial bioenergetics and biosynthesis are required for tumorigenesis and accumulating evidences for metabolic alterations throughout the cell-cycle.
Our preliminary results show intriguing oscillations between oxidative and reductive TCA cycle flux throughout the cell-cycle. We will explore the extent to which cells adapt their metabolism to fulfil the changing energetic and anabolic demands throughout the cell-cycle, how metabolic oscillations are regulated, and their benefit to cells in terms of thermodynamic efficiency. Spatial flux analysis will be instrumental for investigating glutaminolysis - a ‘hallmark’ metabolic adaptation in cancer involving shuttling of metabolic intermediates and cofactors between mitochondria and cytoplasm.
On a clinical front, our spatio-temporal fluxomics analysis will enable to disentangle oncogene-induced flux alterations, having an important tumorigenic role, from artefacts originating from population averaging. A comprehensive view of how cells adapt their metabolism due to oncogenic mutations will reveal novel targets for anti-cancer drugs.
Summary
The metabolism of cancer cells is altered to meet cellular requirements for growth, providing novel means to selectively target tumorigenesis. While extensively studied, our current view of cancer cellular metabolism is fundamentally limited by lack of information on variability in metabolic activity between distinct subcellular compartments and cells.
We propose to develop a spatio-temporal fluxomics approach for quantifying metabolic fluxes in the cytoplasm vs. mitochondria as well as their cell-cycle dynamics, combining mass-spectrometry based isotope tracing with cell synchronization, rapid cellular fractionation, and computational metabolic network modelling.
Spatio-temporal fluxomics will be used to revisit and challenge our current understanding of central metabolism and its induced adaptation to oncogenic events – an important endeavour considering that mitochondrial bioenergetics and biosynthesis are required for tumorigenesis and accumulating evidences for metabolic alterations throughout the cell-cycle.
Our preliminary results show intriguing oscillations between oxidative and reductive TCA cycle flux throughout the cell-cycle. We will explore the extent to which cells adapt their metabolism to fulfil the changing energetic and anabolic demands throughout the cell-cycle, how metabolic oscillations are regulated, and their benefit to cells in terms of thermodynamic efficiency. Spatial flux analysis will be instrumental for investigating glutaminolysis - a ‘hallmark’ metabolic adaptation in cancer involving shuttling of metabolic intermediates and cofactors between mitochondria and cytoplasm.
On a clinical front, our spatio-temporal fluxomics analysis will enable to disentangle oncogene-induced flux alterations, having an important tumorigenic role, from artefacts originating from population averaging. A comprehensive view of how cells adapt their metabolism due to oncogenic mutations will reveal novel targets for anti-cancer drugs.
Max ERC Funding
1 481 250 €
Duration
Start date: 2017-02-01, End date: 2022-01-31
Project acronym CAPRI
Project Clouds and Precipitation Response to Anthropogenic Changes in the Natural Environment
Researcher (PI) Ilan Koren
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), PE10, ERC-2012-StG_20111012
Summary Clouds and precipitation play a crucial role in the Earth's energy balance, global atmospheric circulation and the water cycle. Despite their importance, clouds still pose the largest uncertainty in climate research.
I propose a new approach for studying anthropogenic effects on cloud fields and rain, tackling the challenge from both scientific ends: reductionism and systems approach. We will develop a novel research approach using observations and models interactively that will allow us to “peel apart” detailed physical processes. In parallel we will develop a systems view of cloud fields looking for Emergent Behavior rising out of the complexity, as the end result of all of the coupled processes. Better understanding of key processes on a detailed (reductionist) manner will enable us to formulate the important basic rules that control the field and to look for emergence of the overall effects.
We will merge ideas and methods from four different disciplines: remote sensing and radiative transfer, cloud physics, pattern recognition and computer vision and ideas developed in systems approach. All of this will be done against the backdrop of natural variability of meteorological systems.
The outcomes of this work will include fundamental new understanding of the coupled surface-aerosol-cloud-precipitation system. More importantly this work will emphasize the consequences of human actions on the environment, and how we change our climate and hydrological cycle as we input pollutants and transform the Earth’s surface. This work will open new horizons in cloud research by developing novel methods and employing the bulk knowledge of pattern recognition, complexity, networking and self organization to cloud and climate studies. We are proposing a long-term, open-ended program of study that will have scientific and societal relevance as long as human-caused influences continue, evolve and change.
Summary
Clouds and precipitation play a crucial role in the Earth's energy balance, global atmospheric circulation and the water cycle. Despite their importance, clouds still pose the largest uncertainty in climate research.
I propose a new approach for studying anthropogenic effects on cloud fields and rain, tackling the challenge from both scientific ends: reductionism and systems approach. We will develop a novel research approach using observations and models interactively that will allow us to “peel apart” detailed physical processes. In parallel we will develop a systems view of cloud fields looking for Emergent Behavior rising out of the complexity, as the end result of all of the coupled processes. Better understanding of key processes on a detailed (reductionist) manner will enable us to formulate the important basic rules that control the field and to look for emergence of the overall effects.
We will merge ideas and methods from four different disciplines: remote sensing and radiative transfer, cloud physics, pattern recognition and computer vision and ideas developed in systems approach. All of this will be done against the backdrop of natural variability of meteorological systems.
The outcomes of this work will include fundamental new understanding of the coupled surface-aerosol-cloud-precipitation system. More importantly this work will emphasize the consequences of human actions on the environment, and how we change our climate and hydrological cycle as we input pollutants and transform the Earth’s surface. This work will open new horizons in cloud research by developing novel methods and employing the bulk knowledge of pattern recognition, complexity, networking and self organization to cloud and climate studies. We are proposing a long-term, open-ended program of study that will have scientific and societal relevance as long as human-caused influences continue, evolve and change.
Max ERC Funding
1 428 169 €
Duration
Start date: 2012-09-01, End date: 2017-08-31
Project acronym CASSANDRA
Project Accelerating mass loss of Greenland: firn and the shifting runoff limit
Researcher (PI) Horst MACHGUTH
Host Institution (HI) UNIVERSITE DE FRIBOURG
Call Details Consolidator Grant (CoG), PE10, ERC-2018-COG
Summary Meltwater running off the flanks of the Greenland ice sheet contributes roughly 60% to its mass loss, the rest being due to calving. Only meltwater originating from below the elevation of the runoff limit leaves the ice sheet, contributing to mass loss; melt at higher elevations refreezes in the porous firn and does not drive mass loss. Therefore any shift in the runoff limit modifies mass loss and subsequent sea level rise. New evidence shows surface runoff at increasingly high elevations, outpacing the rate at which the equilibrium line elevation rises. This research proposal focuses on the runoff limit as a powerful yet poorly understood modulator of Greenland mass balance. We will track the runoff limit over the full satellite era using two of the largest and oldest remote sensing archives, Landsat and the Advanced Very High Resolution Radiometer (AVHRR). We will establish time series of the runoff limit for all regions of Greenland to identify the mechanisms driving fluctuations in the runoff limit. This newly gained process understanding and a wealth of in-situ measurements will then be used to build firn hydrology models capable of simulating runoff and the associated runoff limit over time. Eventually, the firn hydrology models will be applied to reconcile estimates of Greenland past, present and future mass balance. Covering the entire satellite era and all of Greenland, the focus on the runoff limit will constitute a paradigm shift leading to major advance in our understanding of how vulnerable the surface of the ice sheet reacts to climate change and how the changing surface impacts runoff and thus Greenland's role in the global sea level budget.
Summary
Meltwater running off the flanks of the Greenland ice sheet contributes roughly 60% to its mass loss, the rest being due to calving. Only meltwater originating from below the elevation of the runoff limit leaves the ice sheet, contributing to mass loss; melt at higher elevations refreezes in the porous firn and does not drive mass loss. Therefore any shift in the runoff limit modifies mass loss and subsequent sea level rise. New evidence shows surface runoff at increasingly high elevations, outpacing the rate at which the equilibrium line elevation rises. This research proposal focuses on the runoff limit as a powerful yet poorly understood modulator of Greenland mass balance. We will track the runoff limit over the full satellite era using two of the largest and oldest remote sensing archives, Landsat and the Advanced Very High Resolution Radiometer (AVHRR). We will establish time series of the runoff limit for all regions of Greenland to identify the mechanisms driving fluctuations in the runoff limit. This newly gained process understanding and a wealth of in-situ measurements will then be used to build firn hydrology models capable of simulating runoff and the associated runoff limit over time. Eventually, the firn hydrology models will be applied to reconcile estimates of Greenland past, present and future mass balance. Covering the entire satellite era and all of Greenland, the focus on the runoff limit will constitute a paradigm shift leading to major advance in our understanding of how vulnerable the surface of the ice sheet reacts to climate change and how the changing surface impacts runoff and thus Greenland's role in the global sea level budget.
Max ERC Funding
1 989 181 €
Duration
Start date: 2019-05-01, End date: 2024-04-30
Project acronym CHROMATINMODWEB
Project Functional and regulatory protein networks of chromatin modifying enzymes
Researcher (PI) Antonis Kirmizis
Host Institution (HI) UNIVERSITY OF CYPRUS
Call Details Starting Grant (StG), LS2, ERC-2010-StG_20091118
Summary Proper and controlled expression of genes is essential for normal cell growth. Chromatin modifying enzymes play a
fundamental role in the control of gene expression and their deregulation is often linked to cancer. In recent years chromatin
modifiers have been considered key targets for cancer therapy and this demands a full understanding of their biological
functions. Previous biochemical and structural studies have focused on the identification of chromatin modifying enzymes
and characterization of their substrate specificities and catalytic mechanisms. However, a comprehensive view of the
biological processes, signaling pathways and regulatory circuits in which these enzymes participate is missing. Protein
arginine methyltransferases (PRMTs), which methylate histones and are evolutionarily conserved from yeast to human,
constitute an example of chromatin modifying enzymes whose functional and regulatory networks remain unexplored. I
propose to use complementary state-of-the-art genomic and proteomic approaches in order to identify the protein networks
and cellular pathways that are linked to PRMTs. In parallel, I will identify novel regulatory circuits and define the molecular
mechanisms that control methylation of specific histone arginine residues. I will utilize the yeast S. cerevisiae as a model
organism because it allows genetic, biochemical and genomic approaches to be combined. Most importantly, many of the
pathways and mechanisms in yeast are highly conserved and therefore, the findings from this study will be pertinent to
human and other eukaryotic organisms. Establishing a global cellular wiring diagram of PRMTs will serve as a paradigm for
other chromatin modifiers and is imperative for assessing the efficacy of these enzymes as therapeutic targets.
Summary
Proper and controlled expression of genes is essential for normal cell growth. Chromatin modifying enzymes play a
fundamental role in the control of gene expression and their deregulation is often linked to cancer. In recent years chromatin
modifiers have been considered key targets for cancer therapy and this demands a full understanding of their biological
functions. Previous biochemical and structural studies have focused on the identification of chromatin modifying enzymes
and characterization of their substrate specificities and catalytic mechanisms. However, a comprehensive view of the
biological processes, signaling pathways and regulatory circuits in which these enzymes participate is missing. Protein
arginine methyltransferases (PRMTs), which methylate histones and are evolutionarily conserved from yeast to human,
constitute an example of chromatin modifying enzymes whose functional and regulatory networks remain unexplored. I
propose to use complementary state-of-the-art genomic and proteomic approaches in order to identify the protein networks
and cellular pathways that are linked to PRMTs. In parallel, I will identify novel regulatory circuits and define the molecular
mechanisms that control methylation of specific histone arginine residues. I will utilize the yeast S. cerevisiae as a model
organism because it allows genetic, biochemical and genomic approaches to be combined. Most importantly, many of the
pathways and mechanisms in yeast are highly conserved and therefore, the findings from this study will be pertinent to
human and other eukaryotic organisms. Establishing a global cellular wiring diagram of PRMTs will serve as a paradigm for
other chromatin modifiers and is imperative for assessing the efficacy of these enzymes as therapeutic targets.
Max ERC Funding
1 498 279 €
Duration
Start date: 2011-01-01, End date: 2016-06-30
Project acronym CHROMATINSYS
Project Systematic Approach to Dissect the Interplay between Chromatin and Transcription
Researcher (PI) Nir Friedman
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS2, ERC-2013-ADG
Summary Epigenetic mechanisms play an important role in regulating and maintaining the functionality of cells and have been implicated in a wide range of human diseases. Histone proteins that form the protein core of nucleosomes are subject to a bewildering array of covalent and structural modifications, which can repress, permit, or promote transcription. These modifications can be added and removed by specialized complexes that are recruited by other covalent modifications, by transcription factors, or by the transcriptional machinery. Advances in genomics led to comprehensive mapping of the ``epigenome'' in a range of tissues and organisms. These maps established the tight connection between histone modifications and transcription programs. These static charts, however, are less successful at uncovering the underlying mechanisms, logic, and function of histone modifications in establishing and maintaining transcriptional programs. Our premise is that we can answer these basic questions by observing the effect of genetic perturbations on the dynamics of both chromatin state and transcriptional activity. We aim to dissect the chromatin-transcription system in a systematic manner by building on our extensive experience in modeling and analysis, and a unique high-throughput experimental system we established in my lab.
We plan to use the budding yeast model organism, which allows for
efficient genetic and experimental manipulations. We will combine two technologies: (1) high-throughput measurements of single-cell
transcriptional output using fluorescence reporters; and (2) high-throughput immunoprecipitation sequencing assays to map chromatin state. Measuring with these the dynamics of response to stimuli under different genetic backgrounds and using advanced stochastic network models, we will chart detailed mechanisms that are opaque to current approaches and elucidate the general principles that govern the interplay between chromatin and transcription.
Summary
Epigenetic mechanisms play an important role in regulating and maintaining the functionality of cells and have been implicated in a wide range of human diseases. Histone proteins that form the protein core of nucleosomes are subject to a bewildering array of covalent and structural modifications, which can repress, permit, or promote transcription. These modifications can be added and removed by specialized complexes that are recruited by other covalent modifications, by transcription factors, or by the transcriptional machinery. Advances in genomics led to comprehensive mapping of the ``epigenome'' in a range of tissues and organisms. These maps established the tight connection between histone modifications and transcription programs. These static charts, however, are less successful at uncovering the underlying mechanisms, logic, and function of histone modifications in establishing and maintaining transcriptional programs. Our premise is that we can answer these basic questions by observing the effect of genetic perturbations on the dynamics of both chromatin state and transcriptional activity. We aim to dissect the chromatin-transcription system in a systematic manner by building on our extensive experience in modeling and analysis, and a unique high-throughput experimental system we established in my lab.
We plan to use the budding yeast model organism, which allows for
efficient genetic and experimental manipulations. We will combine two technologies: (1) high-throughput measurements of single-cell
transcriptional output using fluorescence reporters; and (2) high-throughput immunoprecipitation sequencing assays to map chromatin state. Measuring with these the dynamics of response to stimuli under different genetic backgrounds and using advanced stochastic network models, we will chart detailed mechanisms that are opaque to current approaches and elucidate the general principles that govern the interplay between chromatin and transcription.
Max ERC Funding
2 396 450 €
Duration
Start date: 2014-01-01, End date: 2018-12-31
Project acronym CNIDARIAMICRORNA
Project Elucidation of the evolution of post-transcriptional regulation by characterizing the cnidarian microRNA pathway
Researcher (PI) Yehu Moran
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS2, ERC-2014-STG
Summary Over the past decade small RNAs such as microRNAs (miRNAs) and small interfering RNAs (siRNAs) have been shown to carry pivotal roles in post-transcriptional regulation and genome protection and to play an important part in various physiological processes in animals. miRNAs can be found in a very wide range of animals yet their functions were studied almost exclusively in members of the Bilateria such as insects, nematodes and vertebrates. Hence studying their function in representatives of non-bilaterian phyla such as Cnidaria (sea anemones, corals, hydras and jellyfish) is crucial for understanding the evolution of miRNAs in animals and can provide important insights into their roles in the ancient ancestor of Cnidaria and Bilateria. The sea anemone Nematostella vectensis is an excellent model for such a study since it can be grown in large numbers throughout its life cycle in the lab and because well-established genetic manipulation techniques are available for this species. Our preliminary results indicate that miRNAs in Nematostella frequently have a nearly perfect match to their messenger RNA (mRNA) targets, resulting in cleavage of the target. This mode of action is common for plant miRNAs, but is very rare in Bilateria. This finding together with my recent discovery of a Nematostella homolog of HYL1, a protein involved in miRNA biogenesis in plants, raises the exciting possibility that the miRNA pathway existed in the common ancestor of plants and animals. Here I suggest to bring together an array of advanced biochemical and genetic methods such as gene knockdown, transgenesis, high throughput sequencing and immunoprecipitation in order to obtain - for the first time - a deep understanding of the biogenesis and mechanism of action of small RNAs in Cnidaria. This will provide a novel way to understand the evolution of this important molecular pathway and to evaluate its age and ancestral form.
Summary
Over the past decade small RNAs such as microRNAs (miRNAs) and small interfering RNAs (siRNAs) have been shown to carry pivotal roles in post-transcriptional regulation and genome protection and to play an important part in various physiological processes in animals. miRNAs can be found in a very wide range of animals yet their functions were studied almost exclusively in members of the Bilateria such as insects, nematodes and vertebrates. Hence studying their function in representatives of non-bilaterian phyla such as Cnidaria (sea anemones, corals, hydras and jellyfish) is crucial for understanding the evolution of miRNAs in animals and can provide important insights into their roles in the ancient ancestor of Cnidaria and Bilateria. The sea anemone Nematostella vectensis is an excellent model for such a study since it can be grown in large numbers throughout its life cycle in the lab and because well-established genetic manipulation techniques are available for this species. Our preliminary results indicate that miRNAs in Nematostella frequently have a nearly perfect match to their messenger RNA (mRNA) targets, resulting in cleavage of the target. This mode of action is common for plant miRNAs, but is very rare in Bilateria. This finding together with my recent discovery of a Nematostella homolog of HYL1, a protein involved in miRNA biogenesis in plants, raises the exciting possibility that the miRNA pathway existed in the common ancestor of plants and animals. Here I suggest to bring together an array of advanced biochemical and genetic methods such as gene knockdown, transgenesis, high throughput sequencing and immunoprecipitation in order to obtain - for the first time - a deep understanding of the biogenesis and mechanism of action of small RNAs in Cnidaria. This will provide a novel way to understand the evolution of this important molecular pathway and to evaluate its age and ancestral form.
Max ERC Funding
1 499 587 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym COSIWAX
Project Compound Specific Hydrogen Isotope Analyses of Leaf Wax n-Alkanes as a Novel Tool to Assess Plant and Ecosystem Water Relations Across new Spatial and Temporal Scales
Researcher (PI) Ansgar Kahmen
Host Institution (HI) UNIVERSITAT BASEL
Call Details Starting Grant (StG), PE10, ERC-2011-StG_20101014
Summary "Leaf wax n-alkanes are long-chained lipids that are vital components of plant cuticles. What makes leaf wax n-alkanes unique is that their stable hydrogen isotope composition (δD) contains information on precipitation and plant water relations. In addition, leaf wax n-alkanes are abundant in leaves, soils, sediments and even the atmosphere and can persist with their δD values over millions of years. With this exceptional combination of properties, leaf wax n-alkanes and their δD values are now being celebrated as the much-needed ecohydrological proxy that provides information on the hydrological cycle and plant water relations across spatial and temporal scales that range from leaves to biomes and from weeks to millions of years. Despite the enormous potential that leaf wax n-alkanes have as ecohydrological proxy for a range of different research areas, the exact type of hydrological information that is recorded in the δD values of leaf wax n-alkanes remains still unclear. This is because critical mechanisms that determine the δD values of leaf wax n-alkanes are not understood. This proposal will perform the experimental work that is now needed to resolve the key mechanisms that determine the δD values leaf wax n-alkanes. These experiments will set the basis to develop a new numerical model that will allow to ultimately test what the exact hydrological signal is that leaf wax n-alkanes record in their δD values: a mere hydrological signal reflecting the amount or origin of precipitation or, a plant-shaped signal indicating plant water relations such as evapotranspiration. Building on this new model, COSIWAX will set out to test the potential that leaf wax n-alkane δD values hold as new ecohydrological proxy for ecology and ecosystem sciences. If successful, COSIWAX will establish with this research leaf wax n-alkanes δD values as a new and innovative ecohydrological proxy that has extensive possible applications in paleoclimatology, ecology, earth system sciences."
Summary
"Leaf wax n-alkanes are long-chained lipids that are vital components of plant cuticles. What makes leaf wax n-alkanes unique is that their stable hydrogen isotope composition (δD) contains information on precipitation and plant water relations. In addition, leaf wax n-alkanes are abundant in leaves, soils, sediments and even the atmosphere and can persist with their δD values over millions of years. With this exceptional combination of properties, leaf wax n-alkanes and their δD values are now being celebrated as the much-needed ecohydrological proxy that provides information on the hydrological cycle and plant water relations across spatial and temporal scales that range from leaves to biomes and from weeks to millions of years. Despite the enormous potential that leaf wax n-alkanes have as ecohydrological proxy for a range of different research areas, the exact type of hydrological information that is recorded in the δD values of leaf wax n-alkanes remains still unclear. This is because critical mechanisms that determine the δD values of leaf wax n-alkanes are not understood. This proposal will perform the experimental work that is now needed to resolve the key mechanisms that determine the δD values leaf wax n-alkanes. These experiments will set the basis to develop a new numerical model that will allow to ultimately test what the exact hydrological signal is that leaf wax n-alkanes record in their δD values: a mere hydrological signal reflecting the amount or origin of precipitation or, a plant-shaped signal indicating plant water relations such as evapotranspiration. Building on this new model, COSIWAX will set out to test the potential that leaf wax n-alkane δD values hold as new ecohydrological proxy for ecology and ecosystem sciences. If successful, COSIWAX will establish with this research leaf wax n-alkanes δD values as a new and innovative ecohydrological proxy that has extensive possible applications in paleoclimatology, ecology, earth system sciences."
Max ERC Funding
1 496 342 €
Duration
Start date: 2011-10-01, End date: 2016-09-30
Project acronym CrackEpitranscriptom
Project Cracking the epitranscriptome
Researcher (PI) Schraga SCHWARTZ
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2016-STG
Summary Over 100 types of distinct modifications are catalyzed on RNA molecules post-transcriptionally. In an analogous manner to well-studied chemical modifications on proteins or DNA, modifications on RNA - and particularly on mRNA - harbor the exciting potential of regulating the complex and interlinked life cycle of these molecules. The most abundant modification in mammalian and yeast mRNA is N6-methyladenosine (m6A). We have pioneered approaches for mapping m6A in a transcriptome wide manner, and we and others have identified factors involved in encoding and decoding m6A. While experimental disruption of these factors is associated with severe phenotypes, the role of m6A remains enigmatic. No single methylated site has been shown to causally underlie any physiological or molecular function. This proposal aims to establish a framework for systematically deciphering the molecular function of a modification and its underlying mechanisms and to uncover the physiological role of the modification in regulation of a cellular response. We will apply this framework to m6A in the context of meiosis in budding yeast, as m6A dynamically accumulates on meiotic mRNAs and as the methyltransferase catalyzing m6A is essential for meiosis. We will (1) aim to elucidate the physiological targets of methylation governing entry into meiosis (2) seek to elucidate the function of m6A at the molecular level, and understand its impact on the various steps of the mRNA life cycle, (3) seek to understand the mechanisms underlying its effects. These aims will provide a comprehensive framework for understanding how the epitranscriptome, an emerging post-transcriptional layer of regulation, fine-tunes gene regulation and impacts cellular decision making in a dynamic response, and will set the stage towards dissecting the roles of m6A and of an expanding set of mRNA modifications in more complex and disease related systems.
Summary
Over 100 types of distinct modifications are catalyzed on RNA molecules post-transcriptionally. In an analogous manner to well-studied chemical modifications on proteins or DNA, modifications on RNA - and particularly on mRNA - harbor the exciting potential of regulating the complex and interlinked life cycle of these molecules. The most abundant modification in mammalian and yeast mRNA is N6-methyladenosine (m6A). We have pioneered approaches for mapping m6A in a transcriptome wide manner, and we and others have identified factors involved in encoding and decoding m6A. While experimental disruption of these factors is associated with severe phenotypes, the role of m6A remains enigmatic. No single methylated site has been shown to causally underlie any physiological or molecular function. This proposal aims to establish a framework for systematically deciphering the molecular function of a modification and its underlying mechanisms and to uncover the physiological role of the modification in regulation of a cellular response. We will apply this framework to m6A in the context of meiosis in budding yeast, as m6A dynamically accumulates on meiotic mRNAs and as the methyltransferase catalyzing m6A is essential for meiosis. We will (1) aim to elucidate the physiological targets of methylation governing entry into meiosis (2) seek to elucidate the function of m6A at the molecular level, and understand its impact on the various steps of the mRNA life cycle, (3) seek to understand the mechanisms underlying its effects. These aims will provide a comprehensive framework for understanding how the epitranscriptome, an emerging post-transcriptional layer of regulation, fine-tunes gene regulation and impacts cellular decision making in a dynamic response, and will set the stage towards dissecting the roles of m6A and of an expanding set of mRNA modifications in more complex and disease related systems.
Max ERC Funding
1 402 666 €
Duration
Start date: 2016-11-01, End date: 2021-10-31
Project acronym CRISPRsition
Project Developing CRISPR adaptation platforms for basic and applied research
Researcher (PI) Ehud Itzhak Qimron
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Consolidator Grant (CoG), LS2, ERC-2018-COG
Summary The CRISPR-Cas system has been extensively studied for its ability to cleave DNA. In contrast, studies of the ability of the system to acquire and integrate new DNA from invaders as a form of prokaryotic adaptive immunity, have lagged behind. This delay reflects the extreme enthusiasm surrounding the potential of using the system’s cleavage capabilities as a genome editing tool. However, the enormous potential of the adaptation process can and should arouse a similar degree of enthusiasm. My lab has pioneered studies on the CRISPR adaptation process by establishing new methodologies, and applying them to demonstrate the essential role of the proteins and DNA elements, as well as the molecular mechanisms, operating in this process. In this project, I will establish novel platforms for studying adaptation and develop them into biotechnological applications and research tools. These tools will allow me to identify the first natural and synthetic inhibitors of the adaptation process. This, in turn, will provide genetic tools to control adaptation, as well as advance the understanding of the arms race between bacteria and their invaders. I will also harness the adaptation process as a platform for diversifying genetic elements for phage display, and for extending phage recognition of a wide range of hosts. Lastly, I will provide the first evidence for an association between the CRISPR adaptation system and gene repression. This linkage will form the basis of a molecular scanner and recorder platform that I will develop and that can be used to identify crucial genetic elements in phage genomes as well as novel regulatory circuits in the bacterial genome. Together, my findings will represent a considerable leap in the understanding of CRISPR adaptation with respect to the process, potential applications, and the intriguing evolutionary significance.
Summary
The CRISPR-Cas system has been extensively studied for its ability to cleave DNA. In contrast, studies of the ability of the system to acquire and integrate new DNA from invaders as a form of prokaryotic adaptive immunity, have lagged behind. This delay reflects the extreme enthusiasm surrounding the potential of using the system’s cleavage capabilities as a genome editing tool. However, the enormous potential of the adaptation process can and should arouse a similar degree of enthusiasm. My lab has pioneered studies on the CRISPR adaptation process by establishing new methodologies, and applying them to demonstrate the essential role of the proteins and DNA elements, as well as the molecular mechanisms, operating in this process. In this project, I will establish novel platforms for studying adaptation and develop them into biotechnological applications and research tools. These tools will allow me to identify the first natural and synthetic inhibitors of the adaptation process. This, in turn, will provide genetic tools to control adaptation, as well as advance the understanding of the arms race between bacteria and their invaders. I will also harness the adaptation process as a platform for diversifying genetic elements for phage display, and for extending phage recognition of a wide range of hosts. Lastly, I will provide the first evidence for an association between the CRISPR adaptation system and gene repression. This linkage will form the basis of a molecular scanner and recorder platform that I will develop and that can be used to identify crucial genetic elements in phage genomes as well as novel regulatory circuits in the bacterial genome. Together, my findings will represent a considerable leap in the understanding of CRISPR adaptation with respect to the process, potential applications, and the intriguing evolutionary significance.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-12-01, End date: 2024-11-30
Project acronym CSEM
Project The Collaborative Seismic Earth Model Project
Researcher (PI) Andreas FICHTNER
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Starting Grant (StG), PE10, ERC-2016-STG
Summary Seismic tomography images of the Earth's interior are key to the characterisation of earthquakes, natural resource exploration, seismic risk assessment, tsunami warning, and studies of geodynamic processes. While tomography has drawn a fascinating picture of our planet, today's individual researchers can exploit only a fraction of the rapidly expanding seismic data volume. Applications relying on tomographic images lag behind their potential; fundamental questions remain unanswered: Do mantle plumes exist in the deep Earth? What are the properties of active faults, and how do they affect earthquake ground motion?
To address these questions and to ensure continued progress of seismic tomography in the 'Big Data' era, I propose new technological developments that enable a paradigm shift in Earth model construction towards a Collaborative Seismic Earth Model (CSEM). Fully accounting for the physics of wave propagation in the complex 3D Earth, the CSEM is envisioned to evolve successively through a systematic group effort of my team, thus going beyond the tomographic models that individual researchers may construct today.
I will develop the technological foundation of the CSEM and integrate these developments in studies of large-earthquake rupture processes and the convective pattern of the Earth's mantle in relation to surface geology. The CSEM project will bridge the gap between regional and global tomography, and deliver the first multiscale model of the Earth where crust and mantle are jointly resolved. The CSEM will lead to a dramatic increase in the exploitable seismic data volume, and set new standards for the construction and reproducibility of tomographic Earth models.
Beyond this project, the CSEM will be openly accessible through the European Plate Observing System (EPOS). It will then offer Earth scientists the unique opportunity to join forces in the discovery of multiscale Earth structure by systematically building on each other's results.
Summary
Seismic tomography images of the Earth's interior are key to the characterisation of earthquakes, natural resource exploration, seismic risk assessment, tsunami warning, and studies of geodynamic processes. While tomography has drawn a fascinating picture of our planet, today's individual researchers can exploit only a fraction of the rapidly expanding seismic data volume. Applications relying on tomographic images lag behind their potential; fundamental questions remain unanswered: Do mantle plumes exist in the deep Earth? What are the properties of active faults, and how do they affect earthquake ground motion?
To address these questions and to ensure continued progress of seismic tomography in the 'Big Data' era, I propose new technological developments that enable a paradigm shift in Earth model construction towards a Collaborative Seismic Earth Model (CSEM). Fully accounting for the physics of wave propagation in the complex 3D Earth, the CSEM is envisioned to evolve successively through a systematic group effort of my team, thus going beyond the tomographic models that individual researchers may construct today.
I will develop the technological foundation of the CSEM and integrate these developments in studies of large-earthquake rupture processes and the convective pattern of the Earth's mantle in relation to surface geology. The CSEM project will bridge the gap between regional and global tomography, and deliver the first multiscale model of the Earth where crust and mantle are jointly resolved. The CSEM will lead to a dramatic increase in the exploitable seismic data volume, and set new standards for the construction and reproducibility of tomographic Earth models.
Beyond this project, the CSEM will be openly accessible through the European Plate Observing System (EPOS). It will then offer Earth scientists the unique opportunity to join forces in the discovery of multiscale Earth structure by systematically building on each other's results.
Max ERC Funding
1 367 500 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym deepSLice
Project Deciphering the greenhouse gas record in deepest ice using continuous sublimation extraction / laser spectrometry
Researcher (PI) Hubertus Fischer
Host Institution (HI) UNIVERSITAET BERN
Call Details Advanced Grant (AdG), PE10, ERC-2014-ADG
Summary The recent anthropogenic global warming makes a detailed understanding of coupling processes between climate and biogeochemical cycles of pressing importance. The atmospheric archive of air bubbles enclosed in polar ice cores provides the only direct record of greenhouse gas changes in the past, and the key to understanding the related changes in biogeochemical cycles and climate/greenhouse gas feedbacks.
Crucial questions about greenhouse gas variability on very short (decadal) and very long (orbital) time scales still remain open. To answer these questions, the ice core community has proposed new drilling projects with the goal of nearly doubling the time span of the available ice core record to the last 1.5 million years and of covering the entire Holocene greenhouse gas record in unprecedented decadal resolution. These goals have one thing in common: due to glacier flow most of this record will only be found in a very thin layer in the bottom-most ice of the cores. Completely new analytical approaches are needed to unlock the atmospheric archive in this ice in order to gain high-resolution, high-precision measurements, while at the same time drastically reducing sample consumption compared to established techniques.
The deepSLice project will make such a step change in ice core analytics by developing a novel coupled Continuous Sublimation Extraction-Quantum Cascade Laser Spectrometer system. It will allow us to simultaneously measure CO2, CH4 and N2O concentrations as well as the isotopic composition of CO2 on air samples of only 1-2 ml at standard pressure and temperature, reducing the required sample size by one order of magnitude. This non-destructive analysis will make it also possible for the complete air sample to be recollected after analysis and used for other measurements. This method will be applied to existing and new ice cores in order to study past changes in greenhouse gases and the underlying biogeochemical cycles in unparalleled detail.
Summary
The recent anthropogenic global warming makes a detailed understanding of coupling processes between climate and biogeochemical cycles of pressing importance. The atmospheric archive of air bubbles enclosed in polar ice cores provides the only direct record of greenhouse gas changes in the past, and the key to understanding the related changes in biogeochemical cycles and climate/greenhouse gas feedbacks.
Crucial questions about greenhouse gas variability on very short (decadal) and very long (orbital) time scales still remain open. To answer these questions, the ice core community has proposed new drilling projects with the goal of nearly doubling the time span of the available ice core record to the last 1.5 million years and of covering the entire Holocene greenhouse gas record in unprecedented decadal resolution. These goals have one thing in common: due to glacier flow most of this record will only be found in a very thin layer in the bottom-most ice of the cores. Completely new analytical approaches are needed to unlock the atmospheric archive in this ice in order to gain high-resolution, high-precision measurements, while at the same time drastically reducing sample consumption compared to established techniques.
The deepSLice project will make such a step change in ice core analytics by developing a novel coupled Continuous Sublimation Extraction-Quantum Cascade Laser Spectrometer system. It will allow us to simultaneously measure CO2, CH4 and N2O concentrations as well as the isotopic composition of CO2 on air samples of only 1-2 ml at standard pressure and temperature, reducing the required sample size by one order of magnitude. This non-destructive analysis will make it also possible for the complete air sample to be recollected after analysis and used for other measurements. This method will be applied to existing and new ice cores in order to study past changes in greenhouse gases and the underlying biogeochemical cycles in unparalleled detail.
Max ERC Funding
2 255 788 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
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 DMR-CODE
Project Decoding the Mammalian transcriptional Regulatory code in development and stimulatory responses
Researcher (PI) Ido Amit
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2012-StG_20111109
Summary Transcription factors (TF) regulate genome function by controlling gene expression. Comprehensive characterization of the in vivo binding of TF to the DNA in relevant primary models is a critical step towards a global understanding of the human genome. Recent advances in high-throughput genomic technologies provide an extraordinary opportunity to develop and apply systematic approaches to learn the underline principles and mechanisms of mammalian transcriptional networks. The premise of this proposal is that a tractable set of rules govern how cells commit to a specific cell type or respond to the environment, and that these rules are coded in regulatory elements in the genome. Currently our understanding of the mammalian regulatory code is hampered by the difficulty of directly measuring in vivo binding of large numbers of TFs to DNA across multiple primary cell types and their natural response to physiological stimuli.
Here, we overcome this bottleneck by systematically exploring the genomic binding network of 1. All relevant TFs of key hematopoietic cells in both steady state and under relevant stimuli. 2. Follow the changes in TF networks as cells differentiate 3. Use these models to engineer cell states and responses. To achieve these goals, we developed a new method for automated high throughput ChIP coupled to sequencing (HT-ChIP-Seq). We used this method to measure binding of 40 TFs in 4 time points following stimulation of dendritic cells with pathogen components. We find that TFs vary substantially in their binding dynamics, genomic localization, number of binding events, and degree of interaction with other TFs. The analysis of this data suggests that the TF network is hierarchically organized, and composed of different types of TFs, cell differentiation factors, factors that prime for gene induction, and factors that bind more specifically and dynamically. This proposal revisits and challenges the current understanding of the mammalian regulatory code.
Summary
Transcription factors (TF) regulate genome function by controlling gene expression. Comprehensive characterization of the in vivo binding of TF to the DNA in relevant primary models is a critical step towards a global understanding of the human genome. Recent advances in high-throughput genomic technologies provide an extraordinary opportunity to develop and apply systematic approaches to learn the underline principles and mechanisms of mammalian transcriptional networks. The premise of this proposal is that a tractable set of rules govern how cells commit to a specific cell type or respond to the environment, and that these rules are coded in regulatory elements in the genome. Currently our understanding of the mammalian regulatory code is hampered by the difficulty of directly measuring in vivo binding of large numbers of TFs to DNA across multiple primary cell types and their natural response to physiological stimuli.
Here, we overcome this bottleneck by systematically exploring the genomic binding network of 1. All relevant TFs of key hematopoietic cells in both steady state and under relevant stimuli. 2. Follow the changes in TF networks as cells differentiate 3. Use these models to engineer cell states and responses. To achieve these goals, we developed a new method for automated high throughput ChIP coupled to sequencing (HT-ChIP-Seq). We used this method to measure binding of 40 TFs in 4 time points following stimulation of dendritic cells with pathogen components. We find that TFs vary substantially in their binding dynamics, genomic localization, number of binding events, and degree of interaction with other TFs. The analysis of this data suggests that the TF network is hierarchically organized, and composed of different types of TFs, cell differentiation factors, factors that prime for gene induction, and factors that bind more specifically and dynamically. This proposal revisits and challenges the current understanding of the mammalian regulatory code.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym DROUGHT-HEAT
Project Land-Climate Interactions: Constraints for Droughts and Heatwaves in a Changing Climate
Researcher (PI) Sonia Isabelle Seneviratne
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Consolidator Grant (CoG), PE10, ERC-2013-CoG
Summary "Land-climate interactions mediated through soil moisture and vegetation play a critical role in the climate system, in particular for the occurrence of extreme events such as droughts and heatwaves. They are, however, poorly constrained in current Earth System Models (ESMs), leading to large uncertainties in climate projections. These uncertainties affect the quality and accuracy of projections of temperature, water availability, and carbon concentrations, as well as that of projected impacts on agriculture, ecosystems, and health.
In the past years, in-situ and remote sensing-based datasets of soil moisture, evapotranspiration, and energy and carbon fluxes have become increasingly available, providing untapped potential for reducing associated uncertainties in current climate models. The DROUGHT-HEAT project aims at innovatively exploiting these new information sources in order to 1) derive observations-based diagnostics to quantify and isolate the role of land-climate interactions in past extreme events (""Diagnostic Atlas""), 2) evaluate and improve current ESMs and constrain climate-change projections using the derived diagnostics, and 3) apply the newly gained knowledge to frontier developments in the attribution of climate extremes to land processes and their mitigation through ""land geoengineering"".
The DROUGHT-HEAT project integrates the newest land observational datasets with the latest stream of ESMs. Novel methodologies will be applied to extract functional relationships from the data, and identify key gaps in the ESMs' representation of underlying processes. These will build on physically-based relationships, machine learning tools, and model calibration. In addition, they will encompass the mapping and merging of derived diagnostics in space and time to reduce ""blank spaces"" in the datasets. The project is unprecedented in its breadth and scope and will allow a major breakthrough in our understanding of the processes leading to heatwaves and droughts."
Summary
"Land-climate interactions mediated through soil moisture and vegetation play a critical role in the climate system, in particular for the occurrence of extreme events such as droughts and heatwaves. They are, however, poorly constrained in current Earth System Models (ESMs), leading to large uncertainties in climate projections. These uncertainties affect the quality and accuracy of projections of temperature, water availability, and carbon concentrations, as well as that of projected impacts on agriculture, ecosystems, and health.
In the past years, in-situ and remote sensing-based datasets of soil moisture, evapotranspiration, and energy and carbon fluxes have become increasingly available, providing untapped potential for reducing associated uncertainties in current climate models. The DROUGHT-HEAT project aims at innovatively exploiting these new information sources in order to 1) derive observations-based diagnostics to quantify and isolate the role of land-climate interactions in past extreme events (""Diagnostic Atlas""), 2) evaluate and improve current ESMs and constrain climate-change projections using the derived diagnostics, and 3) apply the newly gained knowledge to frontier developments in the attribution of climate extremes to land processes and their mitigation through ""land geoengineering"".
The DROUGHT-HEAT project integrates the newest land observational datasets with the latest stream of ESMs. Novel methodologies will be applied to extract functional relationships from the data, and identify key gaps in the ESMs' representation of underlying processes. These will build on physically-based relationships, machine learning tools, and model calibration. In addition, they will encompass the mapping and merging of derived diagnostics in space and time to reduce ""blank spaces"" in the datasets. The project is unprecedented in its breadth and scope and will allow a major breakthrough in our understanding of the processes leading to heatwaves and droughts."
Max ERC Funding
1 952 285 €
Duration
Start date: 2014-09-01, End date: 2019-08-31
Project acronym DYNACLOCK
Project Dynamic protein-DNA interactomes and circadian transcription regulatory networks in mammals
Researcher (PI) Felix Naef
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Starting Grant (StG), LS2, ERC-2010-StG_20091118
Summary The aim of this project is to understand the dynamics of protein-DNA interactomes underlying circadian oscillators in mammals, and how these shape circadian transcriptional output programs. Specifically our goal is to solve a fundamental issue in circadian biology: the phase specificity problem underlying circadian gene expression. We have taken a challenging and original multi-disciplinary approach in which molecular biology experiments will be tightly interlinked with computational analyses and biophysical modeling. The approach will generate time resolved protein-DNA interactomes in mouse liver for several key circadian repressors at unprecedented resolution. These experiments will be complemented with chromosome conformation capture (3C) experiments to monitor how looping interactions and 3D genome structure rearrange during the circadian cycle, which will inform on how circadian transcription networks use long-range gene regulatory mechanisms. Novel computational algorithms based on biophysical principles will be developed and implemented to optimally analyze interactome and 3C datasets. For the latter, statistical models from polymer physics will be used to reconstruct the chromatin networks and interaction maps from the 3C data. At the detailed level of individual cells, we will investigate transcription bursts, and how those are involved in the control of circadian gene expression. In particular we will exploit high temporal resolution bioluminescence reporters using a biophysical model of transcription coupled with a Hidden Markov Model (HMM). Through our innovative approach, we expect that the data generated and state-of-the-art analyses performed will lead novel insight into the role and mechanics of circadian transcription in controlling circadian outputs in mammals.
Summary
The aim of this project is to understand the dynamics of protein-DNA interactomes underlying circadian oscillators in mammals, and how these shape circadian transcriptional output programs. Specifically our goal is to solve a fundamental issue in circadian biology: the phase specificity problem underlying circadian gene expression. We have taken a challenging and original multi-disciplinary approach in which molecular biology experiments will be tightly interlinked with computational analyses and biophysical modeling. The approach will generate time resolved protein-DNA interactomes in mouse liver for several key circadian repressors at unprecedented resolution. These experiments will be complemented with chromosome conformation capture (3C) experiments to monitor how looping interactions and 3D genome structure rearrange during the circadian cycle, which will inform on how circadian transcription networks use long-range gene regulatory mechanisms. Novel computational algorithms based on biophysical principles will be developed and implemented to optimally analyze interactome and 3C datasets. For the latter, statistical models from polymer physics will be used to reconstruct the chromatin networks and interaction maps from the 3C data. At the detailed level of individual cells, we will investigate transcription bursts, and how those are involved in the control of circadian gene expression. In particular we will exploit high temporal resolution bioluminescence reporters using a biophysical model of transcription coupled with a Hidden Markov Model (HMM). Through our innovative approach, we expect that the data generated and state-of-the-art analyses performed will lead novel insight into the role and mechanics of circadian transcription in controlling circadian outputs in mammals.
Max ERC Funding
1 500 000 €
Duration
Start date: 2011-03-01, End date: 2016-02-29
Project acronym EARLYEARTH
Project Accretion and Differentiation of Terrestrial Planets
Researcher (PI) Maria Schoenbaechler
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Starting Grant (StG), PE10, ERC-2011-StG_20101014
Summary This proposal aims to constrain the late accretion history of the Earth and the differentiation of the earliest silicate reservoirs in planets. Highly siderophile elements (HSE) constrain the late accretion of material onto the Earth; a process that potentially delivered water to Earth. During core formation, HSE strongly partition into metal. Once core formation ceases, newly accreted HSE-rich material will significantly contribute to the HSE budget of the Earth’s mantle. The HSE are more abundant in the Earth’s mantle than predicted from low temperature partitioning experiments and feature nearly chondritic relative abundances. This implies a significant late accretion of chondritic material (“the late veneer”). This idea is challenged by high pressure/temperature experiments indicating that the HSE were left in the behind in the mantle during core formation, thereby calling into question the late veneer. To address this issue, I propose the setup of new isotopic tracers and utilize (i) nucleosynthetic anomalies and (ii) stable isotope systematics of the HSE to determine the origin of HSE in the Earth’s mantle. Unravelling this issue is a major advance in understanding planetary accretion. Formation of the earliest silicate reservoirs probably occurred contemporary to late accretion. Global differentiation in terrestrial silicate reservoirs may have taken place within the first 30 million years of the Earth’s formation based on Sm-Nd isotope data. This timing has been debated on various grounds. The 92Nb-92Zr decay system is a potentially powerful chronometer to further constrain this issue. Its usefulness, however, has been hindered by uncertainties of the initial 92Nb abundance in the solar system. I propose to obtain unequivocal evidence from old differentiated meteorites to settle this debate. The results will have implications for understanding early silicate differentiation on asteroids and - depending on the initial 92Nb abundance - the Earth and Mars.
Summary
This proposal aims to constrain the late accretion history of the Earth and the differentiation of the earliest silicate reservoirs in planets. Highly siderophile elements (HSE) constrain the late accretion of material onto the Earth; a process that potentially delivered water to Earth. During core formation, HSE strongly partition into metal. Once core formation ceases, newly accreted HSE-rich material will significantly contribute to the HSE budget of the Earth’s mantle. The HSE are more abundant in the Earth’s mantle than predicted from low temperature partitioning experiments and feature nearly chondritic relative abundances. This implies a significant late accretion of chondritic material (“the late veneer”). This idea is challenged by high pressure/temperature experiments indicating that the HSE were left in the behind in the mantle during core formation, thereby calling into question the late veneer. To address this issue, I propose the setup of new isotopic tracers and utilize (i) nucleosynthetic anomalies and (ii) stable isotope systematics of the HSE to determine the origin of HSE in the Earth’s mantle. Unravelling this issue is a major advance in understanding planetary accretion. Formation of the earliest silicate reservoirs probably occurred contemporary to late accretion. Global differentiation in terrestrial silicate reservoirs may have taken place within the first 30 million years of the Earth’s formation based on Sm-Nd isotope data. This timing has been debated on various grounds. The 92Nb-92Zr decay system is a potentially powerful chronometer to further constrain this issue. Its usefulness, however, has been hindered by uncertainties of the initial 92Nb abundance in the solar system. I propose to obtain unequivocal evidence from old differentiated meteorites to settle this debate. The results will have implications for understanding early silicate differentiation on asteroids and - depending on the initial 92Nb abundance - the Earth and Mars.
Max ERC Funding
1 994 545 €
Duration
Start date: 2012-04-01, End date: 2017-12-31
Project acronym EcCRISPR
Project Novel roles, components, and mechanisms of the Escherichia coli CRISPR/Cas system
Researcher (PI) Ehud Itzhak Qimron
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS2, ERC-2013-StG
Summary A novel type of defense system was recently identified in bacteria: the CRISPR array and its associated gene products (Cas). The system inserts short DNA sequences, called spacers, derived from foreign nucleic acid molecules in between direct repeats, thus forming the CRISPR array. The transcribed spacers eventually serve as molecular guides for Cas proteins that monitor and destroy nucleic acids having sequences similar to those spacers. Thorough mapping of the functional components and regulators of the system in a single model organism will be extremely valuable for understanding its mechanism of action. Studying the interactions between bacteria and phages should highlight the evolutionary role of the system and its consequences for shaping ecological systems. These insights will lead to novel ways of exploiting the system to improve molecular biology tools, to protect fermenting bacteria from phage spoilage, to equip phages with anti-CRISPR warfare to fight bacteria, and to prevent horizontal gene transfer between pathogens. Here, I intend to systematically seek out new roles of the system and to identify fundamental mechanisms and components that allow the system to function efficiently. I will address fundamental questions such as how the system avoids sampling self DNA into the CRISPR array. In addition, I will pursue two revolutionary possibilities. One, that the CRISPR/Cas system is not merely an adaptive defense system against phages, but that one of its roles is to serve as molecular machinery for silencing specific harmful genes by generating small silencing RNAs without the need for Cas proteins. The other is to test the system’s ability to prevent horizontal gene transfer of antibiotic resistance genes in an effort to study the system’s ecological value, potentially for applicative uses. My proposed studies will allow deeper understanding of the system, and enable breakthroughs from both basic and applicative aspects of the CRISPR field studies.
Summary
A novel type of defense system was recently identified in bacteria: the CRISPR array and its associated gene products (Cas). The system inserts short DNA sequences, called spacers, derived from foreign nucleic acid molecules in between direct repeats, thus forming the CRISPR array. The transcribed spacers eventually serve as molecular guides for Cas proteins that monitor and destroy nucleic acids having sequences similar to those spacers. Thorough mapping of the functional components and regulators of the system in a single model organism will be extremely valuable for understanding its mechanism of action. Studying the interactions between bacteria and phages should highlight the evolutionary role of the system and its consequences for shaping ecological systems. These insights will lead to novel ways of exploiting the system to improve molecular biology tools, to protect fermenting bacteria from phage spoilage, to equip phages with anti-CRISPR warfare to fight bacteria, and to prevent horizontal gene transfer between pathogens. Here, I intend to systematically seek out new roles of the system and to identify fundamental mechanisms and components that allow the system to function efficiently. I will address fundamental questions such as how the system avoids sampling self DNA into the CRISPR array. In addition, I will pursue two revolutionary possibilities. One, that the CRISPR/Cas system is not merely an adaptive defense system against phages, but that one of its roles is to serve as molecular machinery for silencing specific harmful genes by generating small silencing RNAs without the need for Cas proteins. The other is to test the system’s ability to prevent horizontal gene transfer of antibiotic resistance genes in an effort to study the system’s ecological value, potentially for applicative uses. My proposed studies will allow deeper understanding of the system, and enable breakthroughs from both basic and applicative aspects of the CRISPR field studies.
Max ERC Funding
1 499 000 €
Duration
Start date: 2013-12-01, End date: 2018-11-30
Project acronym ENCODE
Project Design Principles in Encoding Complex Noisy Environments
Researcher (PI) Alon Zaslaver
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS2, ERC-2013-StG
Summary Animals constantly face complex environments consisted of multiple fluctuating cues. Accurate detection and efficient integration of such perplexing information are essential as animals’ fitness and consequently survival depend on making the right behavioral decisions. However, little is known about how multifaceted stimuli are integrated by neural systems, and how this information flows in the neural network in a single-neuron resolution.
Here we aim to address these fundamental questions using C. elegans worms as a model system. With a compact and fully-mapped neural network, C. elegans offers a unique opportunity of generating novel breakthroughs and significantly advance the field.
To study functional dynamics on a network-wide scale with an unprecedented single-neuron resolution, we will construct a comprehensive library of transgenic animals expressing state-of-the-art optogenetic tools and Calcium indicators in individual neurons. Moreover, we will study the entire encoding process, beginning with the sensory layer, through integration in the neural network, to behavioral outputs. At the sensory level, we aim to reveal how small sensory systems efficiently encode the complex external world. Next, we will decipher the design principles by which neural circuits integrate and process information. The optogenetic transgenic animals will allow us interrogating computational roles of various circuits by manipulating individual neurons in the network. At the end, we will integrate the gathered knowledge to study how encoding eventually translates to decision making behavioral outputs.
Throughout this project, we will use a combination of cutting-edge experimental techniques coupled with extensive computational analyses, modelling and theory. The aims of this interdisciplinary project together with the system-level approaches put it in the front line of research in the Systems Biology field.
Summary
Animals constantly face complex environments consisted of multiple fluctuating cues. Accurate detection and efficient integration of such perplexing information are essential as animals’ fitness and consequently survival depend on making the right behavioral decisions. However, little is known about how multifaceted stimuli are integrated by neural systems, and how this information flows in the neural network in a single-neuron resolution.
Here we aim to address these fundamental questions using C. elegans worms as a model system. With a compact and fully-mapped neural network, C. elegans offers a unique opportunity of generating novel breakthroughs and significantly advance the field.
To study functional dynamics on a network-wide scale with an unprecedented single-neuron resolution, we will construct a comprehensive library of transgenic animals expressing state-of-the-art optogenetic tools and Calcium indicators in individual neurons. Moreover, we will study the entire encoding process, beginning with the sensory layer, through integration in the neural network, to behavioral outputs. At the sensory level, we aim to reveal how small sensory systems efficiently encode the complex external world. Next, we will decipher the design principles by which neural circuits integrate and process information. The optogenetic transgenic animals will allow us interrogating computational roles of various circuits by manipulating individual neurons in the network. At the end, we will integrate the gathered knowledge to study how encoding eventually translates to decision making behavioral outputs.
Throughout this project, we will use a combination of cutting-edge experimental techniques coupled with extensive computational analyses, modelling and theory. The aims of this interdisciplinary project together with the system-level approaches put it in the front line of research in the Systems Biology field.
Max ERC Funding
1 498 400 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym EPICROP
Project Dissecting epistasis for enhanced crop productivity
Researcher (PI) Sebastian Soyk
Host Institution (HI) UNIVERSITE DE LAUSANNE
Call Details Starting Grant (StG), LS2, ERC-2018-STG
Summary A major goal in plant biology is to understand how naturally occurring genetic variation leads to quantitative differences in economically important traits. Efforts to navigate the genotype-to-phenotype map are often focused on linear genetic interactions. As a result, crop breeding is mainly driven by loci with predictable additive effects. However, it has become clear that quantitative trait variation often results from perturbations of complex genetic networks. Thus, understanding epistasis, or interactions between genes, is key for our ability to predictably improve crops. To meet this challenge, this project will reveal and dissect epistatic interactions in gene regulatory networks that guide stem cell differentiation in the model crop tomato. In the first aim, I will utilize exhaustive allelic series for epistatic MADS-box genes that quantitatively regulate flower and fruit production as an experimental model system to study fundamental principles of epistasis that can be applied to other genetic networks. Genome-wide transcript profiling will be used to reveal molecular signatures of epistasis and potential targets for predictable crop improvement by advanced CRISPR/Cas9 gene editing technology. Further, my preliminary data suggests that epistasis is widespread and important across major productivity traits in tomato. Thus, in a second aim, I will access this untapped resource of cryptic genetic variation by sensitizing a tomato diversity panel for weak epistatic effects from unknown natural modifier loci of stem cell differentiation using trans-acting CRISPR/Cas9 editing cassettes. This screen represents a new approach to mutagenesis in plants with potential to reveal cryptic variation in other system. The outcomes of this project will advance our knowledge in a fundamental area of plant genome biology, help uncover and understand the functional architecture of epistasis, and have potential to bring significant improvements to agriculture.
Summary
A major goal in plant biology is to understand how naturally occurring genetic variation leads to quantitative differences in economically important traits. Efforts to navigate the genotype-to-phenotype map are often focused on linear genetic interactions. As a result, crop breeding is mainly driven by loci with predictable additive effects. However, it has become clear that quantitative trait variation often results from perturbations of complex genetic networks. Thus, understanding epistasis, or interactions between genes, is key for our ability to predictably improve crops. To meet this challenge, this project will reveal and dissect epistatic interactions in gene regulatory networks that guide stem cell differentiation in the model crop tomato. In the first aim, I will utilize exhaustive allelic series for epistatic MADS-box genes that quantitatively regulate flower and fruit production as an experimental model system to study fundamental principles of epistasis that can be applied to other genetic networks. Genome-wide transcript profiling will be used to reveal molecular signatures of epistasis and potential targets for predictable crop improvement by advanced CRISPR/Cas9 gene editing technology. Further, my preliminary data suggests that epistasis is widespread and important across major productivity traits in tomato. Thus, in a second aim, I will access this untapped resource of cryptic genetic variation by sensitizing a tomato diversity panel for weak epistatic effects from unknown natural modifier loci of stem cell differentiation using trans-acting CRISPR/Cas9 editing cassettes. This screen represents a new approach to mutagenesis in plants with potential to reveal cryptic variation in other system. The outcomes of this project will advance our knowledge in a fundamental area of plant genome biology, help uncover and understand the functional architecture of epistasis, and have potential to bring significant improvements to agriculture.
Max ERC Funding
1 499 903 €
Duration
Start date: 2019-08-01, End date: 2024-07-31
Project acronym EPIGEPLAS
Project Epigenetic determinants of the genome that govern cellular plasticity
Researcher (PI) Dirk Schübeler
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Call Details Starting Grant (StG), LS2, ERC-2007-StG
Summary Differentiation events in mammalian development involve stable resetting of transcriptional programs, which entails changes in the epigenetic state of target sequences defined by modifications of DNA and bound nucleosomes. These recently identified epigenetic layers modulate DNA accessibility in a positive and negative manner and thus could make genetic readouts context-dependent and dynamic. The proposed project aims to quantify the epigenetic contribution to cellular differentiation as a key event in development. By applying parallel genomic approaches we will comprehensively define the epigenome and its plasticity during cellular commitment of pluripotent murine stem cells into defined terminally differentiated cells. We will focus on DNA methylation and its interplay with several histone modifications as a way to achieve stable gene silencing. The resulting global profiles will gain insights into targeting principles and generate statistical, predictive models of regulation. From these mechanistic models will be derived and tested by genetically interfering with genetic and epigenetic regulatory pathways and by dissecting DNA sequence components involved in specifying targets. These experiments aim to unravel the crosstalk between epigenetic regulation and cell plasticity, the underlying molecular circuitry in pluripotent and unipotent cells and ultimately help to incorporate epigenetic regulation into current transcriptional regulatory models.
Summary
Differentiation events in mammalian development involve stable resetting of transcriptional programs, which entails changes in the epigenetic state of target sequences defined by modifications of DNA and bound nucleosomes. These recently identified epigenetic layers modulate DNA accessibility in a positive and negative manner and thus could make genetic readouts context-dependent and dynamic. The proposed project aims to quantify the epigenetic contribution to cellular differentiation as a key event in development. By applying parallel genomic approaches we will comprehensively define the epigenome and its plasticity during cellular commitment of pluripotent murine stem cells into defined terminally differentiated cells. We will focus on DNA methylation and its interplay with several histone modifications as a way to achieve stable gene silencing. The resulting global profiles will gain insights into targeting principles and generate statistical, predictive models of regulation. From these mechanistic models will be derived and tested by genetically interfering with genetic and epigenetic regulatory pathways and by dissecting DNA sequence components involved in specifying targets. These experiments aim to unravel the crosstalk between epigenetic regulation and cell plasticity, the underlying molecular circuitry in pluripotent and unipotent cells and ultimately help to incorporate epigenetic regulation into current transcriptional regulatory models.
Max ERC Funding
1 085 000 €
Duration
Start date: 2008-10-01, End date: 2013-09-30
Project acronym Epiherigans
Project Writing, reading and managing stress with H3K9me
Researcher (PI) Susan GASSER
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Call Details Advanced Grant (AdG), LS2, ERC-2016-ADG
Summary Epigenetic inheritance is the transmission of information, generally in the form of DNA methylation or post-translational modifications on histones that regulate the availability of underlying genetic information for transcription. RNA itself feeds back to contribute to histone modification. Sequence accessibility is both a matter of folding the chromatin fibre to alter access to recognition motifs, and the local concentration of factors needed for efficient transcriptional initiation, elongation, termination or mRNA stability. In heterochromatin we find a subset of regulatory factors in carefully balanced concentrations that are maintained in part by the segregation of active and inactive domains. Histone H3 K9 methylation is key to this compartmentation.
C. elegans provides an ideal system in which to study chromatin-based gene repression. We have demonstrated that histone H3 K9 methylation is the essential signal for the sequestration of heterochromatin at the nuclear envelope in C. elegans. The recognition of H3K9me1/2/3 by an inner nuclear envelope-bound chromodomain protein, CEC-4, actively sequesters heterochromatin in embryos, and contributes redundantly in adult tissues.
Epiherigans has the ambitious goal to determine definitively what targets H3K9 methylation, and identify its physiological roles. We will examine how this mark contributes to the epigenetic recognition of repeat vs non-repeat sequence, and mediates a stress-induced response to oxidative damage. We will examine the link between these and the spatial clustering of heterochromatic domains. Epiherigans will develop an integrated approach to identify in vivo the factors that distinguish repeats from non-repeats, self from non-self within genomes and will examine how H3K9me contributes to a persistent ROS or DNA damage stress response. It represents a crucial step towards understanding of how our genomes use heterochromatin to modulate, stabilize and transmit chromatin organization.
Summary
Epigenetic inheritance is the transmission of information, generally in the form of DNA methylation or post-translational modifications on histones that regulate the availability of underlying genetic information for transcription. RNA itself feeds back to contribute to histone modification. Sequence accessibility is both a matter of folding the chromatin fibre to alter access to recognition motifs, and the local concentration of factors needed for efficient transcriptional initiation, elongation, termination or mRNA stability. In heterochromatin we find a subset of regulatory factors in carefully balanced concentrations that are maintained in part by the segregation of active and inactive domains. Histone H3 K9 methylation is key to this compartmentation.
C. elegans provides an ideal system in which to study chromatin-based gene repression. We have demonstrated that histone H3 K9 methylation is the essential signal for the sequestration of heterochromatin at the nuclear envelope in C. elegans. The recognition of H3K9me1/2/3 by an inner nuclear envelope-bound chromodomain protein, CEC-4, actively sequesters heterochromatin in embryos, and contributes redundantly in adult tissues.
Epiherigans has the ambitious goal to determine definitively what targets H3K9 methylation, and identify its physiological roles. We will examine how this mark contributes to the epigenetic recognition of repeat vs non-repeat sequence, and mediates a stress-induced response to oxidative damage. We will examine the link between these and the spatial clustering of heterochromatic domains. Epiherigans will develop an integrated approach to identify in vivo the factors that distinguish repeats from non-repeats, self from non-self within genomes and will examine how H3K9me contributes to a persistent ROS or DNA damage stress response. It represents a crucial step towards understanding of how our genomes use heterochromatin to modulate, stabilize and transmit chromatin organization.
Max ERC Funding
2 500 000 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym ERNBPTC
Project Expression regulatory networks: beyond promoters and transcription control
Researcher (PI) Yitzhak Pilpel
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2007-StG
Summary "Gene expression in living cells is a most intricate molecular process, occurring in stages, each of which is regulated by a diversity of mechanisms. Among the various stages leading to gene expression, only transcription is relatively well understood, thanks to Genomics and bioinformatics. In contrast to the vast amounts of genome-wide data and a growing understanding of the structure of networks controlling transcription, we still lack quantitative, genome-wide knowledge of the mechanisms underlying regulation of mRNA degradation and translation. Among the unknowns are the identity of the regulators, their kinetic modes of action, and their means of interaction with the sequence features that make-up their targets; how these target combine to produce a higher level ""grammar"" is also unknown. An important part of the project is dedicated to generating genome-wide experimental data that will form the basis for quantitative and more comprehensive analysis of gene expression. Specifically, the primary objectives of our proposed research plan are: 1) to advance our understanding of the transcriptome, by deciphering the code regulating mRNA decay 2) to break the code which controls protein translation efficiency 3) to understand how mRNA degradation and translation efficiency determine noise in protein expression levels. The proposed strategy is based on an innovative combination of computational prediction, synthetic gene design, and genome-wide data acquisition, all culminating in extensive data analysis, mathematical modeling and focused experiments. This highly challenging, multidisciplinary project is likely to greatly enhance our knowledge of the various modes by which organisms regulate expression of their genomes, how these regulatory mechanisms are interrelated, how they generate precise response to environmental challenges and how they have evolved over time."
Summary
"Gene expression in living cells is a most intricate molecular process, occurring in stages, each of which is regulated by a diversity of mechanisms. Among the various stages leading to gene expression, only transcription is relatively well understood, thanks to Genomics and bioinformatics. In contrast to the vast amounts of genome-wide data and a growing understanding of the structure of networks controlling transcription, we still lack quantitative, genome-wide knowledge of the mechanisms underlying regulation of mRNA degradation and translation. Among the unknowns are the identity of the regulators, their kinetic modes of action, and their means of interaction with the sequence features that make-up their targets; how these target combine to produce a higher level ""grammar"" is also unknown. An important part of the project is dedicated to generating genome-wide experimental data that will form the basis for quantitative and more comprehensive analysis of gene expression. Specifically, the primary objectives of our proposed research plan are: 1) to advance our understanding of the transcriptome, by deciphering the code regulating mRNA decay 2) to break the code which controls protein translation efficiency 3) to understand how mRNA degradation and translation efficiency determine noise in protein expression levels. The proposed strategy is based on an innovative combination of computational prediction, synthetic gene design, and genome-wide data acquisition, all culminating in extensive data analysis, mathematical modeling and focused experiments. This highly challenging, multidisciplinary project is likely to greatly enhance our knowledge of the various modes by which organisms regulate expression of their genomes, how these regulatory mechanisms are interrelated, how they generate precise response to environmental challenges and how they have evolved over time."
Max ERC Funding
1 320 000 €
Duration
Start date: 2008-09-01, End date: 2013-08-31
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 EVOEPIC
Project Evolutionary mechanisms of epigenomic and chromosomal aberrations in cancer
Researcher (PI) Amos Tanay
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2012-StG_20111109
Summary Our working hypothesis is that tumorigenesis is an evolutionary process that fundamentally couples few major driving events (point mutations, rearrangements) with a complex flux of minor aberrations, many of which are epigenetic. We believe that these minor events are critical factors in the emergence of the cancer phenotype, and that understanding them is essential to the characterization of the disease. In particular, we hypothesize that a quantitative and principled evolutionary model for carcinogenesis is imperative for understanding the heterogeneity within tumor cell populations and predicting the effects of cancer therapies. We will therefore develop an interdisciplinary scheme that combines theoretical models of cancer evolution with in vitro evolutionary experiments and new methods for assaying the population heterogeneity of epigenomic organization. By developing techniques to interrogate DNA methylation and its interaction with other key epigenetic marks at the single-cell level, we will allow quantitative theoretical predictions to be scrutinized and refined. By combining models describing epigenetic aberrations with direct measurements of chromatin organization using Hi-C and 4C-seq, we shall revisit fundamental questions on the causative nature of epigenetic changes during carcinogenesis. Ultimately, we will apply both theoretical and experimental methodologies to assay and characterize the evolutionary histories of tumor cell populations from multiple mouse models and clinical patient samples.
Summary
Our working hypothesis is that tumorigenesis is an evolutionary process that fundamentally couples few major driving events (point mutations, rearrangements) with a complex flux of minor aberrations, many of which are epigenetic. We believe that these minor events are critical factors in the emergence of the cancer phenotype, and that understanding them is essential to the characterization of the disease. In particular, we hypothesize that a quantitative and principled evolutionary model for carcinogenesis is imperative for understanding the heterogeneity within tumor cell populations and predicting the effects of cancer therapies. We will therefore develop an interdisciplinary scheme that combines theoretical models of cancer evolution with in vitro evolutionary experiments and new methods for assaying the population heterogeneity of epigenomic organization. By developing techniques to interrogate DNA methylation and its interaction with other key epigenetic marks at the single-cell level, we will allow quantitative theoretical predictions to be scrutinized and refined. By combining models describing epigenetic aberrations with direct measurements of chromatin organization using Hi-C and 4C-seq, we shall revisit fundamental questions on the causative nature of epigenetic changes during carcinogenesis. Ultimately, we will apply both theoretical and experimental methodologies to assay and characterize the evolutionary histories of tumor cell populations from multiple mouse models and clinical patient samples.
Max ERC Funding
1 499 998 €
Duration
Start date: 2012-12-01, End date: 2017-11-30
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 FEVER
Project Forecasting the recurrence rate of volcanic eruptions
Researcher (PI) Luca Caricchi
Host Institution (HI) UNIVERSITE DE GENEVE
Call Details Starting Grant (StG), PE10, ERC-2015-STG
Summary Volcanic eruptions occur with a frequency that is inversely proportional to their magnitude. Datasets of natural volcanic events, currently used to determine the recurrence rate of volcanic eruptions are intrinsically biased. Combining physical modelling of processes with detailed statistical analysis has been demonstrated essential for assessing reliably the recurrence rate of natural hazards, such as floods and earthquakes. This would be the first attempt to apply a similar, integrated approach to explosive volcanic eruptions.
The high-gain final target of FEVER is to produce a physically based statistical model able to ForEcast the recurrence rate of Volcanic Eruptions both at regional and global scale. This is the first project of this kind and consequently involves a significant risk. Because 500 million people live in proximity of volcanoes and eruptions have a significant social and economical impact, forecasting the recurrence rate of volcanic eruption remains a great challenge in Science.
This project builds on two main directions of my research: a) Thermo-mechanical and statistical modelling targeting the identification of the main physical factors controlling the recurrence rate of volcanic eruptions. We showed that the flux of magma from depth directly controls the magnitude of the largest possible eruptions. Thus, b) we developed a novel method to determine such magma fluxes. These two lines of research combine perfectly in FEVER and will be integrated to answer questions such as: What is the probability of an eruption similar to the Tambora 1815 to occur in the next 100 years on Earth or in Europe? What is the largest physically possible eruption that can occur in Europe?
The high-gain target of FEVER is to mitigate the impact of volcanic eruptions on our society, by producing research of interest for governmental agencies dealing with location of strategic infrastructures, and for businesses such as aviation.
Summary
Volcanic eruptions occur with a frequency that is inversely proportional to their magnitude. Datasets of natural volcanic events, currently used to determine the recurrence rate of volcanic eruptions are intrinsically biased. Combining physical modelling of processes with detailed statistical analysis has been demonstrated essential for assessing reliably the recurrence rate of natural hazards, such as floods and earthquakes. This would be the first attempt to apply a similar, integrated approach to explosive volcanic eruptions.
The high-gain final target of FEVER is to produce a physically based statistical model able to ForEcast the recurrence rate of Volcanic Eruptions both at regional and global scale. This is the first project of this kind and consequently involves a significant risk. Because 500 million people live in proximity of volcanoes and eruptions have a significant social and economical impact, forecasting the recurrence rate of volcanic eruption remains a great challenge in Science.
This project builds on two main directions of my research: a) Thermo-mechanical and statistical modelling targeting the identification of the main physical factors controlling the recurrence rate of volcanic eruptions. We showed that the flux of magma from depth directly controls the magnitude of the largest possible eruptions. Thus, b) we developed a novel method to determine such magma fluxes. These two lines of research combine perfectly in FEVER and will be integrated to answer questions such as: What is the probability of an eruption similar to the Tambora 1815 to occur in the next 100 years on Earth or in Europe? What is the largest physically possible eruption that can occur in Europe?
The high-gain target of FEVER is to mitigate the impact of volcanic eruptions on our society, by producing research of interest for governmental agencies dealing with location of strategic infrastructures, and for businesses such as aviation.
Max ERC Funding
1 458 192 €
Duration
Start date: 2016-04-01, End date: 2021-03-31
Project acronym FORECASToneMONTH
Project Forecasting Surface Weather and Climate at One-Month Leads through Stratosphere-Troposphere Coupling
Researcher (PI) Chaim Israel Garfinkel
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), PE10, ERC-2015-STG
Summary Anomalies in surface temperatures, winds, and precipitation can significantly alter energy supply and demand, cause flooding, and cripple transportation networks. Better management of these impacts can be achieved by extending the duration of reliable predictions of the atmospheric circulation.
Polar stratospheric variability can impact surface weather for well over a month, and this proposed research presents a novel approach towards understanding the fundamentals of how this coupling occurs. Specifically, we are interested in: 1) how predictable are anomalies in the stratospheric circulation? 2) why do only some stratospheric events modify surface weather? and 3) what is the mechanism whereby stratospheric anomalies reach the surface? While this last question may appear academic, several studies indicate that stratosphere-troposphere coupling drives the midlatitude tropospheric response to climate change; therefore, a clearer understanding of the mechanisms will aid in the interpretation of the upcoming changes in the surface climate.
I propose a multi-pronged effort aimed at addressing these questions and improving monthly forecasting. First, carefully designed modelling experiments using a novel modelling framework will be used to clarify how, and under what conditions, stratospheric variability couples to tropospheric variability. Second, novel linkages between variability external to the stratospheric polar vortex and the stratospheric polar vortex will be pursued, thus improving our ability to forecast polar vortex variability itself. To these ends, my group will develop 1) an analytic model for Rossby wave propagation on the sphere, and 2) a simplified general circulation model, which captures the essential processes underlying stratosphere-troposphere coupling. By combining output from the new models, observational data, and output from comprehensive climate models, the connections between the stratosphere and surface climate will be elucidated.
Summary
Anomalies in surface temperatures, winds, and precipitation can significantly alter energy supply and demand, cause flooding, and cripple transportation networks. Better management of these impacts can be achieved by extending the duration of reliable predictions of the atmospheric circulation.
Polar stratospheric variability can impact surface weather for well over a month, and this proposed research presents a novel approach towards understanding the fundamentals of how this coupling occurs. Specifically, we are interested in: 1) how predictable are anomalies in the stratospheric circulation? 2) why do only some stratospheric events modify surface weather? and 3) what is the mechanism whereby stratospheric anomalies reach the surface? While this last question may appear academic, several studies indicate that stratosphere-troposphere coupling drives the midlatitude tropospheric response to climate change; therefore, a clearer understanding of the mechanisms will aid in the interpretation of the upcoming changes in the surface climate.
I propose a multi-pronged effort aimed at addressing these questions and improving monthly forecasting. First, carefully designed modelling experiments using a novel modelling framework will be used to clarify how, and under what conditions, stratospheric variability couples to tropospheric variability. Second, novel linkages between variability external to the stratospheric polar vortex and the stratospheric polar vortex will be pursued, thus improving our ability to forecast polar vortex variability itself. To these ends, my group will develop 1) an analytic model for Rossby wave propagation on the sphere, and 2) a simplified general circulation model, which captures the essential processes underlying stratosphere-troposphere coupling. By combining output from the new models, observational data, and output from comprehensive climate models, the connections between the stratosphere and surface climate will be elucidated.
Max ERC Funding
1 808 000 €
Duration
Start date: 2016-05-01, End date: 2021-04-30
Project acronym FRONTIERS OF RNAI-II
Project High resolution and chemical genetic approaches to RNA silencing mechanisms
Researcher (PI) Olivier Robert Georges Voinnet
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), LS2, ERC-2012-ADG_20120314
Summary In eukaryotes, silencing small (s)RNAs, including micro (mi)RNAs and small interfering (si)RNAs, regulate many aspects of biology, including cell differentiation, development, hormonal responses, or defense. In particular, many plant and metazoan miRNAs play crucial roles in embryonic/post-embryonic development; the precise timing and localization of their expression is thus crucial to their action. Hence, specific miRNA repertoires underlie specific cell identities, and deviations from such repertoires often have deleterious consequences such as cancer. Many miRNAs also help organisms to adapt to stress, thus, given their importance in virtually all aspects of biology, understanding how, when and where miRNAs exert their actions is of paramount importance. To date, however, the few approaches to miRNA-mediated silencing in whole organisms have not taken into account the exquisite definition, in space and time, of their biogenesis and action, leading to an inaccurate view of the biology of these molecules at the systems level.
Using the root system of the model plant Arabidopsis thaliana, we propose to explore, at single-cell and subcellular resolution levels, the biology of the main miRNA effector protein, ARGONAUTE 1 (AGO1) in intact tissues. Using a combination of state-of the-art technologies for single-cell forward genetics, protein purification and RNA/polysome profiling, we will establish a functional spatiotemporal map of the root AGO1-sRNAome and identify cell-specific modifiers of sRNA biogenesis and action. As a complement to the above approaches, chemical genetics will isolate small molecules allowing direct and specific manipulation of AGO1-dependent sRNA pathways in planta. RNA silencing modifier compounds will also accelerate forward and reverse approaches of RNA silencing in plants with sensitized genetic backgrounds, and uncover novel connections between miRNA/siRNA and physiological or metabolic pathways.
Summary
In eukaryotes, silencing small (s)RNAs, including micro (mi)RNAs and small interfering (si)RNAs, regulate many aspects of biology, including cell differentiation, development, hormonal responses, or defense. In particular, many plant and metazoan miRNAs play crucial roles in embryonic/post-embryonic development; the precise timing and localization of their expression is thus crucial to their action. Hence, specific miRNA repertoires underlie specific cell identities, and deviations from such repertoires often have deleterious consequences such as cancer. Many miRNAs also help organisms to adapt to stress, thus, given their importance in virtually all aspects of biology, understanding how, when and where miRNAs exert their actions is of paramount importance. To date, however, the few approaches to miRNA-mediated silencing in whole organisms have not taken into account the exquisite definition, in space and time, of their biogenesis and action, leading to an inaccurate view of the biology of these molecules at the systems level.
Using the root system of the model plant Arabidopsis thaliana, we propose to explore, at single-cell and subcellular resolution levels, the biology of the main miRNA effector protein, ARGONAUTE 1 (AGO1) in intact tissues. Using a combination of state-of the-art technologies for single-cell forward genetics, protein purification and RNA/polysome profiling, we will establish a functional spatiotemporal map of the root AGO1-sRNAome and identify cell-specific modifiers of sRNA biogenesis and action. As a complement to the above approaches, chemical genetics will isolate small molecules allowing direct and specific manipulation of AGO1-dependent sRNA pathways in planta. RNA silencing modifier compounds will also accelerate forward and reverse approaches of RNA silencing in plants with sensitized genetic backgrounds, and uncover novel connections between miRNA/siRNA and physiological or metabolic pathways.
Max ERC Funding
2 251 600 €
Duration
Start date: 2013-07-01, End date: 2018-06-30
Project acronym Gendever
Project Genome, the Edited Version: DNA and RNA Editing of Mammalian Retroelements
Researcher (PI) Erez Levanon
Host Institution (HI) BAR ILAN UNIVERSITY
Call Details Starting Grant (StG), LS2, ERC-2012-StG_20111109
Summary It is generally thought that an organism contains the exactly same genomic information in all its cells and that a genome remains unaltered throughout the organism’s life, with the exception of rare and random somatic mutations that might occur. This genomic information will also serve as a template for exact RNA copies. However, endogenous and powerful means of creating inner genomic diversity are known to exist: (1) RNA editing that leads to alteration of one nucleotide into another, (mainly A-to-I); (2) DNA editing that changes the DNA’s content by shifting C-into-U; (3) active retroelements that can insert copies of their sequences into new locations in a genome.
Recently, we and others have found that although considered extremely rare, all three mechanisms are active somatically or at least leave traces of their occurrence in the genome, and are linked together, as most editing events occur in retroelements. However, the magnitude and scope of these mechanisms, which can lead to huge diversity and complexity within an organism and even within a cell, are still a mystery. This explosion of genomic variety can have dramatic effect on diverse biological processes, such as brain complexity, cancer and evolution acceleration.
In GENEDVER, we aim to perform the first genome-wide mapping of editing and active retroelements in various genomes using a combination of computational and genomic approaches. Specifically, we will develop a strategy to detect RNA and DNA editing in retroelements, scan for editing events in various genomes, and build the first global editing atlas. In addition, we will exploit the close association between editing and retroelements in to produce a genome-wide approach to detect active retroelements. Finally, we will screen for editing events and retrotranspositions in various biological conditions, in order to expose their involvement in many biological states and evolution.
Summary
It is generally thought that an organism contains the exactly same genomic information in all its cells and that a genome remains unaltered throughout the organism’s life, with the exception of rare and random somatic mutations that might occur. This genomic information will also serve as a template for exact RNA copies. However, endogenous and powerful means of creating inner genomic diversity are known to exist: (1) RNA editing that leads to alteration of one nucleotide into another, (mainly A-to-I); (2) DNA editing that changes the DNA’s content by shifting C-into-U; (3) active retroelements that can insert copies of their sequences into new locations in a genome.
Recently, we and others have found that although considered extremely rare, all three mechanisms are active somatically or at least leave traces of their occurrence in the genome, and are linked together, as most editing events occur in retroelements. However, the magnitude and scope of these mechanisms, which can lead to huge diversity and complexity within an organism and even within a cell, are still a mystery. This explosion of genomic variety can have dramatic effect on diverse biological processes, such as brain complexity, cancer and evolution acceleration.
In GENEDVER, we aim to perform the first genome-wide mapping of editing and active retroelements in various genomes using a combination of computational and genomic approaches. Specifically, we will develop a strategy to detect RNA and DNA editing in retroelements, scan for editing events in various genomes, and build the first global editing atlas. In addition, we will exploit the close association between editing and retroelements in to produce a genome-wide approach to detect active retroelements. Finally, we will screen for editing events and retrotranspositions in various biological conditions, in order to expose their involvement in many biological states and evolution.
Max ERC Funding
1 499 249 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym GeneBodyMethylation
Project Resolving the Nuts and Bolts of Gene Body Methylation
Researcher (PI) Assaf Zemach
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS2, ERC-2015-STG
Summary DNA methylation, the covalent binding of a methyl group (CH3) to cytosine base, regulates the genome activity and plays a fundamental developmental role in eukaryotes. Its epigenetic characteristics of regulating transcription without changing the genetic code together with the ability to be transmitted through DNA replication allow organisms to memorize cellular events for many generations. DNA methylation is mostly known for its role in transcriptional silencing; however, its functional output is more complex and is influenced in part by its DNA context. Recent genomic studies, have found DNA methylation to be targeted inside sequences of actively transcribed genes, thus termed gene body methylation. Despite being an evolutionary conserved and a robust methylation pathway targeted to thousands of genes in animal and plant genomes, the function of gene body methylation is still poorly understood at both the molecular and functional level. Similar to the chicken and egg conundrum, because we do not know what gene body methylation does, therefore scientists could not apply its function to discover its regulators either. Gene body methylation is targeted to a very specific subset and subregions of genes, thus we strongly believe that specific factors exist and are missing simply because that no one has ever searched for them before. Hence, to make major breakthroughs in the field, our approach is to artificially generate gene-body-specific hypomethylated plants that together with customized genetic and biochemical systems will allow us to discover regulators and interpreters of gene body methylation. Using these unique genetic tools and novel molecular factors, we will be able to ultimately explore the particular biological roles of gene body methylation. These findings will fill the gap towards a full comprehension of the entire functional array of DNA methylation, and to its more precise exploitation in yielding better crops and in treating human diseases.
Summary
DNA methylation, the covalent binding of a methyl group (CH3) to cytosine base, regulates the genome activity and plays a fundamental developmental role in eukaryotes. Its epigenetic characteristics of regulating transcription without changing the genetic code together with the ability to be transmitted through DNA replication allow organisms to memorize cellular events for many generations. DNA methylation is mostly known for its role in transcriptional silencing; however, its functional output is more complex and is influenced in part by its DNA context. Recent genomic studies, have found DNA methylation to be targeted inside sequences of actively transcribed genes, thus termed gene body methylation. Despite being an evolutionary conserved and a robust methylation pathway targeted to thousands of genes in animal and plant genomes, the function of gene body methylation is still poorly understood at both the molecular and functional level. Similar to the chicken and egg conundrum, because we do not know what gene body methylation does, therefore scientists could not apply its function to discover its regulators either. Gene body methylation is targeted to a very specific subset and subregions of genes, thus we strongly believe that specific factors exist and are missing simply because that no one has ever searched for them before. Hence, to make major breakthroughs in the field, our approach is to artificially generate gene-body-specific hypomethylated plants that together with customized genetic and biochemical systems will allow us to discover regulators and interpreters of gene body methylation. Using these unique genetic tools and novel molecular factors, we will be able to ultimately explore the particular biological roles of gene body methylation. These findings will fill the gap towards a full comprehension of the entire functional array of DNA methylation, and to its more precise exploitation in yielding better crops and in treating human diseases.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym GENEREGULATION
Project Deciphering the code of gene regulation using massively parallel assays of designed sequence libraries
Researcher (PI) Eran Segal
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS2, ERC-2013-CoG
Summary Many gene expression changes that are associated with disease states have in turn been linked to changes in the genes’ regulatory regions. However, without a ‘regulatory code’ that informs us how DNA sequences determine expression levels, we cannot predict which sequence changes will affect expression, by how much, and by what mechanism.
Here, we aim to arrive at a mechanistic and quantitative understanding of how expression levels are encoded in DNA sequence using a combined experimental and computational approach. To this end, we will construct libraries of >50,000 sequences, fuse them to fluorescent reporters, and genomically integrate them to yeast or human cells. We will then develop methods for accurately measuring, in parallel, the expression of each fused sequence within a single experiment, and for measuring the DNA binding state of each sequence at single cell resolution, resulting in ~1000-fold increase in the scale with which we can study the effect of sequence on expression.
Notably, we will design our experimental system to be modular, allowing us to propose a highly ambitious yet realistic plan in which we will study the effect of sequence on (1) transcriptional and (2) post-transcriptional regulation; (3) Unravel the effect of genetic variation across human individuals on expression; (4) Quantify how cellular fitness depends on the expression level of individual endogenous genes; and (5) Construct a predictive model of the effect of DNA sequence on expression.
Each of our libraries should provide novel insights into a different aspect of gene regulation, leading to new means by which we can interpret whole genome sequencing, which is rapidly being collected for many individuals. In particular, our unified model should allow us to predict expression changes among human individuals based only on their genotypic variation, greatly enhancing the ability to identify common or rare sequence variants that may affect molecular function or cause disease.
Summary
Many gene expression changes that are associated with disease states have in turn been linked to changes in the genes’ regulatory regions. However, without a ‘regulatory code’ that informs us how DNA sequences determine expression levels, we cannot predict which sequence changes will affect expression, by how much, and by what mechanism.
Here, we aim to arrive at a mechanistic and quantitative understanding of how expression levels are encoded in DNA sequence using a combined experimental and computational approach. To this end, we will construct libraries of >50,000 sequences, fuse them to fluorescent reporters, and genomically integrate them to yeast or human cells. We will then develop methods for accurately measuring, in parallel, the expression of each fused sequence within a single experiment, and for measuring the DNA binding state of each sequence at single cell resolution, resulting in ~1000-fold increase in the scale with which we can study the effect of sequence on expression.
Notably, we will design our experimental system to be modular, allowing us to propose a highly ambitious yet realistic plan in which we will study the effect of sequence on (1) transcriptional and (2) post-transcriptional regulation; (3) Unravel the effect of genetic variation across human individuals on expression; (4) Quantify how cellular fitness depends on the expression level of individual endogenous genes; and (5) Construct a predictive model of the effect of DNA sequence on expression.
Each of our libraries should provide novel insights into a different aspect of gene regulation, leading to new means by which we can interpret whole genome sequencing, which is rapidly being collected for many individuals. In particular, our unified model should allow us to predict expression changes among human individuals based only on their genotypic variation, greatly enhancing the ability to identify common or rare sequence variants that may affect molecular function or cause disease.
Max ERC Funding
2 000 000 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym GeoArchMag
Project Beyond the Holocene Geomagnetic field resolution
Researcher (PI) Ron Shaar
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), PE10, ERC-2018-STG
Summary For decades the Holocene has been considered a flat and “boring” epoch from the standpoint of
paleomagnetism, mainly due to insufficient resolution of the available paleomagnetic data. However, recent
archaeomagnetic data have revealed that the Holocene geomagnetic field is anything but stable – presenting
puzzling intervals of extreme decadal-scale fluctuations and unexpected departures from a simple dipolar field
structure. This new information introduced an entirely new paradigm to the study of the geomagnetic field and
to a wide range of research areas relying on paleomagnetic data, such as geochronology, climate research, and
geodynamo exploration.
This proposal aims at breaking the resolution limits in paleomagnetism, and providing a continuous
time series of the geomagnetic field vector throughout the Holocene at decadal resolution and
unprecedented accuracy. To this end I will use an innovative assemblage of data sources, jointly unique to
the Levant, including rare archaeological finds, annual laminated stalagmites, varved sediments, and arid
playa deposits. Together, these sources can provide unprecedented yearly resolution, whereby the “absolute”
archaeomagnetic data can calibrate “relative” terrestrial data.
The geomagnetic data will define an innovative absolute geomagnetic chronology that will be used to
synchronize cosmogenic 10Be data and an extensive body of paleo-climatic indicators. With these in hand, I
will address four ground-breaking problems:
I) Chronology: Developing dating technique for resolving critical controversies in Levantine archaeology and
Quaternary geology.
II) Geophysics: Exploring fine-scale geodynamo features in Earth’s core from new generations of global
geomagnetic models.
III) Cosmogenics: Correlating fast geomagnetic variations with cosmogenic isotope production rate.
IV) Climate: Testing one of the most challenging controversial questions in geomagnetism: “Does the Earth's
magnetic field play a role in climate changes?”
Summary
For decades the Holocene has been considered a flat and “boring” epoch from the standpoint of
paleomagnetism, mainly due to insufficient resolution of the available paleomagnetic data. However, recent
archaeomagnetic data have revealed that the Holocene geomagnetic field is anything but stable – presenting
puzzling intervals of extreme decadal-scale fluctuations and unexpected departures from a simple dipolar field
structure. This new information introduced an entirely new paradigm to the study of the geomagnetic field and
to a wide range of research areas relying on paleomagnetic data, such as geochronology, climate research, and
geodynamo exploration.
This proposal aims at breaking the resolution limits in paleomagnetism, and providing a continuous
time series of the geomagnetic field vector throughout the Holocene at decadal resolution and
unprecedented accuracy. To this end I will use an innovative assemblage of data sources, jointly unique to
the Levant, including rare archaeological finds, annual laminated stalagmites, varved sediments, and arid
playa deposits. Together, these sources can provide unprecedented yearly resolution, whereby the “absolute”
archaeomagnetic data can calibrate “relative” terrestrial data.
The geomagnetic data will define an innovative absolute geomagnetic chronology that will be used to
synchronize cosmogenic 10Be data and an extensive body of paleo-climatic indicators. With these in hand, I
will address four ground-breaking problems:
I) Chronology: Developing dating technique for resolving critical controversies in Levantine archaeology and
Quaternary geology.
II) Geophysics: Exploring fine-scale geodynamo features in Earth’s core from new generations of global
geomagnetic models.
III) Cosmogenics: Correlating fast geomagnetic variations with cosmogenic isotope production rate.
IV) Climate: Testing one of the most challenging controversial questions in geomagnetism: “Does the Earth's
magnetic field play a role in climate changes?”
Max ERC Funding
1 786 381 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym GV-FLU
Project A Genetic View of Influenza Infection
Researcher (PI) Irit Gat-Viks
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS2, ERC-2014-STG
Summary Inherited variation in the quantity and functionality of immune cells plays a key role in determining phenotypic diversity between individuals. Surprisingly little is known, however, about the specific contribution of immune cell subsets to variation in phenotypes such as susceptibility to infectious diseases and the underlying genetic variation. In many complex diseases, we currently have a poor understanding of the driver cell types that are responsible for inherited variation in disease states. A comprehensive mapping of quantities and functions of immune cell types during the course of disease, in large cohorts, bears the potential to transform genetic research; provides understanding of the genetic and immune basis of phenotypes; and reveals the key driver cell subsets.
Here I aim to derive a mechanistic understanding of how variation in quantity and function of immune cell subsets mediates inherited variation in disease states. I propose to develop a computational model that integrates predicted quantities and functions of cell subsets with genotypic and phenotypic information, leading to specific hypotheses on physiological regulation and the particular cell subsets that drive phenotypic diversity. To circumvent the technical difficulty in quantifying a large number of immune cell types, I will profile gene expression and computationally quantify changes in a large number of cell types. I will develop and apply this strategy to dissect Influenza infection in mice.
Since changes in immune responses play a key role in complex diseases, our ability to predict variation in immune responses from genotypes would have important clinical implications. This project has far reaching implications as the paradigm developed here will transform quantitative genetics studies as well as systems immunology research of complex disease. This approach will be applicable to any mammalian disease, allowing researchers to dissect their own systems at unprecedented detail.
Summary
Inherited variation in the quantity and functionality of immune cells plays a key role in determining phenotypic diversity between individuals. Surprisingly little is known, however, about the specific contribution of immune cell subsets to variation in phenotypes such as susceptibility to infectious diseases and the underlying genetic variation. In many complex diseases, we currently have a poor understanding of the driver cell types that are responsible for inherited variation in disease states. A comprehensive mapping of quantities and functions of immune cell types during the course of disease, in large cohorts, bears the potential to transform genetic research; provides understanding of the genetic and immune basis of phenotypes; and reveals the key driver cell subsets.
Here I aim to derive a mechanistic understanding of how variation in quantity and function of immune cell subsets mediates inherited variation in disease states. I propose to develop a computational model that integrates predicted quantities and functions of cell subsets with genotypic and phenotypic information, leading to specific hypotheses on physiological regulation and the particular cell subsets that drive phenotypic diversity. To circumvent the technical difficulty in quantifying a large number of immune cell types, I will profile gene expression and computationally quantify changes in a large number of cell types. I will develop and apply this strategy to dissect Influenza infection in mice.
Since changes in immune responses play a key role in complex diseases, our ability to predict variation in immune responses from genotypes would have important clinical implications. This project has far reaching implications as the paradigm developed here will transform quantitative genetics studies as well as systems immunology research of complex disease. This approach will be applicable to any mammalian disease, allowing researchers to dissect their own systems at unprecedented detail.
Max ERC Funding
1 497 000 €
Duration
Start date: 2015-07-01, End date: 2020-06-30
Project acronym HemTree2.0
Project Single cell genomic analysis and perturbations of hematopoietic progenitors: Towards a refined model of hematopoiesis
Researcher (PI) Ido AMIT
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS2, ERC-2016-COG
Summary Hematopoiesis is an important model for stem cell differentiation with great medical significance.
Heterogeneity within hematopoietic progenitor populations has considerably limited characterization and
molecular understanding of lineage commitment in both health and disease. Advances in single-cell genomic
technologies provide an extraordinary opportunity for unbiased and high resolution mapping of biological
function and regulation. Recently we have developed an experimental and analytical method, termed
massively parallel single-cell RNA-Seq (MARS-Seq), for unbiased classification of individual cells from
their native context and successfully applied it for characterization of immune and hematopoietic
progenitors.
Here, we propose to uncover the hierarchy and regulatory mechanisms controlling hematopoiesis by
combining comprehensive single-cell RNA-Seq analyses, modelling approaches, advanced functional assays,
single-cell CRISPR screens, knockout models and epigenetic profiling. Exciting preliminary result show that
indeed our approach is starting to uncover the complexity of hematopoietic progenitors and the regulatory
circuits driving hematopoietic decisions. We will pursue the following aims: (i) Generate a refined model of
hematopoiesis by comprehensive single-cell RNA-Seq profiling of hematopoietic progenitors, (ii) validate
the predicted model by in vivo functional developmental assays and then (iii) test candidate transcription and
chromatin factors uncovered by our model for their role in controlling progression towards various lineages
using single-cell measurements combined with CRISPR screens. Together, our study is expected to generate
a revised and high-resolution hematopoietic model and decipher the regulatory networks that control
hematopoiesis. Our methods and models may provide a platform for future medical advancements including
a large-scale European collaborative project to discover a comprehensive human hematopoietic tree.
Summary
Hematopoiesis is an important model for stem cell differentiation with great medical significance.
Heterogeneity within hematopoietic progenitor populations has considerably limited characterization and
molecular understanding of lineage commitment in both health and disease. Advances in single-cell genomic
technologies provide an extraordinary opportunity for unbiased and high resolution mapping of biological
function and regulation. Recently we have developed an experimental and analytical method, termed
massively parallel single-cell RNA-Seq (MARS-Seq), for unbiased classification of individual cells from
their native context and successfully applied it for characterization of immune and hematopoietic
progenitors.
Here, we propose to uncover the hierarchy and regulatory mechanisms controlling hematopoiesis by
combining comprehensive single-cell RNA-Seq analyses, modelling approaches, advanced functional assays,
single-cell CRISPR screens, knockout models and epigenetic profiling. Exciting preliminary result show that
indeed our approach is starting to uncover the complexity of hematopoietic progenitors and the regulatory
circuits driving hematopoietic decisions. We will pursue the following aims: (i) Generate a refined model of
hematopoiesis by comprehensive single-cell RNA-Seq profiling of hematopoietic progenitors, (ii) validate
the predicted model by in vivo functional developmental assays and then (iii) test candidate transcription and
chromatin factors uncovered by our model for their role in controlling progression towards various lineages
using single-cell measurements combined with CRISPR screens. Together, our study is expected to generate
a revised and high-resolution hematopoietic model and decipher the regulatory networks that control
hematopoiesis. Our methods and models may provide a platform for future medical advancements including
a large-scale European collaborative project to discover a comprehensive human hematopoietic tree.
Max ERC Funding
2 000 000 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym HUCNC
Project Conserved Non-Coding Sequences; function, variability and phenotypic consequences
Researcher (PI) Stylianos Antonarakis
Host Institution (HI) UNIVERSITE DE GENEVE
Call Details Advanced Grant (AdG), LS2, ERC-2009-AdG
Summary Comparative genomics revealed that ~5% of the human genome is conserved among mammals. This fraction is likely functional, and could harbor pathogenic mutations. We have shown (Nature 2002, Science 2003) that more than half of the constrained fraction of the genome consists of Conserved Non-Coding sequences (CNCs). Model organisms provided evidence for enhancer activity for a fraction of CNCs; in addition another fraction is part of large non-coding RNAs (lincRNA). However, the function of the majority of CNCs is unknown. Importantly, a few pathogenic mutations in CNCs have been associated with genetic disorders. We propose to i) perform functional analysis of CNCs, and ii) identify the spectrum of pathogenic CNC mutations in recognizable human phenotypes. The aims are: 1. Functional genomic connectivity of CNCs 1a. Use 4C in CNCs in various cell types and determine their physical genomic interactions. 1b. Perform targeted disruption of CNCs in cells and assess the functional outcomes. 2. Pathogenic variation of CNCs 2a. Assess the common variation in CNCs: i) common deletion/insertions in 350 samples by aCGH of all human CNCs; ii) common SNP/small indels using DNA selection and High Throughput Sequencing (HTS) of CNCs in 100 samples. 2b. Identify likely pathogenic mutations in developmental syndromes. Search for i) large deletions and duplications of CNCs using aCGH in 1500 samples with malformation syndromes, 1000 from spontaneous abortions, and 500 with X-linked mental retardation; and ii) point mutations in these samples by targeted HTS. The distinction between pathogenic and non-pathogenic variants is difficult, and we propose approaches to meet the challenge. 3. Genetic control (cis and trans eQTLs) of expression variation of CNC lincRNAs, using 200 samples.
Summary
Comparative genomics revealed that ~5% of the human genome is conserved among mammals. This fraction is likely functional, and could harbor pathogenic mutations. We have shown (Nature 2002, Science 2003) that more than half of the constrained fraction of the genome consists of Conserved Non-Coding sequences (CNCs). Model organisms provided evidence for enhancer activity for a fraction of CNCs; in addition another fraction is part of large non-coding RNAs (lincRNA). However, the function of the majority of CNCs is unknown. Importantly, a few pathogenic mutations in CNCs have been associated with genetic disorders. We propose to i) perform functional analysis of CNCs, and ii) identify the spectrum of pathogenic CNC mutations in recognizable human phenotypes. The aims are: 1. Functional genomic connectivity of CNCs 1a. Use 4C in CNCs in various cell types and determine their physical genomic interactions. 1b. Perform targeted disruption of CNCs in cells and assess the functional outcomes. 2. Pathogenic variation of CNCs 2a. Assess the common variation in CNCs: i) common deletion/insertions in 350 samples by aCGH of all human CNCs; ii) common SNP/small indels using DNA selection and High Throughput Sequencing (HTS) of CNCs in 100 samples. 2b. Identify likely pathogenic mutations in developmental syndromes. Search for i) large deletions and duplications of CNCs using aCGH in 1500 samples with malformation syndromes, 1000 from spontaneous abortions, and 500 with X-linked mental retardation; and ii) point mutations in these samples by targeted HTS. The distinction between pathogenic and non-pathogenic variants is difficult, and we propose approaches to meet the challenge. 3. Genetic control (cis and trans eQTLs) of expression variation of CNC lincRNAs, using 200 samples.
Max ERC Funding
2 353 920 €
Duration
Start date: 2010-07-01, End date: 2015-06-30
Project acronym HYDROCARB
Project Hydrogen isotopes in plant-derived organic compounds as new tool to identify changes in the carbon metabolism of plants and ecosystems during the anthropocene
Researcher (PI) Ansgar KAHMEN
Host Institution (HI) UNIVERSITAT BASEL
Call Details Consolidator Grant (CoG), PE10, ERC-2016-COG
Summary HYDROCARB is motivated by the enormous potential that stable hydrogen isotope ratios (δ2H values) in plant compounds have as hydrological proxy, but in particular as new proxy for the carbon metabolism in plants. Current conceptual models suggest that δ2H values in plant organic compounds are composed of (i) hydrological and (ii) metabolic signals. The hydrological information that is contained in δ2H values of plant material is now well understood and is often applied in (paleo-) hydrological research. In contrast, the metabolic information that is contained in plant δ2H values is mostly unknown. Intriguing recent research suggests, however, that metabolic signals in the δ2H values of plant organic compounds reflect the balance of autotrophic and heterotrophic processes in plants. This suggests that exciting and previously unknown opportunities exist to exploit δ2H values in plant compounds for information on the carbohydrate metabolism of plants, which would be relevant for a broad range of biological and biogeochemical disciplines.
The goal of HYDROCARB is to perform the experimental work that is now needed to identify the key biochemical and physiological processes that determine the metabolic information that is recorded in the δ2H values of plant organic compounds such as leaf wax lipids, lignin and cellulose. With this HYDROCARB will provide the basis for semi-mechanistic models that will allow (i) disentangling hydrological from metabolic signals in plant δ2H values and (ii) identifying the precise physiological processes with respect to a plants carbohydrate metabolism that can be deducted from the δ2H values of different plant compounds. If successful, HYDROCARB will establish with this research δ2H values in plant organic compounds as a powerful new proxy that will allow ground-breaking and innovative research on plant and ecosystem carbon cycling, which has implications for plant biology, biogeochemistry and the earth system sciences.
Summary
HYDROCARB is motivated by the enormous potential that stable hydrogen isotope ratios (δ2H values) in plant compounds have as hydrological proxy, but in particular as new proxy for the carbon metabolism in plants. Current conceptual models suggest that δ2H values in plant organic compounds are composed of (i) hydrological and (ii) metabolic signals. The hydrological information that is contained in δ2H values of plant material is now well understood and is often applied in (paleo-) hydrological research. In contrast, the metabolic information that is contained in plant δ2H values is mostly unknown. Intriguing recent research suggests, however, that metabolic signals in the δ2H values of plant organic compounds reflect the balance of autotrophic and heterotrophic processes in plants. This suggests that exciting and previously unknown opportunities exist to exploit δ2H values in plant compounds for information on the carbohydrate metabolism of plants, which would be relevant for a broad range of biological and biogeochemical disciplines.
The goal of HYDROCARB is to perform the experimental work that is now needed to identify the key biochemical and physiological processes that determine the metabolic information that is recorded in the δ2H values of plant organic compounds such as leaf wax lipids, lignin and cellulose. With this HYDROCARB will provide the basis for semi-mechanistic models that will allow (i) disentangling hydrological from metabolic signals in plant δ2H values and (ii) identifying the precise physiological processes with respect to a plants carbohydrate metabolism that can be deducted from the δ2H values of different plant compounds. If successful, HYDROCARB will establish with this research δ2H values in plant organic compounds as a powerful new proxy that will allow ground-breaking and innovative research on plant and ecosystem carbon cycling, which has implications for plant biology, biogeochemistry and the earth system sciences.
Max ERC Funding
1 999 941 €
Duration
Start date: 2017-11-01, End date: 2022-10-31
Project acronym INTEXseas
Project An integrated weather-system perspective on the characteristics, dynamics and impacts of extreme seasons
Researcher (PI) Johann Heinrich WERNLI
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), PE10, ERC-2017-ADG
Summary Single extreme weather events can be hazardous, but for certain socioeconomic sectors the seasonal aggregation of weather is particularly harmful. Extremes on timescales up to two weeks are typically related to specific weather systems, but no such link exists for extreme seasons. Therefore, they are very difficult to meteorologically understand, despite their utmost societal relevance. This project aims at filling this gap, providing a multi-faceted analysis of different types of extreme seasons in a changing climate. Very large ensembles of climate simulations serve to investigate the characteristics and dynamics of the, e.g., hottest and coldest, and wettest and driest, season in regions worldwide. The extreme season characteristics include their spatial scale and their extremeness given the entire distribution of seasonal values in this region. Their dynamics is related to the fundamental understanding of the sequence of weather events that makes a season extreme: is it a single, highly unusual weather event that renders a season the most extreme (e.g., an unprecedented heat wave) or rather an unusual frequency of well-known weather systems (e.g., a series of strongly precipitating cyclones). These paradigms, referred to as “something new” vs. “more of the same”, are particularly relevant when considering extreme seasons in a warming climate. This project will combine state-of-the-art climate modelling, a unique set of weather-system diagnostics informed by profound dynamical understanding, and novel impact assessment pathways to address three main hypotheses: 1) different types of extreme seasons differ in terms of their spatial scale and relation to weather systems; 2) for specific types of extreme seasons, future climate simulations indicate a marked increase of extremeness; and 3) for certain socioeconomic sectors, the consequences of the future modulation of extreme seasons is more severe than inferred from climate change trend considerations alone.
Summary
Single extreme weather events can be hazardous, but for certain socioeconomic sectors the seasonal aggregation of weather is particularly harmful. Extremes on timescales up to two weeks are typically related to specific weather systems, but no such link exists for extreme seasons. Therefore, they are very difficult to meteorologically understand, despite their utmost societal relevance. This project aims at filling this gap, providing a multi-faceted analysis of different types of extreme seasons in a changing climate. Very large ensembles of climate simulations serve to investigate the characteristics and dynamics of the, e.g., hottest and coldest, and wettest and driest, season in regions worldwide. The extreme season characteristics include their spatial scale and their extremeness given the entire distribution of seasonal values in this region. Their dynamics is related to the fundamental understanding of the sequence of weather events that makes a season extreme: is it a single, highly unusual weather event that renders a season the most extreme (e.g., an unprecedented heat wave) or rather an unusual frequency of well-known weather systems (e.g., a series of strongly precipitating cyclones). These paradigms, referred to as “something new” vs. “more of the same”, are particularly relevant when considering extreme seasons in a warming climate. This project will combine state-of-the-art climate modelling, a unique set of weather-system diagnostics informed by profound dynamical understanding, and novel impact assessment pathways to address three main hypotheses: 1) different types of extreme seasons differ in terms of their spatial scale and relation to weather systems; 2) for specific types of extreme seasons, future climate simulations indicate a marked increase of extremeness; and 3) for certain socioeconomic sectors, the consequences of the future modulation of extreme seasons is more severe than inferred from climate change trend considerations alone.
Max ERC Funding
2 500 000 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym IRMIDYN
Project Iron mineral dynamics in redox-affected soils and sediments: Pushing the frontier toward in-situ studies
Researcher (PI) Ruben KRETZSCHMAR
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), PE10, ERC-2017-ADG
Summary IRMIDYN will study the dynamics of redox-driven iron mineral transformation processes in soils and sediments and impacts on nutrient and trace element behavior using a novel approach based on enriched stable isotopes (e.g., 57Fe, 33S, 67Zn, 113Cd, 198Hg) in combination with innovative experiments and cutting-edge analytical techniques, most importantly 57Fe Mössbauer and Raman micro-spectroscopy and imaging. The thermodynamic stability and occurrence of iron minerals in sufficiently stable Earth surface environments is fairly well understood and supported by field observations. However, the kinetics of iron mineral recrystallization and transformation processes under rapidly changing redox conditions is far less understood, and has to date mostly been studied in in mixed reactors with pure minerals or sediment slurries, but rarely in-situ in complex soils and sediments. Thus, we do not know if and how fast certain iron mineral recrystallization and transformation processes observed in the laboratory actually occur in soils and sediments, and which environmental factors control the transformation rates and products. Redox-driven iron mineral recrystallization and transformation processes are key to understanding the biogeochemical cycles of C, N, P, S, and many trace elements (e.g., As, Zn, Cd, Hg, U). In face of current global challenges caused by massive anthropogenic changes in biogeochemical cycles of nutrients and toxic elements, it is paramount that we begin to understand and quantify the dynamics of these processes in-situ and learn how we can apply our mechanistic (but often reductionist) knowledge to the natural environment. This project will take a large step toward a better understanding of iron mineral dynamics in redox-affected Earth surface environments, with wide implications in biogeochemistry and other fields including environmental engineering, corrosion sciences, archaeology and cultural heritage sciences, and planetary sciences.
Summary
IRMIDYN will study the dynamics of redox-driven iron mineral transformation processes in soils and sediments and impacts on nutrient and trace element behavior using a novel approach based on enriched stable isotopes (e.g., 57Fe, 33S, 67Zn, 113Cd, 198Hg) in combination with innovative experiments and cutting-edge analytical techniques, most importantly 57Fe Mössbauer and Raman micro-spectroscopy and imaging. The thermodynamic stability and occurrence of iron minerals in sufficiently stable Earth surface environments is fairly well understood and supported by field observations. However, the kinetics of iron mineral recrystallization and transformation processes under rapidly changing redox conditions is far less understood, and has to date mostly been studied in in mixed reactors with pure minerals or sediment slurries, but rarely in-situ in complex soils and sediments. Thus, we do not know if and how fast certain iron mineral recrystallization and transformation processes observed in the laboratory actually occur in soils and sediments, and which environmental factors control the transformation rates and products. Redox-driven iron mineral recrystallization and transformation processes are key to understanding the biogeochemical cycles of C, N, P, S, and many trace elements (e.g., As, Zn, Cd, Hg, U). In face of current global challenges caused by massive anthropogenic changes in biogeochemical cycles of nutrients and toxic elements, it is paramount that we begin to understand and quantify the dynamics of these processes in-situ and learn how we can apply our mechanistic (but often reductionist) knowledge to the natural environment. This project will take a large step toward a better understanding of iron mineral dynamics in redox-affected Earth surface environments, with wide implications in biogeochemistry and other fields including environmental engineering, corrosion sciences, archaeology and cultural heritage sciences, and planetary sciences.
Max ERC Funding
3 154 658 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym lincSAFARI
Project Sequence and Function Relationships in Long Intervening Noncoding RNAs
Researcher (PI) Igor Ulitsky
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2014-STG
Summary It is now clear that many intergenic regions in eukaryotic genomes give rise to a range of processed and regulated transcripts that do not appear to code for functional proteins. A subset of these are long (>200 nt), capped and polyadenylated RNAs transcribed by RNA polymerase II and collectively called long intervening noncoding RNAs or lincRNAs. The recent estimates are that the human genome encodes >10,000 distinct lincRNAs, many of which show tissue-specific expression and are frequently dysregulated in human disease, including neurodegeneration.
Given the growing number of lincRNAs implicated in human disease or required for proper development, fundamental questions that need to be addressed are: Which lincRNAs are functional? How is functional information encoded in the lincRNA sequence? Is this information interpreted in the context of the mature or the nascent RNA? What are the identities and functional roles of specific sequence domains within lincRNA genes?
Our main hypothesis is that many lincRNA loci play key roles in gene regulation during cell differentiation, both via functionally important transcription events and post-transcriptionally, through the combined action of multiple short sequence domains. We will test this hypothesis using three complementary approaches – comparative genomics, detailed perturbations in mammalian cells followed by quantitative molecular phenotyping, and high-throughput screens for sequences able to carry out specific functions.
We propose an interdisciplinary approach combining computational, molecular and stem cell biology. Our methodology will be scalable, allowing us to tackle completely uncharacterized long RNAs and eventually zoom in and study their individual bases. Upon successful accomplishment of the program, we will delineate modes of action of numerous lincRNAs, report sequence patches that are functionally important and understand how specific bases and structures act in concert to drive lincRNA function.
Summary
It is now clear that many intergenic regions in eukaryotic genomes give rise to a range of processed and regulated transcripts that do not appear to code for functional proteins. A subset of these are long (>200 nt), capped and polyadenylated RNAs transcribed by RNA polymerase II and collectively called long intervening noncoding RNAs or lincRNAs. The recent estimates are that the human genome encodes >10,000 distinct lincRNAs, many of which show tissue-specific expression and are frequently dysregulated in human disease, including neurodegeneration.
Given the growing number of lincRNAs implicated in human disease or required for proper development, fundamental questions that need to be addressed are: Which lincRNAs are functional? How is functional information encoded in the lincRNA sequence? Is this information interpreted in the context of the mature or the nascent RNA? What are the identities and functional roles of specific sequence domains within lincRNA genes?
Our main hypothesis is that many lincRNA loci play key roles in gene regulation during cell differentiation, both via functionally important transcription events and post-transcriptionally, through the combined action of multiple short sequence domains. We will test this hypothesis using three complementary approaches – comparative genomics, detailed perturbations in mammalian cells followed by quantitative molecular phenotyping, and high-throughput screens for sequences able to carry out specific functions.
We propose an interdisciplinary approach combining computational, molecular and stem cell biology. Our methodology will be scalable, allowing us to tackle completely uncharacterized long RNAs and eventually zoom in and study their individual bases. Upon successful accomplishment of the program, we will delineate modes of action of numerous lincRNAs, report sequence patches that are functionally important and understand how specific bases and structures act in concert to drive lincRNA function.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-06-01, End date: 2020-05-31
Project acronym LineageDiscovery
Project Laying the Biological, Computational and Architectural Foundations for Human Cell Lineage Discovery
Researcher (PI) Ehud Shapiro
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS2, ERC-2014-ADG
Summary Within a decade, advances in single-cell genomics would allow humanity to embark on a coordinated international effort to discover the human cell lineage tree. The goal of LineageDiscovery is to lay the biological, computational and architectural foundations for this envisioned project and demonstrate its feasibility and value.
An organismal cell lineage tree is a rooted, labelled binary tree where nodes represent organism cells, edges represent progeny relations and labels capture cell state. The tree of an adult human has about 100 trillion nodes. Many fundamental open questions in biology and medicine are about the structure, dynamics and variance of the human cell lineage tree in development, health, ageing and disease. E.g., which cancer cells give rise to metastases? Do beta cells renew? Which progeny do brain stem cells produce in development, maintenance and ageing?
LineageDiscovery is based on a decade of research on this challenge by Shapiro’s lab and others. It will develop an efficient biological-computational cell lineage discovery workflow that starts with sampled cells and ends with knowledge of their cell lineage tree; and a scalable architecture for the collaborative development and the distributed deployment of this workflow. The workflow will be based on emerging single-cell technologies and will include novel algorithms to analyse single-cell data, to reconstruct cell lineage trees, and to infer ancestral cell type and state dynamics. A programmable meta-system will be developed and used for workflow optimization and evaluation. The workflow and architecture will be deployed and tested in a broad range of proof-of-concept human cell lineage discovery experiments with self-funded collaborators.
Successful execution of this research plan coupled with expected advances in single-cell genomics would establish both the feasibility and the value of the envisioned large-scale human cell lineage discovery project, ideally leading to its launch.
Summary
Within a decade, advances in single-cell genomics would allow humanity to embark on a coordinated international effort to discover the human cell lineage tree. The goal of LineageDiscovery is to lay the biological, computational and architectural foundations for this envisioned project and demonstrate its feasibility and value.
An organismal cell lineage tree is a rooted, labelled binary tree where nodes represent organism cells, edges represent progeny relations and labels capture cell state. The tree of an adult human has about 100 trillion nodes. Many fundamental open questions in biology and medicine are about the structure, dynamics and variance of the human cell lineage tree in development, health, ageing and disease. E.g., which cancer cells give rise to metastases? Do beta cells renew? Which progeny do brain stem cells produce in development, maintenance and ageing?
LineageDiscovery is based on a decade of research on this challenge by Shapiro’s lab and others. It will develop an efficient biological-computational cell lineage discovery workflow that starts with sampled cells and ends with knowledge of their cell lineage tree; and a scalable architecture for the collaborative development and the distributed deployment of this workflow. The workflow will be based on emerging single-cell technologies and will include novel algorithms to analyse single-cell data, to reconstruct cell lineage trees, and to infer ancestral cell type and state dynamics. A programmable meta-system will be developed and used for workflow optimization and evaluation. The workflow and architecture will be deployed and tested in a broad range of proof-of-concept human cell lineage discovery experiments with self-funded collaborators.
Successful execution of this research plan coupled with expected advances in single-cell genomics would establish both the feasibility and the value of the envisioned large-scale human cell lineage discovery project, ideally leading to its launch.
Max ERC Funding
2 250 000 €
Duration
Start date: 2015-09-01, End date: 2020-07-31
Project acronym LIVHET
Project Single cell heterogeneity in the mammalian liver
Researcher (PI) Shaul Shalev Itzkovitz
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2013-StG
Summary The mammalian liver performs critical functions for maintaining metabolic homeostasis. It regulates the body’s glucose and lipid stores, detoxifies blood, and produces bile among a host of other functions. The liver achieves this diversity through the collective behaviour of heterogeneous hepatocytes operating in highly structured microenvironments. Understanding the design principles of the liver is an open challenge requiring analysis of single cells within the intact tissue.
Liver heterogeneity appears at two length scales. At the liver lobule level centripetal blood flow creates gradients of oxygen, nutrients and hormones. The consumption of hepatocytes along the lobule axis determines the inputs available for more centrally located hepatocytes. The resulting spatial division of labor, termed ‘liver zonation’ could enable optimal tissue function in face of these long-range constraints. At the cellular level most hepatocytes are polyploid cells, having either one or two nuclei and a corresponding variability in cell sizes. The functional advantage of liver polyploidy remains unclear.
In this proposal we aim to combine single molecule transcript imaging in the intact liver with theory from systems biology to uncover the design principles of liver heterogeneity. We will examine the hypothesis that spatial zonation and hepatocyte polyploidy evolved to enable the liver to optimally operate. We will characterize the spatial co-expression patterns of key liver genes and theoretically compare the ability of these patterns to excel over alternative patterns. We will also characterize the differential resource allocation of hepatocytes of different ploidy classes.
This interdisciplinary project stands at the forefront of research in mammalian biology, addressing fundamental properties of a major organ at unprecedented single-cell resolution. It will open new avenues for extending the field of systems biology to the analysis of complex tissues in mammalian organisms.
Summary
The mammalian liver performs critical functions for maintaining metabolic homeostasis. It regulates the body’s glucose and lipid stores, detoxifies blood, and produces bile among a host of other functions. The liver achieves this diversity through the collective behaviour of heterogeneous hepatocytes operating in highly structured microenvironments. Understanding the design principles of the liver is an open challenge requiring analysis of single cells within the intact tissue.
Liver heterogeneity appears at two length scales. At the liver lobule level centripetal blood flow creates gradients of oxygen, nutrients and hormones. The consumption of hepatocytes along the lobule axis determines the inputs available for more centrally located hepatocytes. The resulting spatial division of labor, termed ‘liver zonation’ could enable optimal tissue function in face of these long-range constraints. At the cellular level most hepatocytes are polyploid cells, having either one or two nuclei and a corresponding variability in cell sizes. The functional advantage of liver polyploidy remains unclear.
In this proposal we aim to combine single molecule transcript imaging in the intact liver with theory from systems biology to uncover the design principles of liver heterogeneity. We will examine the hypothesis that spatial zonation and hepatocyte polyploidy evolved to enable the liver to optimally operate. We will characterize the spatial co-expression patterns of key liver genes and theoretically compare the ability of these patterns to excel over alternative patterns. We will also characterize the differential resource allocation of hepatocytes of different ploidy classes.
This interdisciplinary project stands at the forefront of research in mammalian biology, addressing fundamental properties of a major organ at unprecedented single-cell resolution. It will open new avenues for extending the field of systems biology to the analysis of complex tissues in mammalian organisms.
Max ERC Funding
1 500 000 €
Duration
Start date: 2013-11-01, End date: 2018-10-31
Project acronym LSO
Project Liver Spatial Omics
Researcher (PI) Shaul Shalev ITZKOVITZ
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS2, ERC-2017-COG
Summary The mammalian liver is a heterogeneous, yet highly structured organ, which performs diverse functions to maintain organismal homeostasis. Hepatocytes operate in repeating hexagonally shaped units termed lobules that are polarized by centripetal blood flow and morphogens. This polarized microenvironment facilitates optimal function by localizing specific processes to distinct lobule layers, a phenomenon known as ‘liver zonation’. While zonation of some key liver functions has been known for years, using spatially resolved single cell transcriptomics, we recently discovered that about 50% of liver genes are zonated. This surprisingly broad spatial heterogeneity raises a fundamental question - do hepatocytes form a uniform population that differs due to spatially graded inputs or are hepatocytes at different zones in fact distinct cell types?
In this proposal we will tackle this question by developing techniques for sorting massive amounts of hepatocytes from defined tissue coordinates at high spatial resolution using zonated surface markers, new zonated reporter mouse models and mRNA content. We will perform a deep and comprehensive profiling of the hepatocyte genome, methylome, epigenome, transcriptome, proteome and metabolome at each zone to characterize liver zonation at all relevant cellular scales. We will also develop an ex-vivo system to functionally characterize the response of hepatocytes from distinct zones to identical input stimuli and the ability of hepatocytes to inter-convert to hepatocytes with differing zonal identities. These experiments will be performed in different metabolic states and along a high fat diet. This project will uncover new features of liver zonation in health and disease and redefine the hepatocyte cell state. Our approach for spatially refined tissue omics can be extended to other structured mammalian organs, thus opening new avenues of research in Systems Biology of mammalian tissues.
Summary
The mammalian liver is a heterogeneous, yet highly structured organ, which performs diverse functions to maintain organismal homeostasis. Hepatocytes operate in repeating hexagonally shaped units termed lobules that are polarized by centripetal blood flow and morphogens. This polarized microenvironment facilitates optimal function by localizing specific processes to distinct lobule layers, a phenomenon known as ‘liver zonation’. While zonation of some key liver functions has been known for years, using spatially resolved single cell transcriptomics, we recently discovered that about 50% of liver genes are zonated. This surprisingly broad spatial heterogeneity raises a fundamental question - do hepatocytes form a uniform population that differs due to spatially graded inputs or are hepatocytes at different zones in fact distinct cell types?
In this proposal we will tackle this question by developing techniques for sorting massive amounts of hepatocytes from defined tissue coordinates at high spatial resolution using zonated surface markers, new zonated reporter mouse models and mRNA content. We will perform a deep and comprehensive profiling of the hepatocyte genome, methylome, epigenome, transcriptome, proteome and metabolome at each zone to characterize liver zonation at all relevant cellular scales. We will also develop an ex-vivo system to functionally characterize the response of hepatocytes from distinct zones to identical input stimuli and the ability of hepatocytes to inter-convert to hepatocytes with differing zonal identities. These experiments will be performed in different metabolic states and along a high fat diet. This project will uncover new features of liver zonation in health and disease and redefine the hepatocyte cell state. Our approach for spatially refined tissue omics can be extended to other structured mammalian organs, thus opening new avenues of research in Systems Biology of mammalian tissues.
Max ERC Funding
2 000 000 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym m1ARNA
Project Transcriptomic m1A - a new key player in the epitranscriptome arena
Researcher (PI) Gideon RECHAVI
Host Institution (HI) MEDICAL RESEARCH INFRASTRUCTURE DEVELOPMENT AND HEALTH SERVICES FUND BY THE SHEBA MEDICAL CENTER
Call Details Advanced Grant (AdG), LS2, ERC-2016-ADG
Summary Reversible epigenetic modifications regulate gene expression to define cell fate and response to environmental stimuli. Gene expression tuning by DNA and chromatin modifications is well studied, yet the effect of RNA modifications on gene expression is only starting to be revealed. More than a hundred chemical modifications decorate RNAs, mainly non-coding ones, expanding their nucleotide vocabulary and mediating their diverse functions. Several modifications were globally mapped in mRNA. Only two, N6-methyladenosine (m6A) and N1-methyladenosine (m1A) exhibit a distinct topology alluding to a functional role. We pioneered the identification of m6A that is located preferentially in distinct transcript landmarks, mostly around stop codons and mediates transcript localization, splicing, decay and translation. We now identified m1A which decorates thousands of genes mainly in the start codon vicinity, upstream to the first splice site. Our preliminary results indicate that m1A dynamically responds to environmental stimuli and plays a central role in translation regulation. The regulation and functions of m1A are still terra incognita. Our objectives are to identify m1A writers and erasers, elucidate m1A readers and the mechanisms whereby m1A dictates downstream outcomes, particularly translation regulation. We will study m1A functions in response to physiologic stimuli and stress conditions in cells and animal models by manipulation of the m1A deposition machinery. As epigenetic marks operate in a context-dependent concerted way we will map m1A marks concomitantly with m6A to decipher their interplay in regulating gene expression via a putative “epigenetic RNA code”. The data obtained from parallel mapping of m1A and m6A at a single nucleotide and a single transcript resolution, will expose the interplay between these two mRNA modifications in the context of multilayer epigenetics. The study of m1A circuits may identify targets amenable to therapeutic manipulations.
Summary
Reversible epigenetic modifications regulate gene expression to define cell fate and response to environmental stimuli. Gene expression tuning by DNA and chromatin modifications is well studied, yet the effect of RNA modifications on gene expression is only starting to be revealed. More than a hundred chemical modifications decorate RNAs, mainly non-coding ones, expanding their nucleotide vocabulary and mediating their diverse functions. Several modifications were globally mapped in mRNA. Only two, N6-methyladenosine (m6A) and N1-methyladenosine (m1A) exhibit a distinct topology alluding to a functional role. We pioneered the identification of m6A that is located preferentially in distinct transcript landmarks, mostly around stop codons and mediates transcript localization, splicing, decay and translation. We now identified m1A which decorates thousands of genes mainly in the start codon vicinity, upstream to the first splice site. Our preliminary results indicate that m1A dynamically responds to environmental stimuli and plays a central role in translation regulation. The regulation and functions of m1A are still terra incognita. Our objectives are to identify m1A writers and erasers, elucidate m1A readers and the mechanisms whereby m1A dictates downstream outcomes, particularly translation regulation. We will study m1A functions in response to physiologic stimuli and stress conditions in cells and animal models by manipulation of the m1A deposition machinery. As epigenetic marks operate in a context-dependent concerted way we will map m1A marks concomitantly with m6A to decipher their interplay in regulating gene expression via a putative “epigenetic RNA code”. The data obtained from parallel mapping of m1A and m6A at a single nucleotide and a single transcript resolution, will expose the interplay between these two mRNA modifications in the context of multilayer epigenetics. The study of m1A circuits may identify targets amenable to therapeutic manipulations.
Max ERC Funding
2 457 500 €
Duration
Start date: 2017-07-01, End date: 2022-06-30
Project acronym MADE-IN-EARTH
Project Interplay between metamorphism and deformation in the Earth’s lithosphere
Researcher (PI) Lucie Tajcmanova
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Starting Grant (StG), PE10, ERC-2013-StG
Summary "A key to understanding the processes operating in the outer part of the Earth is to look at the metamorphic rocks produced in orogenic belts. These rocks now exhumed to the Earth’s surface provide a record of what they experienced, if only they can be correctly interpreted.
The recent use of high resolution devices has revealed the three-dimensional size, shape, composition and distribution of microstructural features in metamorphic rocks down to the nanometre-scale. The new observations show that mechanically maintained pressure variations can be significant (~1 GPa) even on a micro-scale. However, there is currently no satisfactory thermodynamic methodology for a quantitative interpretation of systems with such pressure variations in metamorphic rocks. Ignoring such pressure variations in petrological analysis can lead to errors in depth estimates that are comparable to the typical thickness of the whole continental crust. Such an error may then significantly influence the quality of geodynamic reconstructions.
Here, I propose to develop a revolutionary theoretical and computational method to understand microstructures that reflect pressure variations, based on the chemical and mechanical properties of their constituent minerals. Using the novel theoretical approach, I and my team will perform 3D numerical simulations and give the criteria to correctly understand the key microstructures.
This emerging multi-disciplinary research will provide a quantitative and physically-based framework for interpreting common microstructures in metamorphic rocks. Furthermore, the new approach will not only make a critical contribution to understanding the interplay between metamorphic processes and deformation on the grain scale, but it will also form the basis for a new generation of models for application to large-scale geological scenarios. The results of the project will thus significantly increase our understanding of key processes in the Earth’s lithosphere."
Summary
"A key to understanding the processes operating in the outer part of the Earth is to look at the metamorphic rocks produced in orogenic belts. These rocks now exhumed to the Earth’s surface provide a record of what they experienced, if only they can be correctly interpreted.
The recent use of high resolution devices has revealed the three-dimensional size, shape, composition and distribution of microstructural features in metamorphic rocks down to the nanometre-scale. The new observations show that mechanically maintained pressure variations can be significant (~1 GPa) even on a micro-scale. However, there is currently no satisfactory thermodynamic methodology for a quantitative interpretation of systems with such pressure variations in metamorphic rocks. Ignoring such pressure variations in petrological analysis can lead to errors in depth estimates that are comparable to the typical thickness of the whole continental crust. Such an error may then significantly influence the quality of geodynamic reconstructions.
Here, I propose to develop a revolutionary theoretical and computational method to understand microstructures that reflect pressure variations, based on the chemical and mechanical properties of their constituent minerals. Using the novel theoretical approach, I and my team will perform 3D numerical simulations and give the criteria to correctly understand the key microstructures.
This emerging multi-disciplinary research will provide a quantitative and physically-based framework for interpreting common microstructures in metamorphic rocks. Furthermore, the new approach will not only make a critical contribution to understanding the interplay between metamorphic processes and deformation on the grain scale, but it will also form the basis for a new generation of models for application to large-scale geological scenarios. The results of the project will thus significantly increase our understanding of key processes in the Earth’s lithosphere."
Max ERC Funding
1 499 820 €
Duration
Start date: 2013-10-01, End date: 2018-09-30
Project acronym MATRICS
Project Modern Approaches to Temperature Reconstructions in polar Ice Cores
Researcher (PI) Hubertus Fischer
Host Institution (HI) UNIVERSITAET BERN
Call Details Advanced Grant (AdG), PE10, ERC-2008-AdG
Summary The recent anthropogenic global warming makes a detailed knowledge of variations in the Earth climate system and of the coupling processes between climate and biogeochemical cycles of pressing importance. Studies of climate changes in the past represent a vital part of climate change research which is essential to assess the current warming against the background of natural climate variability. Due to strong limitations in direct observations, climate reconstructions for the past can only be achieved using natural climate archives. The paleoclimatic archive in ice cores provides not only information on climate variability over many thousands of years in high resolution but also on greenhouse gases, aerosol concentrations and more. Crucial questions on climate variability on interannual to orbital time scales and on the coupling processes and teleconnections in the climate system remain still open. To answer these questions novel climate parameters on polar ice cores are needed that go beyond previous studies in terms of temporal resolution, spatial coverage as well as quantitative representativeness. This proposal intends to develop such methods based on latest advances in analytical techniques and to apply them to polar ice cores. The common theme of the new approaches within MATRICs is the reconstruction of new, quantitative temperature information from different regions of the Earth all on the same core avoiding crucial crossdating issues. This comprises (i) continuous quantitative reconstructions of local temperature changes on polar ice sheets in seasonal resolution using new approaches, (ii) estimates of climate changes in continental, not permanently ice covered regions based on concurrent changes in the methane cycle and (iii) a new physical ice core gas thermometer for mean global ocean temperature. Successful implementation of the studies in MATRICs will make a significant contribution to maintain the world leading position of European ice core science.
Summary
The recent anthropogenic global warming makes a detailed knowledge of variations in the Earth climate system and of the coupling processes between climate and biogeochemical cycles of pressing importance. Studies of climate changes in the past represent a vital part of climate change research which is essential to assess the current warming against the background of natural climate variability. Due to strong limitations in direct observations, climate reconstructions for the past can only be achieved using natural climate archives. The paleoclimatic archive in ice cores provides not only information on climate variability over many thousands of years in high resolution but also on greenhouse gases, aerosol concentrations and more. Crucial questions on climate variability on interannual to orbital time scales and on the coupling processes and teleconnections in the climate system remain still open. To answer these questions novel climate parameters on polar ice cores are needed that go beyond previous studies in terms of temporal resolution, spatial coverage as well as quantitative representativeness. This proposal intends to develop such methods based on latest advances in analytical techniques and to apply them to polar ice cores. The common theme of the new approaches within MATRICs is the reconstruction of new, quantitative temperature information from different regions of the Earth all on the same core avoiding crucial crossdating issues. This comprises (i) continuous quantitative reconstructions of local temperature changes on polar ice sheets in seasonal resolution using new approaches, (ii) estimates of climate changes in continental, not permanently ice covered regions based on concurrent changes in the methane cycle and (iii) a new physical ice core gas thermometer for mean global ocean temperature. Successful implementation of the studies in MATRICs will make a significant contribution to maintain the world leading position of European ice core science.
Max ERC Funding
2 100 000 €
Duration
Start date: 2009-01-01, End date: 2014-12-31
Project acronym MATRIX
Project MAchine for Time Reversal and Immersive wave eXperimentation
Researcher (PI) Johan Olof Anders ROBERTSSON
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), PE10, ERC-2015-AdG
Summary There is a need for a radically new laboratory experimental approach for studying the interaction of seismic waves with the complex media of the Earth’s subsurface. We present here a fundamental new approach to seismic wave experimentation that involves fully immersing a physical seismic experiment within a virtual numerical environment. This enormously challenging endeavour, which is relevant to many outstanding issues in seismology, has not been previously attempted. By continuously varying the output of numerous transponders closely spaced around the physical domain using a control algorithm that takes advantage of measurements made by a scanning Laser-Doppler Vibrometer and a novel theory of exact boundary conditions, waves travelling between the physical and numerical domains will seamlessly propagate back and forth between the two domains without being affected by reflections at the boundaries between the two domains. This will allow us to investigate diverse types of Earth materials using frequencies that are much closer to those of seismic waves propagating through the Earth than previously possible.
The novel laboratory enables experimentation under highly controlled conditions. A broad range of long-standing problems in wave propagation and imaging that have eluded Earth scientists and physicists for decades will be addressed. Fine scale heterogeneity, porosity and fluid saturation in real Earth media result in complex frequency-dependent amplitude and phase responses that we can characterize in the laboratory. Synthetically produced complex models can be used in wavefield-focussing experiments and to achieve complete elastic time-reversal for the first time ever. We will study coda waves that can be indicative of slight changes in stress fields before catastrophic fracturing and that might provide pre-cursory signs of earthquakes. Finally, the laboratory is highly relevant to applications such as non-destructive testing, medical imaging and lithotripsy.
Summary
There is a need for a radically new laboratory experimental approach for studying the interaction of seismic waves with the complex media of the Earth’s subsurface. We present here a fundamental new approach to seismic wave experimentation that involves fully immersing a physical seismic experiment within a virtual numerical environment. This enormously challenging endeavour, which is relevant to many outstanding issues in seismology, has not been previously attempted. By continuously varying the output of numerous transponders closely spaced around the physical domain using a control algorithm that takes advantage of measurements made by a scanning Laser-Doppler Vibrometer and a novel theory of exact boundary conditions, waves travelling between the physical and numerical domains will seamlessly propagate back and forth between the two domains without being affected by reflections at the boundaries between the two domains. This will allow us to investigate diverse types of Earth materials using frequencies that are much closer to those of seismic waves propagating through the Earth than previously possible.
The novel laboratory enables experimentation under highly controlled conditions. A broad range of long-standing problems in wave propagation and imaging that have eluded Earth scientists and physicists for decades will be addressed. Fine scale heterogeneity, porosity and fluid saturation in real Earth media result in complex frequency-dependent amplitude and phase responses that we can characterize in the laboratory. Synthetically produced complex models can be used in wavefield-focussing experiments and to achieve complete elastic time-reversal for the first time ever. We will study coda waves that can be indicative of slight changes in stress fields before catastrophic fracturing and that might provide pre-cursory signs of earthquakes. Finally, the laboratory is highly relevant to applications such as non-destructive testing, medical imaging and lithotripsy.
Max ERC Funding
3 498 330 €
Duration
Start date: 2016-12-01, End date: 2021-11-30
Project acronym MEDEA
Project Mechanisms of Epigenetic regulation in Development, Evolution and Adaptation
Researcher (PI) Ulrich Grossniklaus
Host Institution (HI) UNIVERSITAT ZURICH
Call Details Advanced Grant (AdG), LS2, ERC-2009-AdG
Summary Over the last decade epigenetic gene regulation has become a major focus of scientific research as it was shown to play an important role in normal plant and animal development, but also in the ontogeny of human disease. A role of epigenetic processes in evolution, however, has found little general support to date. The goal of this project is to understand the complex interplay of epigenetic mechanisms in plant development and evolution. Many of the approaches we use rely on the recent advances in sequencing technologies, which allow the analysis of molecular characters at an unprecedented level and speed. To achieve our goal, we will focus on two epigenetic paradigms. In Program A, we will focus on dissecting the mechanisms of genomic imprinting at the MEDEA (MEA) locus in Arabidopsis, which we will investigate using genetic, molecular, and innovative biochemical approaches to gain a comprehensive picture of the complex interplay of various epigenetic pathways. In program B, we will analyze the role of epigenetic change in adaptation and evolution using (i) an experimental selection approach in Arabidopsis, where genome-wide analyses of epigenetic modifications have become possible, and (ii) a stable, heritable, epigenetic change occurring in Mimulus populations. In this system, an epigenetic switch of the pollinator syndrome leads to reproductive isolation and, therefore, has an effect on population structure and thus the evolutionary trajectory. These experimental systems each offer unique opportunities to shed light onto the underlying mechanisms controlling epigenetic states. In combination with the new methodologies used, these analyses promise to provide step change in our understanding of epigenetic processes at the level of genes, organisms, and populations.
Summary
Over the last decade epigenetic gene regulation has become a major focus of scientific research as it was shown to play an important role in normal plant and animal development, but also in the ontogeny of human disease. A role of epigenetic processes in evolution, however, has found little general support to date. The goal of this project is to understand the complex interplay of epigenetic mechanisms in plant development and evolution. Many of the approaches we use rely on the recent advances in sequencing technologies, which allow the analysis of molecular characters at an unprecedented level and speed. To achieve our goal, we will focus on two epigenetic paradigms. In Program A, we will focus on dissecting the mechanisms of genomic imprinting at the MEDEA (MEA) locus in Arabidopsis, which we will investigate using genetic, molecular, and innovative biochemical approaches to gain a comprehensive picture of the complex interplay of various epigenetic pathways. In program B, we will analyze the role of epigenetic change in adaptation and evolution using (i) an experimental selection approach in Arabidopsis, where genome-wide analyses of epigenetic modifications have become possible, and (ii) a stable, heritable, epigenetic change occurring in Mimulus populations. In this system, an epigenetic switch of the pollinator syndrome leads to reproductive isolation and, therefore, has an effect on population structure and thus the evolutionary trajectory. These experimental systems each offer unique opportunities to shed light onto the underlying mechanisms controlling epigenetic states. In combination with the new methodologies used, these analyses promise to provide step change in our understanding of epigenetic processes at the level of genes, organisms, and populations.
Max ERC Funding
2 496 641 €
Duration
Start date: 2010-04-01, End date: 2015-12-31
Project acronym MEL-Interactions
Project An integrative approach for the exploration of melanoma genetic and immunological interactions
Researcher (PI) Yardena Rachel SAMUELS
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS2, ERC-2017-COG
Summary Tumor development emerges from an accumulation of somatic alterations that together enable malignant growth. These alterations are immensely diverse and the fate of a cell acquiring an alteration may depend on other alterations already present. Despite our progress in mapping the cancer genetic landscape and an expanding catalogue of cancer genes, a need arises to establish how alterations in cancer genes interact to transform healthy cells into cancer cells. Many fundamental questions regarding genomic interactions remain open. For example, we do not know which proteins make up signaling pathway hubs and in which genetic contexts, how genetic alterations interact functionally, how cancer genetic alterations influence the interaction with T cells and how these affect patient response to therapy. The recent growth in the number of genomics data sets gives rise to a parallel increase in statistical power to detect more complex associations allowing robust analyses of complex interrelated genomic networks. In this proposal we suggest to employ our expertise and unique toolsets, to shed new light on the complex interrelated networks formed in melanoma. We propose to combine state-of-the art high content tools with mechanistic studies to discover the structure of signaling-hub organization in melanoma (Aim 1), functionally characterize the complex genetic interactions within the melanoma genome using genome engineering approaches (Aim 2), and to decipher the immuno-genetic interactions between melanoma and T cells (Aim 3). Importantly, we will try to bridge the knowledge gap in deciphering melanoma-specific gene interactions, protein interactions and interactions with T cells by creating new tools and experimental models. Our findings should make an important step towards an unprecedented, thorough and multifaceted understanding of melanoma biology. More broadly, we believe these approaches provide a paradigm for addressing similarly complex questions in other cancers.
Summary
Tumor development emerges from an accumulation of somatic alterations that together enable malignant growth. These alterations are immensely diverse and the fate of a cell acquiring an alteration may depend on other alterations already present. Despite our progress in mapping the cancer genetic landscape and an expanding catalogue of cancer genes, a need arises to establish how alterations in cancer genes interact to transform healthy cells into cancer cells. Many fundamental questions regarding genomic interactions remain open. For example, we do not know which proteins make up signaling pathway hubs and in which genetic contexts, how genetic alterations interact functionally, how cancer genetic alterations influence the interaction with T cells and how these affect patient response to therapy. The recent growth in the number of genomics data sets gives rise to a parallel increase in statistical power to detect more complex associations allowing robust analyses of complex interrelated genomic networks. In this proposal we suggest to employ our expertise and unique toolsets, to shed new light on the complex interrelated networks formed in melanoma. We propose to combine state-of-the art high content tools with mechanistic studies to discover the structure of signaling-hub organization in melanoma (Aim 1), functionally characterize the complex genetic interactions within the melanoma genome using genome engineering approaches (Aim 2), and to decipher the immuno-genetic interactions between melanoma and T cells (Aim 3). Importantly, we will try to bridge the knowledge gap in deciphering melanoma-specific gene interactions, protein interactions and interactions with T cells by creating new tools and experimental models. Our findings should make an important step towards an unprecedented, thorough and multifaceted understanding of melanoma biology. More broadly, we believe these approaches provide a paradigm for addressing similarly complex questions in other cancers.
Max ERC Funding
2 000 000 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym MELGEN
Project Mutational and Functional Analysis of the Melanoma Genome
Researcher (PI) Yardena Rachel Samuels
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2013-StG
Summary Cancer is a genetic disease that involves the accumulation of somatic mutations. Comprehensive understanding of the mutations that cause cancer and their functional effects is a critical step towards: a) global understanding of cancer development and tumor phenotypes, b) sub-classification of cancer based on mutational signatures, and c) developing targeted therapies based on mutations in specific tumors.
Recent advances in high-throughput genomic technologies provide an unprecedented opportunity to systematically interrogate the cancer genetic landscape. However, although ample genomic sequences are becoming available, so far these have only provided limited information concerning the complexity of the cancer genome. Furthermore, our understanding of the functional effects of identified mutations is hampered by the difficulty in comprehensively assessing their biological and biochemical effects in a physiological manner.
I propose to overcome this major challenge in our understanding of cancer genomics by integrating functional and genetic techniques using melanoma as an example. I propose pioneering high-throughput applications of somatic cell knockout technologies to evaluate newly discovered mutations. The tools developed by this proposal will make it possible to decisively determine the function(s) of proteins that are intimately linked to the pathogenesis of cancer. Funding for this project will enable us to demonstrate the feasibility of initiating a larger scale approach to the ongoing identification of a dynamic and evolving cancer interactome. Furthermore, we will analyze 300 melanoma whole exomes, the largest melanoma dataset to date, to: (i) identify new targetable mutations and (ii) determine the roles of synonymous mutations, which have rarely been studied in cancer. The proposed research will reveal the mechanisms underlying melanoma tumorgenesis, which will in turn generate insights to aid the treatment of melanoma patients.
Summary
Cancer is a genetic disease that involves the accumulation of somatic mutations. Comprehensive understanding of the mutations that cause cancer and their functional effects is a critical step towards: a) global understanding of cancer development and tumor phenotypes, b) sub-classification of cancer based on mutational signatures, and c) developing targeted therapies based on mutations in specific tumors.
Recent advances in high-throughput genomic technologies provide an unprecedented opportunity to systematically interrogate the cancer genetic landscape. However, although ample genomic sequences are becoming available, so far these have only provided limited information concerning the complexity of the cancer genome. Furthermore, our understanding of the functional effects of identified mutations is hampered by the difficulty in comprehensively assessing their biological and biochemical effects in a physiological manner.
I propose to overcome this major challenge in our understanding of cancer genomics by integrating functional and genetic techniques using melanoma as an example. I propose pioneering high-throughput applications of somatic cell knockout technologies to evaluate newly discovered mutations. The tools developed by this proposal will make it possible to decisively determine the function(s) of proteins that are intimately linked to the pathogenesis of cancer. Funding for this project will enable us to demonstrate the feasibility of initiating a larger scale approach to the ongoing identification of a dynamic and evolving cancer interactome. Furthermore, we will analyze 300 melanoma whole exomes, the largest melanoma dataset to date, to: (i) identify new targetable mutations and (ii) determine the roles of synonymous mutations, which have rarely been studied in cancer. The proposed research will reveal the mechanisms underlying melanoma tumorgenesis, which will in turn generate insights to aid the treatment of melanoma patients.
Max ERC Funding
1 500 000 €
Duration
Start date: 2013-10-01, End date: 2018-09-30
Project acronym MERIR
Project Methane related iron reduction processes in sediments: Hidden couplings and their significance for carbon and iron cycles
Researcher (PI) Orit Sivan
Host Institution (HI) BEN-GURION UNIVERSITY OF THE NEGEV
Call Details Consolidator Grant (CoG), PE10, ERC-2018-COG
Summary About one-third of annual methane (CH4) emissions to the atmosphere originate from natural, nonanthropogenic
sources. However, if all the naturally produced methane actually did reach the atmosphere, its
levels would increase by an order of magnitude, dwarfing anthropogenic CO2 emissions. Fortunately, natural
scavengers of this methane near its production zone limit its release. One of these scavengers, iron (Fe) oxide,
can become a major sink for methane when sulfate concentrations are low. Methane-iron couplings in
established sediments, however, are poorly understood. Specifically, significant iron oxide reduction has been
observed in many aquatic sediments at depths well below its expected redox zone, where methane is produced
by methanogenesis, often accompanied by decreases in methane concentrations. These observations challenge
our understandings of iron-methane couplings and microbial players in the deep methanogenic zone and their
impacts on the carbon, iron and other cycles. I aim in the proposed research to elucidate the unexplored
mechanisms of methane-related iron reduction (MERIR) in the methanogenic zone of established
sedimentary profiles under various environmental conditions and their impact on global biogeochemical
cycles. I will resolve two striking yet unexplained phenomena: (1) the active involvement of aerobic
methanotrophs in iron-coupled anaerobic oxidation of methane (AOM), and (2) the unusual reactivity
of iron minerals toward reduction that is accompanied by intensive authigenic magnetite precipitation, and
the effects of this mineralogy change on sedimentary magnetism. My expertise will enable me to achieve the
objectives of this interdisciplinary proposed work using novel approaches from different fields. The project
will likely lead to breakthroughs in our understanding of microbial survival strategies, reveal novel pathways
for aerobic methanotrophs, and change our perspectives on iron mineral reactivities and sedimentary
magnetism.
Summary
About one-third of annual methane (CH4) emissions to the atmosphere originate from natural, nonanthropogenic
sources. However, if all the naturally produced methane actually did reach the atmosphere, its
levels would increase by an order of magnitude, dwarfing anthropogenic CO2 emissions. Fortunately, natural
scavengers of this methane near its production zone limit its release. One of these scavengers, iron (Fe) oxide,
can become a major sink for methane when sulfate concentrations are low. Methane-iron couplings in
established sediments, however, are poorly understood. Specifically, significant iron oxide reduction has been
observed in many aquatic sediments at depths well below its expected redox zone, where methane is produced
by methanogenesis, often accompanied by decreases in methane concentrations. These observations challenge
our understandings of iron-methane couplings and microbial players in the deep methanogenic zone and their
impacts on the carbon, iron and other cycles. I aim in the proposed research to elucidate the unexplored
mechanisms of methane-related iron reduction (MERIR) in the methanogenic zone of established
sedimentary profiles under various environmental conditions and their impact on global biogeochemical
cycles. I will resolve two striking yet unexplained phenomena: (1) the active involvement of aerobic
methanotrophs in iron-coupled anaerobic oxidation of methane (AOM), and (2) the unusual reactivity
of iron minerals toward reduction that is accompanied by intensive authigenic magnetite precipitation, and
the effects of this mineralogy change on sedimentary magnetism. My expertise will enable me to achieve the
objectives of this interdisciplinary proposed work using novel approaches from different fields. The project
will likely lead to breakthroughs in our understanding of microbial survival strategies, reveal novel pathways
for aerobic methanotrophs, and change our perspectives on iron mineral reactivities and sedimentary
magnetism.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym MFECE
Project Magnetostrophic Flow in Experiments and the Core of the Earth
Researcher (PI) Andrew Jackson
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), PE10, ERC-2009-AdG
Summary We describe here an innovative strategy for understanding the so-called magnetostrophic regime of fluid flow in the Earth s core, and thus the mechanisms by which the Earth s magnetic field is sustained over time. The magnetostrophic regime is the state in which Lorentz (magnetic) forces are balanced by Coriolis (rotational) forces and pressure gradients and is thought to be the zeroth order force balance in the core. We propose a series of ground-breaking experiments using liquid sodium contained in a rapidly rotating sphere containing a differentially rotating solid inner sphere. For the first time electric current is injected into the fluid in different configurations in order that the Lorentz force is everywhere significant. Various other magnetic fields can be applied from the exterior and the interior. The influence of turbulence, viscous and magnetic boundary layers will be examined. The presence of instabilities and wave motion will be studied, and the existence of steady solutions will be naturally determined. Diagnostic measurements of magnetic fields and electrical potentials, and Doppler velocimetry will characterise the experiment. These unique experiments are backed by numerical calculations. Complementary studies will analyse the observed magnetic field over the last 400 years in the same magnetostrophic framework. An inverse method will be developed to find the initial state of the field that evolves in a manner compatible with observations. This will elucidate the interior structure of the magnetic field for the first time, determining the amplitude and morphology of the field. The importance of magnetic diffusion (Joule heating) will arise naturally, and fluid motion in the entire core will be found, allowing comparison with geodetic observations.
Summary
We describe here an innovative strategy for understanding the so-called magnetostrophic regime of fluid flow in the Earth s core, and thus the mechanisms by which the Earth s magnetic field is sustained over time. The magnetostrophic regime is the state in which Lorentz (magnetic) forces are balanced by Coriolis (rotational) forces and pressure gradients and is thought to be the zeroth order force balance in the core. We propose a series of ground-breaking experiments using liquid sodium contained in a rapidly rotating sphere containing a differentially rotating solid inner sphere. For the first time electric current is injected into the fluid in different configurations in order that the Lorentz force is everywhere significant. Various other magnetic fields can be applied from the exterior and the interior. The influence of turbulence, viscous and magnetic boundary layers will be examined. The presence of instabilities and wave motion will be studied, and the existence of steady solutions will be naturally determined. Diagnostic measurements of magnetic fields and electrical potentials, and Doppler velocimetry will characterise the experiment. These unique experiments are backed by numerical calculations. Complementary studies will analyse the observed magnetic field over the last 400 years in the same magnetostrophic framework. An inverse method will be developed to find the initial state of the field that evolves in a manner compatible with observations. This will elucidate the interior structure of the magnetic field for the first time, determining the amplitude and morphology of the field. The importance of magnetic diffusion (Joule heating) will arise naturally, and fluid motion in the entire core will be found, allowing comparison with geodetic observations.
Max ERC Funding
3 116 900 €
Duration
Start date: 2010-05-01, End date: 2016-04-30
Project acronym MIRTURN
Project Mechanisms of microRNA biogenesis and turnover
Researcher (PI) Helge Grosshans
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Call Details Starting Grant (StG), LS2, ERC-2009-StG
Summary MicroRNAs (miRNAs) are a novel class of genes, accounting for >1% of genes in a typical animal genome. They constitute an important layer of gene regulation that affects diverse processes such as cell differentiation, apoptosis, and metabolism. Despite such critical roles, deciphering the mechanism of action of miRNAs has been difficult, leading to multiple, partially contradictory, models of miRNA activity. Moreover, adding an additional layer of complexity, it is now emerging that miRNA activity is regulated by various mechanisms that we are only beginning to identify. Our objective is to understand how miRNAs are regulated under physiological conditions, in the roundworm Caenorhabditis elegans. We will focus on pathways of miRNA turnover, an issue of fundamental importance that has received little attention because miRNAs are widely held to be highly stable molecules. However, miRNA over-accumulation causes aberrant development and disease, prompting us to test rigorously whether degradation can antagonize miRNA activity and either identify the machinery involved, or confirm the dominance of other regulatory modalities, whose components we will identify. C. elegans is the organism in which miRNAs and many components of the miRNA machinery were discovered. However, previous studies emphasized genetics and cell biology approaches, limiting the degree of mechanistic insight that could be obtained. In addition to exploiting the traditional strengths of C. elegans, we will therefore develop and apply biochemical and genomic techniques to obtain a comprehensive understanding of miRNA regulation, enabling us to demonstrate both molecular mechanisms and physiological relevance. Given the importance of miRNAs in development and disease, identifying the regulators of these tiny gene regulators will be both of scientific interest and biomedical relevance.
Summary
MicroRNAs (miRNAs) are a novel class of genes, accounting for >1% of genes in a typical animal genome. They constitute an important layer of gene regulation that affects diverse processes such as cell differentiation, apoptosis, and metabolism. Despite such critical roles, deciphering the mechanism of action of miRNAs has been difficult, leading to multiple, partially contradictory, models of miRNA activity. Moreover, adding an additional layer of complexity, it is now emerging that miRNA activity is regulated by various mechanisms that we are only beginning to identify. Our objective is to understand how miRNAs are regulated under physiological conditions, in the roundworm Caenorhabditis elegans. We will focus on pathways of miRNA turnover, an issue of fundamental importance that has received little attention because miRNAs are widely held to be highly stable molecules. However, miRNA over-accumulation causes aberrant development and disease, prompting us to test rigorously whether degradation can antagonize miRNA activity and either identify the machinery involved, or confirm the dominance of other regulatory modalities, whose components we will identify. C. elegans is the organism in which miRNAs and many components of the miRNA machinery were discovered. However, previous studies emphasized genetics and cell biology approaches, limiting the degree of mechanistic insight that could be obtained. In addition to exploiting the traditional strengths of C. elegans, we will therefore develop and apply biochemical and genomic techniques to obtain a comprehensive understanding of miRNA regulation, enabling us to demonstrate both molecular mechanisms and physiological relevance. Given the importance of miRNAs in development and disease, identifying the regulators of these tiny gene regulators will be both of scientific interest and biomedical relevance.
Max ERC Funding
1 782 200 €
Duration
Start date: 2010-05-01, End date: 2015-04-30
Project acronym NOVCARBFIX
Project Analysis, Design and Experimental Evolution of Novel Carbon Fixation Pathways
Researcher (PI) Ron Milo
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS2, ERC-2014-CoG
Summary Carbon fixation is a prerequisite for accumulating biomass and storing energy in most of the living world. As such, it supplies our food and dominates land and water usage by humanity. In agriculture, where water and nutrients are abundant, the rate of carbon fixation often limits growth rate. Therefore increasing the rate of carbon fixation is of global importance towards agricultural and energetic sustainability.
What are the limits on the possible rate of carbon fixation? Attempts to improve RuBisCO, the key enzyme in the Calvin-Benson cycle, have achieved only limited results. My lab focuses on trying to overcome this global challenge by building synthetic pathways for carbon fixation. We create a computational framework that designs and scores pathways and creates step-wise selection strategies for in-vivo experimental implementation. Our most promising synthetic carbon fixation pathways are found to utilize the highly effective carboxylating enzyme, PEP carboxylase. We experimentally test these pathways in the most genetically tractable context by constructing an E.coli strain that depends on atmospheric CO2 fixation. We will gradually incorporate the pathways, initially as essential reaction steps for biomass production, and finally with CO2 as sole carbon input of the cell.
As a stepping-stone towards this challenging goal, we will construct an autotrophic E.coli strain that uses the Calvin-Benson cycle. We systematically convert this synthetic biology grand challenge into a gradual evolutionary ladder with independently selectable steps. We recently achieved key steps in the ladder, such as semi-autotrophic growth, serving as powerful proofs of concept.
The proposed research will advance our basic-science understanding of evolutionary plasticity of metabolic pathways. It also paves the way for a hybrid rational-design/experimental-evolution approach to revisit and advance the capacity of metabolism for agricultural productivity and renewable energy storage.
Summary
Carbon fixation is a prerequisite for accumulating biomass and storing energy in most of the living world. As such, it supplies our food and dominates land and water usage by humanity. In agriculture, where water and nutrients are abundant, the rate of carbon fixation often limits growth rate. Therefore increasing the rate of carbon fixation is of global importance towards agricultural and energetic sustainability.
What are the limits on the possible rate of carbon fixation? Attempts to improve RuBisCO, the key enzyme in the Calvin-Benson cycle, have achieved only limited results. My lab focuses on trying to overcome this global challenge by building synthetic pathways for carbon fixation. We create a computational framework that designs and scores pathways and creates step-wise selection strategies for in-vivo experimental implementation. Our most promising synthetic carbon fixation pathways are found to utilize the highly effective carboxylating enzyme, PEP carboxylase. We experimentally test these pathways in the most genetically tractable context by constructing an E.coli strain that depends on atmospheric CO2 fixation. We will gradually incorporate the pathways, initially as essential reaction steps for biomass production, and finally with CO2 as sole carbon input of the cell.
As a stepping-stone towards this challenging goal, we will construct an autotrophic E.coli strain that uses the Calvin-Benson cycle. We systematically convert this synthetic biology grand challenge into a gradual evolutionary ladder with independently selectable steps. We recently achieved key steps in the ladder, such as semi-autotrophic growth, serving as powerful proofs of concept.
The proposed research will advance our basic-science understanding of evolutionary plasticity of metabolic pathways. It also paves the way for a hybrid rational-design/experimental-evolution approach to revisit and advance the capacity of metabolism for agricultural productivity and renewable energy storage.
Max ERC Funding
1 999 843 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym NucleolusChromatin
Project Analysis of the nucleolus in genome organization and function
Researcher (PI) Raffaella SANTORO
Host Institution (HI) UNIVERSITAT ZURICH
Call Details Advanced Grant (AdG), LS2, ERC-2017-ADG
Summary In eukaryotic cells, the higher-order organization of genomes is functionally important to ensure correct execution of gene expression programs. For instance, as cells differentiate into specialized cell types, chromosomes undergo diverse structural and organizational changes that affect gene expression and other cellular functions. However, how this process is achieved is still poorly understood. The elucidation of the mechanisms that control the spatial architecture of the genome and its contribution to gene regulation is a key open issue in molecular biology, relevant for physiological and pathological processes.
Increasing evidence indicated that large-scale folding of chromatin may affect gene expression by locating genes to specific nuclear subcompartments that are either stimulatory or inhibitory to transcription. Nuclear periphery (NP) and nucleolus are two important nuclear landmarks where repressive chromatin domains are often located. The interaction of chromosomes with NP and nucleolus is thought to contribute to a basal chromosome architecture and genome function. However, while the role of NP in genome organization has been well documented, the function of the nucleolus remains yet elusive.
To fully understand how genome organization regulates chromatin and gene expression states, it is necessary to obtain a comprehensive functional map of genome compartmentalization. However, so far, only domains associating with NP (LADs) have been identified and characterized while nucleolar-associated domains (NADs) remained under-investigated. The aim of this project is to fill this gap by developing methods to identify and characterize NADs and analyse the role of the nucleolus in genome organization, moving toward the obtainment of a comprehensive functional map of genome compartmentalization for each cell state and providing novel insights into basic principles of genome organization and its role in gene expression and cell function that yet remain elusive.
Summary
In eukaryotic cells, the higher-order organization of genomes is functionally important to ensure correct execution of gene expression programs. For instance, as cells differentiate into specialized cell types, chromosomes undergo diverse structural and organizational changes that affect gene expression and other cellular functions. However, how this process is achieved is still poorly understood. The elucidation of the mechanisms that control the spatial architecture of the genome and its contribution to gene regulation is a key open issue in molecular biology, relevant for physiological and pathological processes.
Increasing evidence indicated that large-scale folding of chromatin may affect gene expression by locating genes to specific nuclear subcompartments that are either stimulatory or inhibitory to transcription. Nuclear periphery (NP) and nucleolus are two important nuclear landmarks where repressive chromatin domains are often located. The interaction of chromosomes with NP and nucleolus is thought to contribute to a basal chromosome architecture and genome function. However, while the role of NP in genome organization has been well documented, the function of the nucleolus remains yet elusive.
To fully understand how genome organization regulates chromatin and gene expression states, it is necessary to obtain a comprehensive functional map of genome compartmentalization. However, so far, only domains associating with NP (LADs) have been identified and characterized while nucleolar-associated domains (NADs) remained under-investigated. The aim of this project is to fill this gap by developing methods to identify and characterize NADs and analyse the role of the nucleolus in genome organization, moving toward the obtainment of a comprehensive functional map of genome compartmentalization for each cell state and providing novel insights into basic principles of genome organization and its role in gene expression and cell function that yet remain elusive.
Max ERC Funding
2 500 000 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym OOID
Project The Ocean's Oxygen Isotopes Deciphered: Combining Observations, Experiments and Models
Researcher (PI) Itay HALEVY
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), PE10, ERC-2017-STG
Summary The isotopic composition of O in seawater is a fundamental property of Earth's oceans, key to paleoclimate reconstructions and to our understanding of the origin of water on Earth, the water-rock reactions that govern seawater chemistry, and the conditions under which life emerged. Despite more than five decades of research, the geologic history of seawater 18O/16O remains a topic of intense debate. Without exception, well-preserved 18O/16O records from marine precipitates reflect both the minerals' formation temperature, and the isotopic composition of seawater. This duality has prevented unique interpretation of a long-term secular trend, in which 18O/16O in sedimentary rocks (e.g., carbonates, cherts) has increased by ~15 ‰ since the Archean. Here I outline an inter-disciplinary research program to address this fundamental problem, which integrates new geochemical observations, laboratory experiments, and numerical models.
We will generate geologic records of 18O/16O in two previously untapped repositories: iron oxides and iron-bearing authigenic clays. Several characteristics of both, and preliminary results, suggest that these repositories hold the potential to settle the long-standing debate about seawater 18O/16O. We will determine the temperature dependence of mineral-water O isotope fractionation in laboratory experiments and observations of natural systems. We will experimentally test the resistance of these minerals to O isotope exchange under geologically-relevant conditions, with the aim of evaluating the potential for late-stage isotopic resetting. Finally, we will develop models of the marine O isotope cycle, which account for the processes that govern seawater 18O/16O over long timescales, and which will be used to provide a quantitative understanding of the new records. With these new insights, we will explore implications for the geologic history of seawater chemistry, atmospheric composition, climate and biology.
Summary
The isotopic composition of O in seawater is a fundamental property of Earth's oceans, key to paleoclimate reconstructions and to our understanding of the origin of water on Earth, the water-rock reactions that govern seawater chemistry, and the conditions under which life emerged. Despite more than five decades of research, the geologic history of seawater 18O/16O remains a topic of intense debate. Without exception, well-preserved 18O/16O records from marine precipitates reflect both the minerals' formation temperature, and the isotopic composition of seawater. This duality has prevented unique interpretation of a long-term secular trend, in which 18O/16O in sedimentary rocks (e.g., carbonates, cherts) has increased by ~15 ‰ since the Archean. Here I outline an inter-disciplinary research program to address this fundamental problem, which integrates new geochemical observations, laboratory experiments, and numerical models.
We will generate geologic records of 18O/16O in two previously untapped repositories: iron oxides and iron-bearing authigenic clays. Several characteristics of both, and preliminary results, suggest that these repositories hold the potential to settle the long-standing debate about seawater 18O/16O. We will determine the temperature dependence of mineral-water O isotope fractionation in laboratory experiments and observations of natural systems. We will experimentally test the resistance of these minerals to O isotope exchange under geologically-relevant conditions, with the aim of evaluating the potential for late-stage isotopic resetting. Finally, we will develop models of the marine O isotope cycle, which account for the processes that govern seawater 18O/16O over long timescales, and which will be used to provide a quantitative understanding of the new records. With these new insights, we will explore implications for the geologic history of seawater chemistry, atmospheric composition, climate and biology.
Max ERC Funding
1 490 596 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym ORDER IN DISORDER
Project Order in Disorder: Context-dependent strategies for integrating peptide-mediated interactions
Researcher (PI) Ora Schueler-Furman
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS2, ERC-2012-StG_20111109
Summary Peptide-mediated protein interactions are emerging as key regulators of many important regulatory processes in the cell. Short motifs, usually embedded into disordered regions of the protein, interact with specific sets of protein partners in a transient way and allow efficient and malleable propagation of signals towards their targets. While regular protein interactions have been intensively studied, many aspects of peptide-mediated interactions have not yet been elucidated.
The aim of this proposal is to improve our understanding of the basic strategies that are employed by peptide-mediated interactions to achieve different types of outcomes in different settings, and how the context of the peptide influences this outcome. Towards this aim, we will establish two complementary strategies, namely (1) a significant extension of our modeling tools for peptide-protein complex structures that will allow modeling of effects of the surrounding flexible linker, and (2) the establishment of an experimental lab that will allow us to independently validate and complement our modeling results. Targeted modulation of peptide affinity, specificity, and linker length and sequence, using both computational design as well as experimental in vitro evolution, will dissect different contributions to the functional outcome of a peptide-mediated interaction within its context. We can thus study in detail the interplay of the interaction with additional features in the linker sequence, such as posttranslational modification sites, as well as additional peptide binding motifs and interactions.
Interactions mediated by intrinsically disordered regions are omnipresent. Their accurate characterization, modeling and manipulation holds therefore many promises towards applications for the development of better drugs and basic insights for better fundamental understanding of the underlying basis of regulatory interactions.
Summary
Peptide-mediated protein interactions are emerging as key regulators of many important regulatory processes in the cell. Short motifs, usually embedded into disordered regions of the protein, interact with specific sets of protein partners in a transient way and allow efficient and malleable propagation of signals towards their targets. While regular protein interactions have been intensively studied, many aspects of peptide-mediated interactions have not yet been elucidated.
The aim of this proposal is to improve our understanding of the basic strategies that are employed by peptide-mediated interactions to achieve different types of outcomes in different settings, and how the context of the peptide influences this outcome. Towards this aim, we will establish two complementary strategies, namely (1) a significant extension of our modeling tools for peptide-protein complex structures that will allow modeling of effects of the surrounding flexible linker, and (2) the establishment of an experimental lab that will allow us to independently validate and complement our modeling results. Targeted modulation of peptide affinity, specificity, and linker length and sequence, using both computational design as well as experimental in vitro evolution, will dissect different contributions to the functional outcome of a peptide-mediated interaction within its context. We can thus study in detail the interplay of the interaction with additional features in the linker sequence, such as posttranslational modification sites, as well as additional peptide binding motifs and interactions.
Interactions mediated by intrinsically disordered regions are omnipresent. Their accurate characterization, modeling and manipulation holds therefore many promises towards applications for the development of better drugs and basic insights for better fundamental understanding of the underlying basis of regulatory interactions.
Max ERC Funding
1 499 808 €
Duration
Start date: 2012-12-01, End date: 2017-11-30
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 PALAEO-RA
Project A Palaeoreanalysis To Understand Decadal Climate Variability
Researcher (PI) Stefan BRÖNNIMANN
Host Institution (HI) UNIVERSITAET BERN
Call Details Advanced Grant (AdG), PE10, ERC-2017-ADG
Summary Climatic variations at decadal scales, such as phases of accelerated warming, weak monsoons, or widespread subtropical drought, have profound effects on society and the economy. Understanding such variations requires insights from the past. However, no data sets of past climate are available to study decadal variability of large-scale climate with state-of-the-art diagnostic methods. Currently available data sets are limited to statistical reconstructions of local or regional surface climate. The PALAEO-RA project will produce the first ever comprehensive, 3-dimensional, physically consistent reconstruction of the global climate system at a monthly scale for the past six centuries. This palaeoreanalysis is based on combining information from early instrumental measurements, historical documents (e.g., capitalizing on large amounts of newly available data from China), and proxies (e.g., tree rings) with a large ensemble of climate model simulations. To achieve this novel combination, a completely new data assimilation system for palaeoclimatological data will be developed. The unique data sets produced in this project will become reference data sets for studying past climatic variations (i) for diagnostic studies of interannual-to-decadal variability, (ii) as a benchmark for model simulations and (iii) for climate impact studies. Using the data produced, the project will analyse episodes of slowed or accelerated global warming, decadal subtropical drought periods, episodes of expanding or contracting tropics, slowed or strengthened monsoons, changes in storm tracks, blocking and associated weather extremes, and links between Arctic and midlatitude climate. The analyses will provide new insights into the processes governing decadal variability of weather and climate.
Summary
Climatic variations at decadal scales, such as phases of accelerated warming, weak monsoons, or widespread subtropical drought, have profound effects on society and the economy. Understanding such variations requires insights from the past. However, no data sets of past climate are available to study decadal variability of large-scale climate with state-of-the-art diagnostic methods. Currently available data sets are limited to statistical reconstructions of local or regional surface climate. The PALAEO-RA project will produce the first ever comprehensive, 3-dimensional, physically consistent reconstruction of the global climate system at a monthly scale for the past six centuries. This palaeoreanalysis is based on combining information from early instrumental measurements, historical documents (e.g., capitalizing on large amounts of newly available data from China), and proxies (e.g., tree rings) with a large ensemble of climate model simulations. To achieve this novel combination, a completely new data assimilation system for palaeoclimatological data will be developed. The unique data sets produced in this project will become reference data sets for studying past climatic variations (i) for diagnostic studies of interannual-to-decadal variability, (ii) as a benchmark for model simulations and (iii) for climate impact studies. Using the data produced, the project will analyse episodes of slowed or accelerated global warming, decadal subtropical drought periods, episodes of expanding or contracting tropics, slowed or strengthened monsoons, changes in storm tracks, blocking and associated weather extremes, and links between Arctic and midlatitude climate. The analyses will provide new insights into the processes governing decadal variability of weather and climate.
Max ERC Funding
2 499 975 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym PhageResist
Project Beyond CRISPR: Systematic characterization of novel anti-phage defense systems in the microbial pan-genome
Researcher (PI) Rotem Sorek
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE LTD
Call Details Consolidator Grant (CoG), LS2, ERC-2015-CoG
Summary The perpetual arms race between bacteria and phage has resulted in the evolution of efficient resistance systems that protect bacteria from phage infection. Such systems, which include restriction enzymes and CRISPR-Cas, have major influence on the evolution of both bacteria and phage, and have also proven to be invaluable for molecular and biotechnological applications. Although much have been learned on the biology of bacterial defense against phage, more than half of all sequenced bacteria do not contain CRISPR-Cas, and it is estimated that many additional, yet-uncharacterized anti-phage defense systems are encoded in bacterial genomes.
The goal of this project is to systematically understand the arsenal of defense mechanisms that are at the disposal of microbes in their struggle against phages. The project combines computational genomics, synthetic biology, high-throughput robotic assays, and deep genetic and biochemical experiments to discover, verify, and study the properties of anti-phage defense systems.
Summary
The perpetual arms race between bacteria and phage has resulted in the evolution of efficient resistance systems that protect bacteria from phage infection. Such systems, which include restriction enzymes and CRISPR-Cas, have major influence on the evolution of both bacteria and phage, and have also proven to be invaluable for molecular and biotechnological applications. Although much have been learned on the biology of bacterial defense against phage, more than half of all sequenced bacteria do not contain CRISPR-Cas, and it is estimated that many additional, yet-uncharacterized anti-phage defense systems are encoded in bacterial genomes.
The goal of this project is to systematically understand the arsenal of defense mechanisms that are at the disposal of microbes in their struggle against phages. The project combines computational genomics, synthetic biology, high-throughput robotic assays, and deep genetic and biochemical experiments to discover, verify, and study the properties of anti-phage defense systems.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym PNET
Project Principles of biomolecular networks
Researcher (PI) Naama Barkai
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS2, ERC-2013-ADG
Summary Cells process information using biochemical circuits of interacting proteins and genes. We wish to define principles guiding the design of those circuits. The interplay between variability and robustness is of key interest to us. Bio-molecular processes are stochastic, environmental conditions fluctuate, and sequence polymorphisms are abundant. How is variability buffered to maintain reproducible outcomes? Can variability enhance computational abilities? What is the impact of variability on bio-molecular circuit design? We will explore those fundamental questions in three contexts:
Source of variability in Gene expression: We previously examined the mechanistic basis of expression variability, defining promoter structures associated with low vs. high variability. We will now address the more challenging question: what evolutionary pressures shape the expression program? On the network level, we will define mutual effects of selection for increased expression and for optimal growth. On the metabolic level, we will define which aspect of the expression process is limiting and the genomic consequences of this limitation.
Role of expression variability in Nutrient homeostasis: We recently reported that repression of high affinity transporter in rich nutrient (the ‘dual-transporter’ motif) enables advanced preparation to nutrient depletion. We will now validate an additional predicted property of this motif: cells become committed to the starvation program, escaping it due to expression noise only. To this end, we will introduce a novel method for modulating expression noise while maintaining mean abundance.
Buffering variability in Embryonic patterning: Buffering fluctuations is essential in embryonic patterning. We previously established that the embryonic DV axis of Drosophila is robustly patterned through the newly defined shuttling mechanism. We will quantify the ability of this system to buffer size variations (scaling), and reveal the underlying scaling mechanism.
Summary
Cells process information using biochemical circuits of interacting proteins and genes. We wish to define principles guiding the design of those circuits. The interplay between variability and robustness is of key interest to us. Bio-molecular processes are stochastic, environmental conditions fluctuate, and sequence polymorphisms are abundant. How is variability buffered to maintain reproducible outcomes? Can variability enhance computational abilities? What is the impact of variability on bio-molecular circuit design? We will explore those fundamental questions in three contexts:
Source of variability in Gene expression: We previously examined the mechanistic basis of expression variability, defining promoter structures associated with low vs. high variability. We will now address the more challenging question: what evolutionary pressures shape the expression program? On the network level, we will define mutual effects of selection for increased expression and for optimal growth. On the metabolic level, we will define which aspect of the expression process is limiting and the genomic consequences of this limitation.
Role of expression variability in Nutrient homeostasis: We recently reported that repression of high affinity transporter in rich nutrient (the ‘dual-transporter’ motif) enables advanced preparation to nutrient depletion. We will now validate an additional predicted property of this motif: cells become committed to the starvation program, escaping it due to expression noise only. To this end, we will introduce a novel method for modulating expression noise while maintaining mean abundance.
Buffering variability in Embryonic patterning: Buffering fluctuations is essential in embryonic patterning. We previously established that the embryonic DV axis of Drosophila is robustly patterned through the newly defined shuttling mechanism. We will quantify the ability of this system to buffer size variations (scaling), and reveal the underlying scaling mechanism.
Max ERC Funding
2 311 000 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym POPRNASEQ
Project Population transcriptional genomics in humans using high throughput sequencing
Researcher (PI) Emmanouil Dermitzakis
Host Institution (HI) UNIVERSITE DE GENEVE
Call Details Starting Grant (StG), LS2, ERC-2010-StG_20091118
Summary Gene expression is one of the marks of cellular state and function. The relative abundance of transcripts defines and is a result of the differentiation status of a cell. Interrogation of gene expression levels and patterns in the human and other genomes can be informative about perturbations from the average pattern due to external stimuli or internal factors such as genetic variants. Gene expression profiles have been extensively used to assess developmental processes, pathways contributing to cell differentiation, and predicting the outcome of disease status.
Understanding the effects of genetic variation in basic cellular processes such as gene expression is key to the dissection of the genetic contributions to whole organism phenotypes.
We propose to interrogate the transcriptome of primary fibroblasts, primary T-cells and EBV-transformed B-cell (lymphoblastoid cell lines or LCLs) from umbilical cords of 200 individuals of European descent using next generation sequencing (mRNAseq). A subset will also be interrogated for transcriptionally engaged RNA polymerases (GROseq) and protein abundance. These data will be analyzed for the detection of eQTLs and other genetic effects associated with variation in alternative splicing and other properties of the transcripts and dissection of the genetic effects from primary transcription to protein and their tissue specific effects. These data will be integrated with genome-wide association studies and other efforts to dissect the genetic basis of complex traits and diseases in humans. In addition, we will develop bioinformatic models to understand the fine scale regulatory signals that are responsible for the regulatory patterns observed and how sequence variants have an effect on them.
Summary
Gene expression is one of the marks of cellular state and function. The relative abundance of transcripts defines and is a result of the differentiation status of a cell. Interrogation of gene expression levels and patterns in the human and other genomes can be informative about perturbations from the average pattern due to external stimuli or internal factors such as genetic variants. Gene expression profiles have been extensively used to assess developmental processes, pathways contributing to cell differentiation, and predicting the outcome of disease status.
Understanding the effects of genetic variation in basic cellular processes such as gene expression is key to the dissection of the genetic contributions to whole organism phenotypes.
We propose to interrogate the transcriptome of primary fibroblasts, primary T-cells and EBV-transformed B-cell (lymphoblastoid cell lines or LCLs) from umbilical cords of 200 individuals of European descent using next generation sequencing (mRNAseq). A subset will also be interrogated for transcriptionally engaged RNA polymerases (GROseq) and protein abundance. These data will be analyzed for the detection of eQTLs and other genetic effects associated with variation in alternative splicing and other properties of the transcripts and dissection of the genetic effects from primary transcription to protein and their tissue specific effects. These data will be integrated with genome-wide association studies and other efforts to dissect the genetic basis of complex traits and diseases in humans. In addition, we will develop bioinformatic models to understand the fine scale regulatory signals that are responsible for the regulatory patterns observed and how sequence variants have an effect on them.
Max ERC Funding
1 500 000 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym POSTTRANS
Project An interdisciplinary genome-wide study of posttranscriptional regulation by small RNAs: from individual interactions to networks and evolution
Researcher (PI) Hanah Margalit
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS2, ERC-2012-ADG_20120314
Summary Deciphering the interactions within and between the three major components of living organisms, DNA, RNA and Protein, is at the heart of biological research. New large-scale experimental methods have dramatically advanced genome-wide detection of protein-protein, protein-DNA, protein-RNA and protein-mediated RNA-RNA interactions. However, at present there is no large-scale method that could detect all RNA-RNA interactions independent of a mediator protein, or when the mediator protein is unknown. Attaining such a method is of utmost importance and is very timely, as it is now evident that RNA-RNA interactions play central roles in cellular life. In particular, hundreds of expressed small RNA (sRNA) molecules were discovered in both pro- and eukaryotes, many of which act as posttranscriptional regulators of gene expression by base-pairing with their mRNA targets. It seems that in many organisms the layer of posttranscriptional regulation is as widespread as transcription regulation, presenting a major challenge towards achieving functional and mechanistic understanding of this regulation level. Here we propose to develop an innovative methodology for genome-wide detection of the sRNA targetome, all mRNA targets of cellular sRNAs. This new methodology combines in vivo structural probing with deep sequencing and is independent of protein considerations. We will apply this method to deciper the sRNA targetome of the model organism Escherichia coli, which encodes over 100 sRNAs. We will use the sRNA targetome data as the foundation for a systematic ‘bottom-up’ computational analysis of multifaceted aspects of sRNA-mediated posttranscriptional regulation, encompassing the basic underlying rules of sRNA-mRNA target recognition, the design principles of the posttranscriptional regulatory network and its integration with the transcriptional and metabolic networks, and the evolution of posttranscriptional regulation.
Summary
Deciphering the interactions within and between the three major components of living organisms, DNA, RNA and Protein, is at the heart of biological research. New large-scale experimental methods have dramatically advanced genome-wide detection of protein-protein, protein-DNA, protein-RNA and protein-mediated RNA-RNA interactions. However, at present there is no large-scale method that could detect all RNA-RNA interactions independent of a mediator protein, or when the mediator protein is unknown. Attaining such a method is of utmost importance and is very timely, as it is now evident that RNA-RNA interactions play central roles in cellular life. In particular, hundreds of expressed small RNA (sRNA) molecules were discovered in both pro- and eukaryotes, many of which act as posttranscriptional regulators of gene expression by base-pairing with their mRNA targets. It seems that in many organisms the layer of posttranscriptional regulation is as widespread as transcription regulation, presenting a major challenge towards achieving functional and mechanistic understanding of this regulation level. Here we propose to develop an innovative methodology for genome-wide detection of the sRNA targetome, all mRNA targets of cellular sRNAs. This new methodology combines in vivo structural probing with deep sequencing and is independent of protein considerations. We will apply this method to deciper the sRNA targetome of the model organism Escherichia coli, which encodes over 100 sRNAs. We will use the sRNA targetome data as the foundation for a systematic ‘bottom-up’ computational analysis of multifaceted aspects of sRNA-mediated posttranscriptional regulation, encompassing the basic underlying rules of sRNA-mRNA target recognition, the design principles of the posttranscriptional regulatory network and its integration with the transcriptional and metabolic networks, and the evolution of posttranscriptional regulation.
Max ERC Funding
2 329 360 €
Duration
Start date: 2013-02-01, End date: 2019-01-31
Project acronym PROKRNA
Project Prokaryotic RNomics: Unravelling the RNA-mediated regulatory layers
Researcher (PI) Rotem Sorek
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2010-StG_20091118
Summary Pioneering studies from the recent year, including those published by the PI of this proposal, are revolutionizing our perception of prokaryotic transcriptomes, and reveal unexpected regulatory complexity. Two central concepts are arising: the unanticipated abundance of cis-antisense RNAs overlapping protein coding genes, and alternative transcripts resulting from a dynamic behaviour of operon structures (where genes can be included or excluded from a polycistronic transcript in response to environmental cues). Understanding these phenomena holds a great potential for our ability to decipher how bacteria regulate their complex life styles and pathogenic behaviours, but their dynamics, regulatory roles, and effects on combinatorially increasing the regulatory capacity of the genome are completely unknown.
The primary objectives of this proposed research are: i) to understand the extent, regulatory roles, and evolutionary consequences of cis-antisense
RNAs in prokaryotes; ii) to understand the regulatory code, combinatorial effects and dynamics of alternative operon structures; and, in parallel iii) to develop a unified framework for comparative prokaryotic transcriptomics.
Our strategy is based on a combination of deep sequencing technologies, computational modelling and data analyses, systems biology
approaches, and focused molecular biology experiments. We will identify the extent and the impact of these RNA-based regulatory layers in representative pathogenic and non-pathogenic species across the prokaryotic tree of life, study their functional and evolutionary consequences, and break the regulatory code controlling them. Our planned research has the potential of producing
methodological and conceptual breakthroughs in the emerging field of prokaryotic transcriptomics.
Summary
Pioneering studies from the recent year, including those published by the PI of this proposal, are revolutionizing our perception of prokaryotic transcriptomes, and reveal unexpected regulatory complexity. Two central concepts are arising: the unanticipated abundance of cis-antisense RNAs overlapping protein coding genes, and alternative transcripts resulting from a dynamic behaviour of operon structures (where genes can be included or excluded from a polycistronic transcript in response to environmental cues). Understanding these phenomena holds a great potential for our ability to decipher how bacteria regulate their complex life styles and pathogenic behaviours, but their dynamics, regulatory roles, and effects on combinatorially increasing the regulatory capacity of the genome are completely unknown.
The primary objectives of this proposed research are: i) to understand the extent, regulatory roles, and evolutionary consequences of cis-antisense
RNAs in prokaryotes; ii) to understand the regulatory code, combinatorial effects and dynamics of alternative operon structures; and, in parallel iii) to develop a unified framework for comparative prokaryotic transcriptomics.
Our strategy is based on a combination of deep sequencing technologies, computational modelling and data analyses, systems biology
approaches, and focused molecular biology experiments. We will identify the extent and the impact of these RNA-based regulatory layers in representative pathogenic and non-pathogenic species across the prokaryotic tree of life, study their functional and evolutionary consequences, and break the regulatory code controlling them. Our planned research has the potential of producing
methodological and conceptual breakthroughs in the emerging field of prokaryotic transcriptomics.
Max ERC Funding
1 499 540 €
Duration
Start date: 2011-01-01, End date: 2016-06-30
Project acronym PROTEOMICAN
Project Discovery of breast cancer aggressiveness markers using topo-proteomics mapping
Researcher (PI) Tamar Geiger
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS2, ERC-2014-STG
Summary "In the proposed research we will explore the functional proteomic diversity of histologically-defined regions within human breast tumors, aiming to identify novel protein biomarkers of tumor aggressiveness. Once identified, these proteins will serve as potent diagnostic markers and therapeutic targets. Towards this aim we will perform genome-scale proteomic profiling on tumor regions displaying diverse histopathology. This will be followed by functional investigation of these cancer cell sub-populations to determine their tumorigenic potential, and search for microparticle-based proteomic biomarkers from serum samples towards identification of cancer aggressiveness in blood tests.
Analysis of the proteomic diversity holds a promise to reveal yet unidentified regulators of the tumorigenic phenotype as quantitative protein profiling is expected to most faithfully predict cellular phenotypes. This will be accomplished using the 'super-SILAC' technology, which I developed during my post-doctoral research. Using this technology, we identified over 12,000 proteins in formalin-fixed paraffin embedded breast cancer tumors. In the current project we will take one large step further, namely, microdissect and analyze selected regions in breast tumors based on local histopathological characteristics, such as the expression of known markers, cancer cell density, the vicinity to blood vessels and to the tumor invasive front. This ""topological map"" of the proteome will be followed by functional in vitro and in vivo studies, directly probing the aggressiveness of these cell populations, manifested by an accelerated proliferation and invasive/metastatic capacity. Finally, proteins associated with tumor aggressiveness will serve as blood-based biomarkers for predicting the tumorigenic phenotype using non-invasive tests. This work will set the basis for quantitative probing of tumor heterogeneity, which is crucial for accurate diagnosis and effective therapy.
"
Summary
"In the proposed research we will explore the functional proteomic diversity of histologically-defined regions within human breast tumors, aiming to identify novel protein biomarkers of tumor aggressiveness. Once identified, these proteins will serve as potent diagnostic markers and therapeutic targets. Towards this aim we will perform genome-scale proteomic profiling on tumor regions displaying diverse histopathology. This will be followed by functional investigation of these cancer cell sub-populations to determine their tumorigenic potential, and search for microparticle-based proteomic biomarkers from serum samples towards identification of cancer aggressiveness in blood tests.
Analysis of the proteomic diversity holds a promise to reveal yet unidentified regulators of the tumorigenic phenotype as quantitative protein profiling is expected to most faithfully predict cellular phenotypes. This will be accomplished using the 'super-SILAC' technology, which I developed during my post-doctoral research. Using this technology, we identified over 12,000 proteins in formalin-fixed paraffin embedded breast cancer tumors. In the current project we will take one large step further, namely, microdissect and analyze selected regions in breast tumors based on local histopathological characteristics, such as the expression of known markers, cancer cell density, the vicinity to blood vessels and to the tumor invasive front. This ""topological map"" of the proteome will be followed by functional in vitro and in vivo studies, directly probing the aggressiveness of these cell populations, manifested by an accelerated proliferation and invasive/metastatic capacity. Finally, proteins associated with tumor aggressiveness will serve as blood-based biomarkers for predicting the tumorigenic phenotype using non-invasive tests. This work will set the basis for quantitative probing of tumor heterogeneity, which is crucial for accurate diagnosis and effective therapy.
"
Max ERC Funding
1 699 261 €
Duration
Start date: 2015-07-01, End date: 2020-06-30
Project acronym PROTEOMICS V3.0
Project Proteomics v3.0: Development, Implementation and Dissemination of a Third Generation Proteomics Technology
Researcher (PI) Rudolf Aebersold
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), LS2, ERC-2008-AdG
Summary Quantitative proteomics is a key technology for the life sciences in general and for systems biology in particular. So far, however, technical limitations have made it impossible to analyze the complete proteome of any species. It is the general goal of this proposal to develop, implement, apply and disseminate a new proteomic strategy that has the potential to generate quantitative proteomic datasets at an unprecedented depth, throughput, accuracy and robustness. Specifically, the new technology will identify and quantify every protein in a proteome. The title of the project Proteomics v3.0 was chosen to indicate the transformation of proteomics into its third phase, after 2D gel electrophoresis and LC-MS/MS based shotgun proteomics. Proteomics v3.0 is based on two sequential steps, emulating the strategy that has been immensely successful in the genomic sciences. In the first step the proteomic space is completely mapped out to generate a proteomic resource that is akin to the genomic sequence database. In the second step rapid and accurate assays will be developed to unambiguously identify and quantify any protein of the respective proteome in a multitude of samples. These assays will be made publicly accessible to support quantitative proteomic studies in the respective species. The strategy will first be implemented and tested in the yeast S. cerevisiae. In a later stage of the project it will be extended to the more complicated human proteome and include the development of assays that also probe the state of modification, splice forms and other types of protein variants generated by a specific open reading frame. Overall, the project will transform quantitative proteomics from a highly specialized technology practiced at a high level in a few laboratories worldwide into a commodity technology accessible, in principle to every group.
Summary
Quantitative proteomics is a key technology for the life sciences in general and for systems biology in particular. So far, however, technical limitations have made it impossible to analyze the complete proteome of any species. It is the general goal of this proposal to develop, implement, apply and disseminate a new proteomic strategy that has the potential to generate quantitative proteomic datasets at an unprecedented depth, throughput, accuracy and robustness. Specifically, the new technology will identify and quantify every protein in a proteome. The title of the project Proteomics v3.0 was chosen to indicate the transformation of proteomics into its third phase, after 2D gel electrophoresis and LC-MS/MS based shotgun proteomics. Proteomics v3.0 is based on two sequential steps, emulating the strategy that has been immensely successful in the genomic sciences. In the first step the proteomic space is completely mapped out to generate a proteomic resource that is akin to the genomic sequence database. In the second step rapid and accurate assays will be developed to unambiguously identify and quantify any protein of the respective proteome in a multitude of samples. These assays will be made publicly accessible to support quantitative proteomic studies in the respective species. The strategy will first be implemented and tested in the yeast S. cerevisiae. In a later stage of the project it will be extended to the more complicated human proteome and include the development of assays that also probe the state of modification, splice forms and other types of protein variants generated by a specific open reading frame. Overall, the project will transform quantitative proteomics from a highly specialized technology practiced at a high level in a few laboratories worldwide into a commodity technology accessible, in principle to every group.
Max ERC Funding
2 400 000 €
Duration
Start date: 2009-04-01, End date: 2014-03-31
Project acronym PROTEOMICS4D
Project Proteomics 4D: The proteome in context
Researcher (PI) Rudolf Aebersold
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), LS2, ERC-2014-ADG
Summary Elements operating in the context of a system generate results that are different from the simple addition of the results of each element. This notion is one of the basic tenants of systems science. In systems biology/medicine complex (disease) phenotypes arise from multiple interacting factors, specifically proteins. Yet, the biochemical and mechanistic base of complex phenotypes remain elusive.
An array of powerful genomic technologies including GWAS, WGS, transcriptomics, epigenetic analyses and proteomics have identified numerous factors that contribute to complex phenotypes. It can be expected that over the next few years, genetic factors contributing to specific complex phenotypes will be comprehensively identified, while their interactions will remain elusive.
The project “Proteomics 4D: The proteome in context “explores the concept, that complex phenotypes arise from the perturbation of modules of interacting proteins and that these modules integrate seemingly independent genomic variants into a single biochemical response. We will develop and apply a generic technology to directly measure the composition, topology and structure of wild type and genetically perturbed protein modules and relate structural changes to their functional output.
This will be achieved by a the integration of quantitative proteomic and phosphoproteomic technologies determining molecular phenotypes, and hybrid structural methods consisting of chemical cross-linking and mass spectrometry, cryoEM and computational data integration to probe structural perturbations.
The project will focus initially on the structural and functional effects of cancer associated mutations in protein kinase modules and then generalize to study perturbed modules in any tissue and disease state. The resources supporting this technology will be disseminated to catalyze a broad transformation of biology and molecular medicine towards the analysis of the proteome as a modular entity, the proteome in context.
Summary
Elements operating in the context of a system generate results that are different from the simple addition of the results of each element. This notion is one of the basic tenants of systems science. In systems biology/medicine complex (disease) phenotypes arise from multiple interacting factors, specifically proteins. Yet, the biochemical and mechanistic base of complex phenotypes remain elusive.
An array of powerful genomic technologies including GWAS, WGS, transcriptomics, epigenetic analyses and proteomics have identified numerous factors that contribute to complex phenotypes. It can be expected that over the next few years, genetic factors contributing to specific complex phenotypes will be comprehensively identified, while their interactions will remain elusive.
The project “Proteomics 4D: The proteome in context “explores the concept, that complex phenotypes arise from the perturbation of modules of interacting proteins and that these modules integrate seemingly independent genomic variants into a single biochemical response. We will develop and apply a generic technology to directly measure the composition, topology and structure of wild type and genetically perturbed protein modules and relate structural changes to their functional output.
This will be achieved by a the integration of quantitative proteomic and phosphoproteomic technologies determining molecular phenotypes, and hybrid structural methods consisting of chemical cross-linking and mass spectrometry, cryoEM and computational data integration to probe structural perturbations.
The project will focus initially on the structural and functional effects of cancer associated mutations in protein kinase modules and then generalize to study perturbed modules in any tissue and disease state. The resources supporting this technology will be disseminated to catalyze a broad transformation of biology and molecular medicine towards the analysis of the proteome as a modular entity, the proteome in context.
Max ERC Funding
2 208 150 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym PROTEOTOXNET
Project Unraveling the cellular responses to aberrantly-folded and aggregated proteins
Researcher (PI) Paola Picotti
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Starting Grant (StG), LS2, ERC-2013-StG
Summary Protein aggregation diseases are associated with the intracellular accumulation of specific misfolded protein aggregates, that are directly implicated in cellular dysfunction. A range of human disorders of previously unknown nature now falls into this category. The mechanisms of aggregation-induced cell degeneration are however unclear, resulting in poor therapeutic strategies. In this project we will characterize the network of cellular responses to a set of disease-related aggregation-prone proteins (APPs) and determine how to efficiently modulate proteotoxicity. In a discovery phase, we will use proteome-wide screens to identify functional modules that sequentially respond to APP aggregation. A novel method to probe conformational changes of proteins in their biological matrix will be applied to quantify concomitant changes in APP conformation during the toxicity cascade. Our eminently quantitative approach will allow us to characterize the kinetics of intracellular aggregate formation and the resulting cellular responses. A set of biological markers for different functional modules and toxicity stages will be measured using a selected reaction monitoring-fingerprint assay, through a set of genetic and chemical modulators of proteotoxicity. This will unravel how modulators rewire or compensate for the deregulated networks and suggest their most efficient combinations. The multi-level information will be iteratively integrated with prior data and network structure. Emerging hypotheses will be tested using a combination of genetic and biochemical tools and targeted proteomic experiments. This project features novel concepts and methods and will allow us to unravel the molecular events accompanying the onset of proteinopathies and their modulation. It will positively impact biomedical research on protein aggregation diseases, guide identification of suitable therapeutic strategies and advance our understanding of the biology of protein aggregation.
Summary
Protein aggregation diseases are associated with the intracellular accumulation of specific misfolded protein aggregates, that are directly implicated in cellular dysfunction. A range of human disorders of previously unknown nature now falls into this category. The mechanisms of aggregation-induced cell degeneration are however unclear, resulting in poor therapeutic strategies. In this project we will characterize the network of cellular responses to a set of disease-related aggregation-prone proteins (APPs) and determine how to efficiently modulate proteotoxicity. In a discovery phase, we will use proteome-wide screens to identify functional modules that sequentially respond to APP aggregation. A novel method to probe conformational changes of proteins in their biological matrix will be applied to quantify concomitant changes in APP conformation during the toxicity cascade. Our eminently quantitative approach will allow us to characterize the kinetics of intracellular aggregate formation and the resulting cellular responses. A set of biological markers for different functional modules and toxicity stages will be measured using a selected reaction monitoring-fingerprint assay, through a set of genetic and chemical modulators of proteotoxicity. This will unravel how modulators rewire or compensate for the deregulated networks and suggest their most efficient combinations. The multi-level information will be iteratively integrated with prior data and network structure. Emerging hypotheses will be tested using a combination of genetic and biochemical tools and targeted proteomic experiments. This project features novel concepts and methods and will allow us to unravel the molecular events accompanying the onset of proteinopathies and their modulation. It will positively impact biomedical research on protein aggregation diseases, guide identification of suitable therapeutic strategies and advance our understanding of the biology of protein aggregation.
Max ERC Funding
1 500 000 €
Duration
Start date: 2014-03-01, End date: 2019-06-30
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 RAPLODAPT
Project Ploidy change as a rapid mechanism of adaptation
Researcher (PI) Judith Berman
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Advanced Grant (AdG), LS2, ERC-2013-ADG
Summary Fungi are particularly challenging pathogens; because they and their human hosts are eukaryotes. We will study how new traits such as drugR arise rapidly using Candida albicans, the most prevalent fungal pathogen of humans. The work explores the ground-breaking concept that alterations in genome ploidy are prevalent in drugR isolates because these genome states promote persistence and drug resistance.
We recently found that C. albicans, thought to be an “obligate diploid”, can form haploids. This represents a major paradigm shift both technologically and conceptually. We are poised to exploit this unique opportunity to reinvent genomic approaches for C. albicans by leveraging next generation sequencing, high throughput analyses and more traditional genetics. Because haploids are much less fit than heterozygous diploids, our working hypothesis is that changes in ploidy, including whole genome ploidy and aneuploidy, occur frequently under drug stress and that they make major contributions to the rapid appearance of genotypic and phenotypic diversity, in part by promoting persistence.
The objectives of this proposal are to develop next-generation technologies that leverage haploids; to characterize the conditions and genes that promote ploidy transitions, especially in the presence of drug, in vitro and in vivo and to analyze their fitness consequences. This multi-disciplinary research program will integrate = approaches at the genetic, genomic, molecular, cellular and population levels and includes computational approaches to model evolutionary processes.
The project will lead to unparalleled advances in tools for the research community, and important insights concerning how diversity arises rapidly. It will assist in efforts to design diagnostic and therapeutic strategies for preventing and treating fungal diseases, prividing insights into the rapid appearance of drug resistance in eukaryotic pathogens, and chemotherapy resistance in cancer cells.
Summary
Fungi are particularly challenging pathogens; because they and their human hosts are eukaryotes. We will study how new traits such as drugR arise rapidly using Candida albicans, the most prevalent fungal pathogen of humans. The work explores the ground-breaking concept that alterations in genome ploidy are prevalent in drugR isolates because these genome states promote persistence and drug resistance.
We recently found that C. albicans, thought to be an “obligate diploid”, can form haploids. This represents a major paradigm shift both technologically and conceptually. We are poised to exploit this unique opportunity to reinvent genomic approaches for C. albicans by leveraging next generation sequencing, high throughput analyses and more traditional genetics. Because haploids are much less fit than heterozygous diploids, our working hypothesis is that changes in ploidy, including whole genome ploidy and aneuploidy, occur frequently under drug stress and that they make major contributions to the rapid appearance of genotypic and phenotypic diversity, in part by promoting persistence.
The objectives of this proposal are to develop next-generation technologies that leverage haploids; to characterize the conditions and genes that promote ploidy transitions, especially in the presence of drug, in vitro and in vivo and to analyze their fitness consequences. This multi-disciplinary research program will integrate = approaches at the genetic, genomic, molecular, cellular and population levels and includes computational approaches to model evolutionary processes.
The project will lead to unparalleled advances in tools for the research community, and important insights concerning how diversity arises rapidly. It will assist in efforts to design diagnostic and therapeutic strategies for preventing and treating fungal diseases, prividing insights into the rapid appearance of drug resistance in eukaryotic pathogens, and chemotherapy resistance in cancer cells.
Max ERC Funding
2 365 000 €
Duration
Start date: 2014-01-01, End date: 2019-12-31
Project acronym RAVEN
Project Rapid mass loss of debris covered glaciers in High Mountain Asia
Researcher (PI) Francesca PELLICCIOTTI
Host Institution (HI) EIDGENOESSISCHE FORSCHUNGSANSTALT WSL
Call Details Consolidator Grant (CoG), PE10, ERC-2017-COG
Summary The research proposed uses an integrated data-modelling approach to elucidate the role that debris-covered glaciers play in the water cycle of High Mountain Asia (HMA) and establish how future HMA glacier and runoff will evolve. Debris-covered glaciers are of great significance for the hydrology of HMA, with large contributions to headwater streamflow. Despite this, their mass balance, hydrological role and future changes are poorly constrained, challenging model predictions of future water resources. Debris mantles insulate the ice and reduce ablation, but large-scale research indicates that HMA debris-covered glaciers are losing mass at rates similar to debris-free glaciers. This anomalous behaviour has profound implications for future glacier mass balance and runoff, but has not been reproduced with models, a fundamental limitation to a global assessment. I aim to establish that: 1) supraglacial cliffs and ponds are responsible for higher than expected mass losses of HMA debris-covered glaciers, because they act as windows of energy transfer through the debris; and that 2) their inclusion into models of glacier evolution will provide essential new estimates of glacier changes and future water availability in HMA. RAVEN will achieve these aims through combination of high-resolution satellite observations, field data and physically-based models in four sites along the Himalayan arc. This unprecedented setup captures the variety of climate and glaciers across HMA. Using satellite images I will investigate the spatial distribution and temporal evolution of cliffs and ponds; the insights will be used to develop physically-based models of cliff and pond ablation, which will be included in a glacio-hydrological model. Future glacier and runoff response will be projected using downscaled climate scenarios, allowing new estimates of glacier changes and future runoff for a data-starved region where millions of people depend on the water resources from glaciers and snow.
Summary
The research proposed uses an integrated data-modelling approach to elucidate the role that debris-covered glaciers play in the water cycle of High Mountain Asia (HMA) and establish how future HMA glacier and runoff will evolve. Debris-covered glaciers are of great significance for the hydrology of HMA, with large contributions to headwater streamflow. Despite this, their mass balance, hydrological role and future changes are poorly constrained, challenging model predictions of future water resources. Debris mantles insulate the ice and reduce ablation, but large-scale research indicates that HMA debris-covered glaciers are losing mass at rates similar to debris-free glaciers. This anomalous behaviour has profound implications for future glacier mass balance and runoff, but has not been reproduced with models, a fundamental limitation to a global assessment. I aim to establish that: 1) supraglacial cliffs and ponds are responsible for higher than expected mass losses of HMA debris-covered glaciers, because they act as windows of energy transfer through the debris; and that 2) their inclusion into models of glacier evolution will provide essential new estimates of glacier changes and future water availability in HMA. RAVEN will achieve these aims through combination of high-resolution satellite observations, field data and physically-based models in four sites along the Himalayan arc. This unprecedented setup captures the variety of climate and glaciers across HMA. Using satellite images I will investigate the spatial distribution and temporal evolution of cliffs and ponds; the insights will be used to develop physically-based models of cliff and pond ablation, which will be included in a glacio-hydrological model. Future glacier and runoff response will be projected using downscaled climate scenarios, allowing new estimates of glacier changes and future runoff for a data-starved region where millions of people depend on the water resources from glaciers and snow.
Max ERC Funding
2 000 000 €
Duration
Start date: 2018-05-01, End date: 2023-04-30
Project acronym ReaDMe
Project Readout of DNA methylation
Researcher (PI) Dirk Schubeler
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Call Details Advanced Grant (AdG), LS2, ERC-2014-ADG
Summary DNA and chromatin modifications are essential for proper control of gene expression during development. How these marks alter transcriptional programs and modulate binding patterns of sequence specific transcription factors (TF) remains poorly understood. This currently limits our interpretation of epigenomic maps towards their incorporation into predictive models of gene regulation.
ReaDMe has the ambitious goal to systematically define the sensitivity of TFs to local levels of DNA methylation in vivo. We will use a combination of genomics, genome editing and proteomics tools to comprehensively identify transcriptional regulators that respond to DNA methylation. As a first approach, we will interrogate changes in the global TF binding landscape when DNA methylation is ablated from the genome. Using both embryonic stem cells and somatic cells, these experiments are aimed at identifying sites that are occupied by TFs in a DNA methylation dependent manner within different cellular context. Secondly, we will combine parallelized chromosomal insertions with targeted footprinting to determine the link between DNA sequence context, methylation density and TF binding. In a third approach we will define the global chromatin proteome as a function of DNA methylation. Through the use of a novel and orthogonal proteomics assay, we will characterize DNA methylation sensitive changes in the chromatin-bound proteome. Candidate factors predicted from all approaches will be validated and functionally characterized through direct genome-wide mapping as well as loss of function analysis.
ERC funding would enable ReaDMe to develop an integrated setup to in vivo identify and characterize where DNA methylation influences the cis-regulatory landscape by modulating binding profiles of trans-acting factors. This goal represents a crucial step towards comprehensive understanding of the genomic readout of DNA methylation and its impact on gene regulation.
Summary
DNA and chromatin modifications are essential for proper control of gene expression during development. How these marks alter transcriptional programs and modulate binding patterns of sequence specific transcription factors (TF) remains poorly understood. This currently limits our interpretation of epigenomic maps towards their incorporation into predictive models of gene regulation.
ReaDMe has the ambitious goal to systematically define the sensitivity of TFs to local levels of DNA methylation in vivo. We will use a combination of genomics, genome editing and proteomics tools to comprehensively identify transcriptional regulators that respond to DNA methylation. As a first approach, we will interrogate changes in the global TF binding landscape when DNA methylation is ablated from the genome. Using both embryonic stem cells and somatic cells, these experiments are aimed at identifying sites that are occupied by TFs in a DNA methylation dependent manner within different cellular context. Secondly, we will combine parallelized chromosomal insertions with targeted footprinting to determine the link between DNA sequence context, methylation density and TF binding. In a third approach we will define the global chromatin proteome as a function of DNA methylation. Through the use of a novel and orthogonal proteomics assay, we will characterize DNA methylation sensitive changes in the chromatin-bound proteome. Candidate factors predicted from all approaches will be validated and functionally characterized through direct genome-wide mapping as well as loss of function analysis.
ERC funding would enable ReaDMe to develop an integrated setup to in vivo identify and characterize where DNA methylation influences the cis-regulatory landscape by modulating binding profiles of trans-acting factors. This goal represents a crucial step towards comprehensive understanding of the genomic readout of DNA methylation and its impact on gene regulation.
Max ERC Funding
2 136 969 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym RECONMET
Project Reconstruction of methane flux from lakes: development and application of a new approach
Researcher (PI) Oliver Heiri
Host Institution (HI) UNIVERSITAET BERN
Call Details Starting Grant (StG), PE10, ERC-2009-StG
Summary Reconstruction of methane flux from lakes: development and application of a new approach
Summary
Reconstruction of methane flux from lakes: development and application of a new approach
Max ERC Funding
1 554 000 €
Duration
Start date: 2009-12-01, End date: 2014-11-30
Project acronym RegRNA
Project Mechanistic principles of regulation by small RNAs
Researcher (PI) Hanah Margalit
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS2, ERC-2018-ADG
Summary Small RNAs (sRNAs) are major regulators of gene expression in bacteria, exerting their regulation in trans by base pairing with target RNAs. Traditionally, sRNAs were considered post-transcriptional regulators, mainly regulating translation by blocking or exposing the ribosome binding site. However, accumulating evidence suggest that sRNAs can exploit the base pairing to manipulate their targets in different ways, assisting or interfering with various molecular processes involving the target RNA. Currently there are a few examples of these alternative regulation modes, but their extent and implications in the cellular circuitry have not been assessed. Here we propose to take advantage of the power of RNA-seq-based technologies to develop innovative approaches to address these challenges transcriptome-wide. These approaches will enable us to map the regulatory mechanism a sRNA employs per target through its effect on a certain molecular process. For feasibility we propose studying three processes: RNA cleavage by RNase E, pre-mature Rho-dependent transcription termination, and transcription elongation pausing. Finding targets regulated by sRNA manipulation of the two latter processes would be especially intriguing, as it would suggest that sRNAs can function as gene-specific transcription regulators (alluded to by our preliminary results). As a basis of our research we will use the network of ~2400 sRNA-target pairs in Escherichia coli, deciphered by RIL-seq (a method we recently developed for global in vivo detection of sRNA targets). Revealing the regulatory mechanism(s) employed per target will shed light on the principles underlying the integration of distinct sRNA regulation modes in specific regulatory circuits and cellular contexts, with direct implications to synthetic biology and pathogenic bacteria. Our study may change the way sRNAs are perceived, from post-transcriptional to versatile regulators that apply different regulation modes to different targets.
Summary
Small RNAs (sRNAs) are major regulators of gene expression in bacteria, exerting their regulation in trans by base pairing with target RNAs. Traditionally, sRNAs were considered post-transcriptional regulators, mainly regulating translation by blocking or exposing the ribosome binding site. However, accumulating evidence suggest that sRNAs can exploit the base pairing to manipulate their targets in different ways, assisting or interfering with various molecular processes involving the target RNA. Currently there are a few examples of these alternative regulation modes, but their extent and implications in the cellular circuitry have not been assessed. Here we propose to take advantage of the power of RNA-seq-based technologies to develop innovative approaches to address these challenges transcriptome-wide. These approaches will enable us to map the regulatory mechanism a sRNA employs per target through its effect on a certain molecular process. For feasibility we propose studying three processes: RNA cleavage by RNase E, pre-mature Rho-dependent transcription termination, and transcription elongation pausing. Finding targets regulated by sRNA manipulation of the two latter processes would be especially intriguing, as it would suggest that sRNAs can function as gene-specific transcription regulators (alluded to by our preliminary results). As a basis of our research we will use the network of ~2400 sRNA-target pairs in Escherichia coli, deciphered by RIL-seq (a method we recently developed for global in vivo detection of sRNA targets). Revealing the regulatory mechanism(s) employed per target will shed light on the principles underlying the integration of distinct sRNA regulation modes in specific regulatory circuits and cellular contexts, with direct implications to synthetic biology and pathogenic bacteria. Our study may change the way sRNAs are perceived, from post-transcriptional to versatile regulators that apply different regulation modes to different targets.
Max ERC Funding
2 278 125 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym REGULATORYCIRCUITS
Project Novel Systematic Strategies for Elucidating Cellular Regulatory Circuits
Researcher (PI) Nir Friedman
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS2, ERC-2008-AdG
Summary The precise regulation of gene expression has been the subject of extensive scrutiny. Nonetheless, there is a big gap between genomic characterization of transcriptional responses and our predictions based on known molecular mechanisms and networks and of transcription regulation. In this proposal I argue for an approach to bridge this gap by using a novel experimental strategy that exploits the recent maturation of two technologies: the use of fluorescence reporter techniques to monitor promoter activity and high-throughput genetic manipulations for the construction of combinatorial genetic perturbations. By combining these, we will screen for genes that modulate the transcriptional response of target promoters, use genetic interactions between them to better resolve their functional dependencies, and build detailed quantitative models of transcriptional processes. We will use the budding yeast model organism, which allows for efficient manipulations, to dissect two transcriptional responses that are prototypical of many regulatory networks in living cells: [1] The early response to osmotic stress, which is mediated by at least two signaling pathways and multiple transcription factors, and [2] the central carbon metabolism response to shifts in carbon source, which involves multiple sensing and signaling pathways to maintain homeostasis. Our approach will elucidate mechanisms that are opaque to classical screens and facilitate building detailed predictive models of these responses. These results will lead to understanding of general principles that govern transcriptional networks. This is the first approach to comprehensively characterize the molecular mechanisms that modulate a transcriptional response, and arrange them in a coherent network. It will open many questions for detailed biochemical investigations, as well as set the stage to extend these ideas to use more detailed phenotypic assays and in more complex organisms.
Summary
The precise regulation of gene expression has been the subject of extensive scrutiny. Nonetheless, there is a big gap between genomic characterization of transcriptional responses and our predictions based on known molecular mechanisms and networks and of transcription regulation. In this proposal I argue for an approach to bridge this gap by using a novel experimental strategy that exploits the recent maturation of two technologies: the use of fluorescence reporter techniques to monitor promoter activity and high-throughput genetic manipulations for the construction of combinatorial genetic perturbations. By combining these, we will screen for genes that modulate the transcriptional response of target promoters, use genetic interactions between them to better resolve their functional dependencies, and build detailed quantitative models of transcriptional processes. We will use the budding yeast model organism, which allows for efficient manipulations, to dissect two transcriptional responses that are prototypical of many regulatory networks in living cells: [1] The early response to osmotic stress, which is mediated by at least two signaling pathways and multiple transcription factors, and [2] the central carbon metabolism response to shifts in carbon source, which involves multiple sensing and signaling pathways to maintain homeostasis. Our approach will elucidate mechanisms that are opaque to classical screens and facilitate building detailed predictive models of these responses. These results will lead to understanding of general principles that govern transcriptional networks. This is the first approach to comprehensively characterize the molecular mechanisms that modulate a transcriptional response, and arrange them in a coherent network. It will open many questions for detailed biochemical investigations, as well as set the stage to extend these ideas to use more detailed phenotypic assays and in more complex organisms.
Max ERC Funding
2 199 899 €
Duration
Start date: 2009-01-01, End date: 2013-12-31
Project acronym REpiReg
Project RNAi-mediated Epigenetic Gene Regulation
Researcher (PI) Marc Bühler
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Call Details Consolidator Grant (CoG), LS2, ERC-2015-CoG
Summary RNAi refers to the ability of small RNAs to silence expression of homologous sequences. A surprising link between epigenetics and RNAi was discovered more than a decade ago, and I was fortunate enough to be involved in this exciting field of research from the beginning. It is now well established that endogenous small RNAs have a direct impact on the genome in various organisms. Yet, the initiation of chromatin modifications in trans by exogenously introduced small RNAs has been inherently difficult to achieve in all eukaryotic cells. This has sparked controversy about the importance and conservation of RNAi-mediated epigenome regulation and hampered systematic mechanistic dissection of this phenomenon.
Using fission yeast, we have discovered a counter-acting mechanism that impedes small RNA-directed formation of heterochromatin and constitutes the foundation of this proposal. Our goal is to close several knowledge gaps and test the intriguing possibility that the suppressive mechanism we discovered is conserved in mammalian cells. We will employ yeast and embryonic stem cells and use cutting-edge technologies (i.e., chemical mutagenesis combined with whole-genome sequencing, genome editing with engineered nucleases, and single-cell RNA sequencing) to address fundamental, as yet unanswered questions.
My proposal consists of four major aims. In aim 1, I propose to use proteomics approaches and to perform yeast genetic screens to define additional pathway components and regulatory factors. Aim 2 builds on our ability to finally trigger de novo formation of heterochromatin by synthetic siRNAs acting in trans, addressing many of the outstanding mechanistic questions that could not be addressed in the past. In Aims 3 and 4, experiments conducted in yeast and mouse cells will elucidate missing fragments critical to our understanding of the conserved principles behind RNAi-mediated epigenetic gene regulation.
Summary
RNAi refers to the ability of small RNAs to silence expression of homologous sequences. A surprising link between epigenetics and RNAi was discovered more than a decade ago, and I was fortunate enough to be involved in this exciting field of research from the beginning. It is now well established that endogenous small RNAs have a direct impact on the genome in various organisms. Yet, the initiation of chromatin modifications in trans by exogenously introduced small RNAs has been inherently difficult to achieve in all eukaryotic cells. This has sparked controversy about the importance and conservation of RNAi-mediated epigenome regulation and hampered systematic mechanistic dissection of this phenomenon.
Using fission yeast, we have discovered a counter-acting mechanism that impedes small RNA-directed formation of heterochromatin and constitutes the foundation of this proposal. Our goal is to close several knowledge gaps and test the intriguing possibility that the suppressive mechanism we discovered is conserved in mammalian cells. We will employ yeast and embryonic stem cells and use cutting-edge technologies (i.e., chemical mutagenesis combined with whole-genome sequencing, genome editing with engineered nucleases, and single-cell RNA sequencing) to address fundamental, as yet unanswered questions.
My proposal consists of four major aims. In aim 1, I propose to use proteomics approaches and to perform yeast genetic screens to define additional pathway components and regulatory factors. Aim 2 builds on our ability to finally trigger de novo formation of heterochromatin by synthetic siRNAs acting in trans, addressing many of the outstanding mechanistic questions that could not be addressed in the past. In Aims 3 and 4, experiments conducted in yeast and mouse cells will elucidate missing fragments critical to our understanding of the conserved principles behind RNAi-mediated epigenetic gene regulation.
Max ERC Funding
1 998 557 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym RINEC
Project River networks as ecological corridors for biodiversity, populations and waterborne disease (RINEC)
Researcher (PI) Andrea Rinaldo
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), PE10, ERC-2008-AdG
Summary The proposal hinges on the noteworthy scientific perspectives provided by ecohydrological studies of river basins,seen as a natural laboratory for complex system perspectives integrating hydrologic, ecological and geomorphological dynamics. Moving from morphological and functional analyses of dendritic geometries observed in Nature over a wide range of scales,my claim is that essential processes sustaining human life and societies taking place along dendritic structures can be predicted. Population migrations and human settlements historically proceeded along river networks to follow water supply routes. Riparian systems,critically important ecosystems positioned along streams and rivers, play crucial roles in their watersheds,including nutrient filtering, biogeochemical processing,shade and resource provisioning, and stream bank stabilization. Devastating water-borne disease,such as cholera, and invading foreign species spread through water bodies linked by river networks. Although the dynamics of such systems has been extensively studied, existing approaches were mostly within the framework of mean-field or two-dimensional landscapes that ignore directionality of dispersal implied by the network acting as environmental matrix. How does connectivity within a a river network affect the emergent spreading of water-borne infections? Does the river basin act as a template for biodiversity? Are there hydrologic controls on the spreading of water-borne disease? To answer such questions, the present proposal addresses the study of biodiversity in the river basin (freshwater fish and riparian vegetation); cholera dynamics and zebra mussel invasions along river networks. Observational data and theoretical models, in a comparative mode,will be analyzed within a unified theoretical framework.This is intended to prove of crucial interest for understanding the functioning of river basins as a whole,including its ecosystem structure and function.
Summary
The proposal hinges on the noteworthy scientific perspectives provided by ecohydrological studies of river basins,seen as a natural laboratory for complex system perspectives integrating hydrologic, ecological and geomorphological dynamics. Moving from morphological and functional analyses of dendritic geometries observed in Nature over a wide range of scales,my claim is that essential processes sustaining human life and societies taking place along dendritic structures can be predicted. Population migrations and human settlements historically proceeded along river networks to follow water supply routes. Riparian systems,critically important ecosystems positioned along streams and rivers, play crucial roles in their watersheds,including nutrient filtering, biogeochemical processing,shade and resource provisioning, and stream bank stabilization. Devastating water-borne disease,such as cholera, and invading foreign species spread through water bodies linked by river networks. Although the dynamics of such systems has been extensively studied, existing approaches were mostly within the framework of mean-field or two-dimensional landscapes that ignore directionality of dispersal implied by the network acting as environmental matrix. How does connectivity within a a river network affect the emergent spreading of water-borne infections? Does the river basin act as a template for biodiversity? Are there hydrologic controls on the spreading of water-borne disease? To answer such questions, the present proposal addresses the study of biodiversity in the river basin (freshwater fish and riparian vegetation); cholera dynamics and zebra mussel invasions along river networks. Observational data and theoretical models, in a comparative mode,will be analyzed within a unified theoretical framework.This is intended to prove of crucial interest for understanding the functioning of river basins as a whole,including its ecosystem structure and function.
Max ERC Funding
1 146 200 €
Duration
Start date: 2009-01-01, End date: 2013-12-31
Project acronym RNAflashbacks
Project The Neuronal Code of Inheritance
Researcher (PI) Oded Rechavi
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Consolidator Grant (CoG), LS2, ERC-2018-COG
Summary Little is known about non-DNA mediated transgenerational inheritance of parental responses. If inheritance of non-genetic materials is prevalent, it could challenge our conceptions regarding the rules and limits of heredity. We are in particularly intrigued by the possibility that the nervous system can produce heritable changes. Mechanisms for propagation of responses from the soma to the germline are known. Until very recently, the possibility that any type of environmental response could become heritable, let alone somatic responses, was considered blasphemous. In C. elegans nematodes, exogenously-supplied artificial dsRNA transfers from the soma to the germline, triggering transgenerational small RNA-mediated RNA interference. It is unknown whether endogenous small RNAs can transmit specific information about the environment to the progeny. We will investigate if endogenous siRNAs, which are naturally made in somatic tissues, and in particular in neurons, produce transgenerational responses. Specifically, we will test which RNA molecules act transgenerationally, how do they mediate non-cell autonomous gene regulation, and which responses can be communicated to the progeny. What is the code that transforms particular environmental responses to specific arsenals of heritable RNA molecules? We will answer these questions, and moreover study the implications that this completely new form of hereditary has for the offspring’s survival. Can heritable small RNAs retain adaptive memory? Not only will we elucidate natural transmission of the parents’ activity from generation-to-generation, we will moreover devise means to control these mechanisms. We will engineer tools to diagnose, erase, maintain, and modulate the heritable effects, which would be important for basic research and hopefully also translational in the future.
Summary
Little is known about non-DNA mediated transgenerational inheritance of parental responses. If inheritance of non-genetic materials is prevalent, it could challenge our conceptions regarding the rules and limits of heredity. We are in particularly intrigued by the possibility that the nervous system can produce heritable changes. Mechanisms for propagation of responses from the soma to the germline are known. Until very recently, the possibility that any type of environmental response could become heritable, let alone somatic responses, was considered blasphemous. In C. elegans nematodes, exogenously-supplied artificial dsRNA transfers from the soma to the germline, triggering transgenerational small RNA-mediated RNA interference. It is unknown whether endogenous small RNAs can transmit specific information about the environment to the progeny. We will investigate if endogenous siRNAs, which are naturally made in somatic tissues, and in particular in neurons, produce transgenerational responses. Specifically, we will test which RNA molecules act transgenerationally, how do they mediate non-cell autonomous gene regulation, and which responses can be communicated to the progeny. What is the code that transforms particular environmental responses to specific arsenals of heritable RNA molecules? We will answer these questions, and moreover study the implications that this completely new form of hereditary has for the offspring’s survival. Can heritable small RNAs retain adaptive memory? Not only will we elucidate natural transmission of the parents’ activity from generation-to-generation, we will moreover devise means to control these mechanisms. We will engineer tools to diagnose, erase, maintain, and modulate the heritable effects, which would be important for basic research and hopefully also translational in the future.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-08-01, End date: 2024-07-31
Project acronym RNAiGenReg
Project RNAi-mediated genome regulation
Researcher (PI) Marc Bühler
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Call Details Starting Grant (StG), LS2, ERC-2011-StG_20101109
Summary RNA interference (RNAi) is a highly conserved, sequence-specific gene regulatory mechanism among eukaryotes. It is critical for a variety of important biological functions and is being pursued as a promising new tool for the treatment of a variety of human maladies. A surprising link between heterochromatin and the RNAi pathway was discovered a few years ago in fission yeast and plants, and similar mechanisms have more recently been described in various eukaryotes. However, to what extent the mechanisms we have been studying in yeast are conserved up to humans remains unknown. The goal of this proposal is to further our understanding of RNAi-mediated heterochromatin assembly by using fission yeast as a model organism, but also to investigate to role of RNAi in the nucleus of human cells.
My proposal consists of three major aims. In aim 1 I propose to combine light and electron microscopy to address important and largely unanswered questions such as subcellular localization and temporal regulation of the RNAi pathway. Aim 2 builds on our recent discovery that RNAi factors physically associate with chromatin to control genome activity also outside constitutive heterochromatin. I am proposing experiments in fission yeast that aim at understanding the biological role of this new mode of genome regulation and its mechanistic dissection . However, we will also extend our analysis to human cells which will shed new light on the role of the RNAi pathway in the nucleus of higher eukaryotes. Finally, we are aiming at identifying the features a target locus in the S. pombe genome must have to become susceptible to RNAi-mediated silencing at the level of chromatin. Thus, the outcome of these experiments may substantially influence the developments of siRNA-based therapeutics.
Summary
RNA interference (RNAi) is a highly conserved, sequence-specific gene regulatory mechanism among eukaryotes. It is critical for a variety of important biological functions and is being pursued as a promising new tool for the treatment of a variety of human maladies. A surprising link between heterochromatin and the RNAi pathway was discovered a few years ago in fission yeast and plants, and similar mechanisms have more recently been described in various eukaryotes. However, to what extent the mechanisms we have been studying in yeast are conserved up to humans remains unknown. The goal of this proposal is to further our understanding of RNAi-mediated heterochromatin assembly by using fission yeast as a model organism, but also to investigate to role of RNAi in the nucleus of human cells.
My proposal consists of three major aims. In aim 1 I propose to combine light and electron microscopy to address important and largely unanswered questions such as subcellular localization and temporal regulation of the RNAi pathway. Aim 2 builds on our recent discovery that RNAi factors physically associate with chromatin to control genome activity also outside constitutive heterochromatin. I am proposing experiments in fission yeast that aim at understanding the biological role of this new mode of genome regulation and its mechanistic dissection . However, we will also extend our analysis to human cells which will shed new light on the role of the RNAi pathway in the nucleus of higher eukaryotes. Finally, we are aiming at identifying the features a target locus in the S. pombe genome must have to become susceptible to RNAi-mediated silencing at the level of chromatin. Thus, the outcome of these experiments may substantially influence the developments of siRNA-based therapeutics.
Max ERC Funding
1 599 992 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym SAMIT
Project Systems Analysis of Plant Metabolism through the Integration of Heterogeneous Data from Genetics, Informatics and Metabolomics
Researcher (PI) Asaph Aharoni
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2007-StG
Summary "The term METABOLISM describes all the chemical reactions and interactions that take place in a biological system. The regulation of metabolic pathways is constantly tuned in order to suit the needs of development and fitness. The main goal of the proposed research is to unravel networks of genes and proteins which coordinate the activity of metabolic pathways during plant development and stress response. In Phase I of the project, an infrastructure will be set-up that includes metabolite analyses technologies, a large tomato mutant population and a collection of tomato transcription factor genes. In Phase II, the population will be screened for novel mutants and genes associated with the research interests of the lab. The screens will include: a) visual screening, b) HTP, non laborious assays, c) non-targeted metabolite analysis for the detection of differential metabolites using LC-MS, and d) screening by a reverse genetic approach (from sequence to mutant) through transposon display. The collection of transcription factors will be used for rapid gene function analysis by means of Virus Induced Gene silencing (VIGS), and for the generation of a tomato transcription factors ""Interactome"" map. In Phase III, selected genes and mutants will be subjected to a detailed characterization including: a) the regulatory networks controlling fruit development, b) the regulation of metabolic pathways associated with plant surface metabolism and c) the regulation of the Isoprenoid pathway (including the glycoalkaloids and carotenoids). Data gathered from diverse platforms and screens will be integrated using computational tools to provide new knowledge on the genetic control of metabolic pathways that is currently very limited. This research addresses a major challenge, namely, the extensive acquisition of an heterogeneous set of data (genetic, gene expression, protein interaction and metabolic) and their integration to identify regulatory networks controlling plant metabolism."
Summary
"The term METABOLISM describes all the chemical reactions and interactions that take place in a biological system. The regulation of metabolic pathways is constantly tuned in order to suit the needs of development and fitness. The main goal of the proposed research is to unravel networks of genes and proteins which coordinate the activity of metabolic pathways during plant development and stress response. In Phase I of the project, an infrastructure will be set-up that includes metabolite analyses technologies, a large tomato mutant population and a collection of tomato transcription factor genes. In Phase II, the population will be screened for novel mutants and genes associated with the research interests of the lab. The screens will include: a) visual screening, b) HTP, non laborious assays, c) non-targeted metabolite analysis for the detection of differential metabolites using LC-MS, and d) screening by a reverse genetic approach (from sequence to mutant) through transposon display. The collection of transcription factors will be used for rapid gene function analysis by means of Virus Induced Gene silencing (VIGS), and for the generation of a tomato transcription factors ""Interactome"" map. In Phase III, selected genes and mutants will be subjected to a detailed characterization including: a) the regulatory networks controlling fruit development, b) the regulation of metabolic pathways associated with plant surface metabolism and c) the regulation of the Isoprenoid pathway (including the glycoalkaloids and carotenoids). Data gathered from diverse platforms and screens will be integrated using computational tools to provide new knowledge on the genetic control of metabolic pathways that is currently very limited. This research addresses a major challenge, namely, the extensive acquisition of an heterogeneous set of data (genetic, gene expression, protein interaction and metabolic) and their integration to identify regulatory networks controlling plant metabolism."
Max ERC Funding
1 009 000 €
Duration
Start date: 2008-07-01, End date: 2013-06-30
Project acronym scAssembly
Project Algorithms and experimental tools for integrating very large-scale single cell genomics data
Researcher (PI) Amos TANAY
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS2, ERC-2016-COG
Summary Robust and flexible tissue- and cell-type specific gene regulation is a definitive prerequisite for complex function in any multi-cellular organism. Modern genomics and epigenomics provide us with catalogues of gene regulatory elements and maps illustrating their activity in different tissues. Nevertheless, we are far from being able to explain emergence and maintenance of cellular states from such data, partly because we so far lacked characterization of individual molecular states and genome control mechanisms at their native resolution - the single cell. Recently, new approaches developed by the single cell genomics community, with several contributions from our group, allow massive acquisition of data on the transcriptional, epigenomic and chromosomal conformation states in large cohorts of single cells. In this research program, we aim to move forward rapidly to bridge a major gap between these experimental breakthroughs and models of genome regulation in complex tissues. We will develop algorithms and models for representing data the transcriptional profiles, DNA methylation landscapes and Hi-C maps of literally millions of cells. Our tools will be designed specifically to leverage on new single cell RNA-seq, single cell Hi-C, single cell capture-pBat and higher order 4C-seq that we will continue to develop experimentally. Furthermore, we shall enhance and optimize our interdisciplinary framework hand in hand with a working model aiming at unprecedentedly comprehensive single cell analysis of E8-E10 mouse embryos. This will provide us with hundreds of worked-out cases of tissue specific gene regulation. The techniques and insights from these studies will then be used to characterize cell type aberrations and epigenetic reprogramming in tumors. The open algorithms, techniques and methodology we shall develop can accelerate research in multiple groups that will utilize single cell genomics to study numerous questions on gene regulation in the coming years.
Summary
Robust and flexible tissue- and cell-type specific gene regulation is a definitive prerequisite for complex function in any multi-cellular organism. Modern genomics and epigenomics provide us with catalogues of gene regulatory elements and maps illustrating their activity in different tissues. Nevertheless, we are far from being able to explain emergence and maintenance of cellular states from such data, partly because we so far lacked characterization of individual molecular states and genome control mechanisms at their native resolution - the single cell. Recently, new approaches developed by the single cell genomics community, with several contributions from our group, allow massive acquisition of data on the transcriptional, epigenomic and chromosomal conformation states in large cohorts of single cells. In this research program, we aim to move forward rapidly to bridge a major gap between these experimental breakthroughs and models of genome regulation in complex tissues. We will develop algorithms and models for representing data the transcriptional profiles, DNA methylation landscapes and Hi-C maps of literally millions of cells. Our tools will be designed specifically to leverage on new single cell RNA-seq, single cell Hi-C, single cell capture-pBat and higher order 4C-seq that we will continue to develop experimentally. Furthermore, we shall enhance and optimize our interdisciplinary framework hand in hand with a working model aiming at unprecedentedly comprehensive single cell analysis of E8-E10 mouse embryos. This will provide us with hundreds of worked-out cases of tissue specific gene regulation. The techniques and insights from these studies will then be used to characterize cell type aberrations and epigenetic reprogramming in tumors. The open algorithms, techniques and methodology we shall develop can accelerate research in multiple groups that will utilize single cell genomics to study numerous questions on gene regulation in the coming years.
Max ERC Funding
2 437 500 €
Duration
Start date: 2017-12-01, End date: 2022-11-30
Project acronym SCIPER
Project Studying Cancer Individuality by Personal and Predictive Drug Screening and Differential OMICs
Researcher (PI) Berend SNIJDER
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Starting Grant (StG), LS2, ERC-2018-STG
Summary The cellular and molecular systems that determine drug responses in cancer are complex, highly individual, and incompletely understood. As a result, many cancer patients receive ineffective or even harmful therapies, which endangers lives, burdens healthcare systems, and prevents new therapies from reaching clinical approval.
To address this problem, we are developing a platform that measures hundreds of ex vivo drug responses from small patient biopsies by immunofluorescence, automated confocal microscopy, single-cell image analysis, and machine learning. We preserve cellular memory and maximize physiological relevance by not culturing or sorting cells prior to drug exposure. Sub-cellular, single-cell, and cell population-wide image analysis reveals on-target drug responses and disentangles multicellular ones. In a first interventional clinical trial, this phenotypic information alone led to strongly improved treatment of patients with aggressive hematologic malignancies.
Enabled by this high-throughput, predictive, and phenotypic information, I here propose to identify the molecular and cellular systems that govern treatment response individuality in cancer. (Aim 1) We will combine drug response profiling with RNA sequencing and proteomic measurements of malignant and healthy cells from the same biopsies. Critically, the patient-internal comparisons in both screening and OMICs allow neutralizing complex confounding factors. (Aim 2) New multiplexed immunofluorescence and convolutional neural network-based analyses will identify multiclass cell-types and -states, and quantify non-cell-autonomous responses. (Aim 3) Computational integration and causal inference will identify the molecular determinants and governing principles of drug response individuality in cancer, amenable to further validation. This proposal will thus improve our mechanistic understanding of cancer individuality and develop powerful new tools for OMICs-based precision medicine.
Summary
The cellular and molecular systems that determine drug responses in cancer are complex, highly individual, and incompletely understood. As a result, many cancer patients receive ineffective or even harmful therapies, which endangers lives, burdens healthcare systems, and prevents new therapies from reaching clinical approval.
To address this problem, we are developing a platform that measures hundreds of ex vivo drug responses from small patient biopsies by immunofluorescence, automated confocal microscopy, single-cell image analysis, and machine learning. We preserve cellular memory and maximize physiological relevance by not culturing or sorting cells prior to drug exposure. Sub-cellular, single-cell, and cell population-wide image analysis reveals on-target drug responses and disentangles multicellular ones. In a first interventional clinical trial, this phenotypic information alone led to strongly improved treatment of patients with aggressive hematologic malignancies.
Enabled by this high-throughput, predictive, and phenotypic information, I here propose to identify the molecular and cellular systems that govern treatment response individuality in cancer. (Aim 1) We will combine drug response profiling with RNA sequencing and proteomic measurements of malignant and healthy cells from the same biopsies. Critically, the patient-internal comparisons in both screening and OMICs allow neutralizing complex confounding factors. (Aim 2) New multiplexed immunofluorescence and convolutional neural network-based analyses will identify multiclass cell-types and -states, and quantify non-cell-autonomous responses. (Aim 3) Computational integration and causal inference will identify the molecular determinants and governing principles of drug response individuality in cancer, amenable to further validation. This proposal will thus improve our mechanistic understanding of cancer individuality and develop powerful new tools for OMICs-based precision medicine.
Max ERC Funding
1 500 000 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym SCrIPT
Project Stable Chromium Isotopes as a Productivity Tracer
Researcher (PI) Samuel JACCARD
Host Institution (HI) UNIVERSITAET BERN
Call Details Consolidator Grant (CoG), PE10, ERC-2018-COG
Summary The overall concept of this proposal is to investigate the main biogeochemical processes modulating spatial and temporal changes in marine export productivity, and assess their role in regulating atmospheric carbon dioxide (CO2) concentrations, both under present conditions and in the geological past. The exchange of CO2 between the atmosphere and ocean interior mediated by the oceanic ecosystem is a pivotal mechanism modulating the global carbon cycle, and thus, a substantial driver of the Earth’s climatic evolution.
The overarching objective of this research proposal is to develop a novel proxy to trace changes in the global strength of the marine biological carbon pump (BCP) based on stable Chromium (Cr) isotopes. Despite its significance for the global carbon cycle, the BCP is still poorly constrained. This project will explore a tracer that has recently been developed to investigate the rise of atmospheric oxygen in the early history of the Earth and develop it thoroughly through a comprehensive, multidisciplinary calibration program and apply it to the much more subtle redox variations associated with organic matter remineralization in the ocean. The proposed approach includes phytoplankton culture experiments, water-column investigations and sedimentary analysis and will aim at elucidating the mechanisms governing the reduction of Cr and its associated isotopic fractionation. The proxy will subsequently be used to reconstruct export production variability in the past and assess its role in modulating glacial/interglacial climate oscillations. These past changes tended to be much slower than the current, anthropogenic change. Nonetheless, they can help to appraise sensitivities and point toward potentially dominant mechanisms of change. The observations gathered within the framework of this research program will enable refining the evolution of the marine carbon cycle and the rapidly declining buffering capacity of the ocean.
Summary
The overall concept of this proposal is to investigate the main biogeochemical processes modulating spatial and temporal changes in marine export productivity, and assess their role in regulating atmospheric carbon dioxide (CO2) concentrations, both under present conditions and in the geological past. The exchange of CO2 between the atmosphere and ocean interior mediated by the oceanic ecosystem is a pivotal mechanism modulating the global carbon cycle, and thus, a substantial driver of the Earth’s climatic evolution.
The overarching objective of this research proposal is to develop a novel proxy to trace changes in the global strength of the marine biological carbon pump (BCP) based on stable Chromium (Cr) isotopes. Despite its significance for the global carbon cycle, the BCP is still poorly constrained. This project will explore a tracer that has recently been developed to investigate the rise of atmospheric oxygen in the early history of the Earth and develop it thoroughly through a comprehensive, multidisciplinary calibration program and apply it to the much more subtle redox variations associated with organic matter remineralization in the ocean. The proposed approach includes phytoplankton culture experiments, water-column investigations and sedimentary analysis and will aim at elucidating the mechanisms governing the reduction of Cr and its associated isotopic fractionation. The proxy will subsequently be used to reconstruct export production variability in the past and assess its role in modulating glacial/interglacial climate oscillations. These past changes tended to be much slower than the current, anthropogenic change. Nonetheless, they can help to appraise sensitivities and point toward potentially dominant mechanisms of change. The observations gathered within the framework of this research program will enable refining the evolution of the marine carbon cycle and the rapidly declining buffering capacity of the ocean.
Max ERC Funding
1 997 625 €
Duration
Start date: 2019-05-01, End date: 2024-04-30
Project acronym SEXGENTRANSEVOLUTION
Project Sex-biased genome and transcriptome evolution in mammals
Researcher (PI) Henrik Kaessmann
Host Institution (HI) UNIVERSITE DE LAUSANNE
Call Details Starting Grant (StG), LS2, ERC-2009-StG
Summary Mammalian males and females have many phenotypic differences. These differences, collectively referred to as sexual dimorphism, are the consequence of natural and sexual selection for phenotypic traits that affect the fitness of each sex and are encoded in the genome. Part of the underlying genomic differences between the sexes are found on sex specific (the Y) or sex biased chromosomes (the X), while many sexually dimorphic traits probably result from autosomal gene expression differences in sex specific or somatic tissues. However, the origin and evolution of sex-biased genes in mammals has not been studied in detail. I propose to generate the first detailed qualitative and quantitative transcriptome data using next generation sequencing technologies for a unique collection of germline and somatic tissues from representatives of all major mammalian lineages: placental mammals, marsupials, and the egg-laying monotremes. Together with detailed transcriptome data from birds (the evolutionary sister lineage), complementary experiments (e.g. methylome analyses), and available genomic resources from these species, these unprecedented data will allow an integrated analysis of the origin and functional evolution of mammalian sex chromosomes, the emergence of new sex biased genes, and the evolution of gene expression in germline versus somatic tissues in mammals at large. The proposed work will thus substantially increase our power to understand how mammalian genomes evolved the capacity to produce such pronounced sexually dimorphic traits. Beyond research pertaining to sex biased genome evolution, our data will represent a unique resource for future investigations of mammalian gene functions and serve as a basis for exploring the evolution of other mammal specific phenotypes.
Summary
Mammalian males and females have many phenotypic differences. These differences, collectively referred to as sexual dimorphism, are the consequence of natural and sexual selection for phenotypic traits that affect the fitness of each sex and are encoded in the genome. Part of the underlying genomic differences between the sexes are found on sex specific (the Y) or sex biased chromosomes (the X), while many sexually dimorphic traits probably result from autosomal gene expression differences in sex specific or somatic tissues. However, the origin and evolution of sex-biased genes in mammals has not been studied in detail. I propose to generate the first detailed qualitative and quantitative transcriptome data using next generation sequencing technologies for a unique collection of germline and somatic tissues from representatives of all major mammalian lineages: placental mammals, marsupials, and the egg-laying monotremes. Together with detailed transcriptome data from birds (the evolutionary sister lineage), complementary experiments (e.g. methylome analyses), and available genomic resources from these species, these unprecedented data will allow an integrated analysis of the origin and functional evolution of mammalian sex chromosomes, the emergence of new sex biased genes, and the evolution of gene expression in germline versus somatic tissues in mammals at large. The proposed work will thus substantially increase our power to understand how mammalian genomes evolved the capacity to produce such pronounced sexually dimorphic traits. Beyond research pertaining to sex biased genome evolution, our data will represent a unique resource for future investigations of mammalian gene functions and serve as a basis for exploring the evolution of other mammal specific phenotypes.
Max ERC Funding
1 901 522 €
Duration
Start date: 2010-02-01, End date: 2015-01-31
Project acronym Signaling 3D
Project Three Dimensional Single Cell Analysis of the Cancer Stem Cell Inducing Epithelial-Mesenchymal Transition Signaling Networks in Breast Cancer by Mass Cytometry
Researcher (PI) Bernd Bodenmiller
Host Institution (HI) UNIVERSITAT ZURICH
Call Details Starting Grant (StG), LS2, ERC-2013-StG
Summary Tumor metastases, relapse, and resistance to therapy are the main causes of death in cancer patients. Cancer stem cells (CSCs) drive cancer growth, are likely responsible for cancer reoccurrence, and provide the potential to colonize a metastatic site. A cell plasticity process called epithelial-mesenchymal transition (EMT) generates CSCs from epithelial cancer cells and simultaneously equips these cells with motility and invasiveness, features prerequisite for metastasis. Consequently, therapeutic strategies that target EMT and CSC signaling networks are highly attractive. However, the properties of these signaling networks and their dependency on the tumor microenvironment and cancer genotypes are poorly understood. Here we propose to generate quantitative, time-resolved biomarker signatures and network models of EMT stages and CSCs on the single-cell level and to define their dependency on breast cancer tumor microenvironments and genotypes. Using mass cytometry, a technology able to quantify up to 100 proteins and phosphorylation sites simultaneously in a single-cell, the signaling network structure of EMT and CSC state will be gauged by modulation of cancer-related and signaling genes. These data will be used to infer mathematical signaling network descriptions of EMT and the CSC states, and to determine their master regulators and regulatory sub-networks. By extending mass cytometry to spatially resolved measurements, the EMT/CSC network states and their microenvironment will be analyzed in three dimensions in patient samples, and data will be correlated with associated genomic and clinical information. Based on these in vivo data, follow-up experiments in mouse models will be performed to validate the identified network states and cell-to-cell interaction as therapeutic targets. Finally, the in vivo dataset and its correlation with genomic and clinical information will be used to identify biomarkers for personalized medicine approaches.
Summary
Tumor metastases, relapse, and resistance to therapy are the main causes of death in cancer patients. Cancer stem cells (CSCs) drive cancer growth, are likely responsible for cancer reoccurrence, and provide the potential to colonize a metastatic site. A cell plasticity process called epithelial-mesenchymal transition (EMT) generates CSCs from epithelial cancer cells and simultaneously equips these cells with motility and invasiveness, features prerequisite for metastasis. Consequently, therapeutic strategies that target EMT and CSC signaling networks are highly attractive. However, the properties of these signaling networks and their dependency on the tumor microenvironment and cancer genotypes are poorly understood. Here we propose to generate quantitative, time-resolved biomarker signatures and network models of EMT stages and CSCs on the single-cell level and to define their dependency on breast cancer tumor microenvironments and genotypes. Using mass cytometry, a technology able to quantify up to 100 proteins and phosphorylation sites simultaneously in a single-cell, the signaling network structure of EMT and CSC state will be gauged by modulation of cancer-related and signaling genes. These data will be used to infer mathematical signaling network descriptions of EMT and the CSC states, and to determine their master regulators and regulatory sub-networks. By extending mass cytometry to spatially resolved measurements, the EMT/CSC network states and their microenvironment will be analyzed in three dimensions in patient samples, and data will be correlated with associated genomic and clinical information. Based on these in vivo data, follow-up experiments in mouse models will be performed to validate the identified network states and cell-to-cell interaction as therapeutic targets. Finally, the in vivo dataset and its correlation with genomic and clinical information will be used to identify biomarkers for personalized medicine approaches.
Max ERC Funding
1 500 000 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym SoilLife
Project The Hidden Frontier: Quantitative Exploration of Physical and Ecological Origins of Microbial Diversity in Soil
Researcher (PI) Dani Or
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), PE10, ERC-2012-ADG_20120216
Summary By some accounts exploring the origins of soil microbial diversity represents a scientific frontier similar to that of space exploration in its scope, described by Curtis and Sloan (2004) as ”an immense and unexplored frontier in science of astronomical dimensions and of astonishing complexity”. The complexity is attributed to soil ecological heterogeneity reflecting interplay of spatio-temporal, physical, and nutritional variables delineating spheres of influence that define microbial habitats and function. Key to microbial life in soil is a flickering aqueous network that defines nutrient diffusional pathways and shapes microbial dispersion and interactions. We propose to develop an individual-based and spatially resolved modeling platform that explicitly considers soil pore structure and aqueous phase configuration and associated biophysical processes forming a virtual soil microcosm. The assembly of these complex ingredients into a computational platform will enable systematic hypotheses testing concerning central questions in microbial ecology that are neither addressed by present ecological theories nor emerge from standard continuum models. Specifically, the project will provide quantitative insights into effects of hydration extremes on survival strategies, the roles of space and diffusional heterogeneity, aspects of dispersion and trophic interactions, self-organization of consortia, and emergence of temporal niches in soil. The research will transform quantitative understanding of soil biophysical processes, a gap that presently limits coherent interpretation of the rapidly growing molecular-based estimates of soil biodiversity, and is essential for guiding future data collection. The research lies at the interface between environmental microbiology and soil physics cutting across disciplinary boundaries and addressing broad issues impacting soil and water quality, the functioning of global bio-geochemical cycles, and the fate of anthropogenic pollutants.
Summary
By some accounts exploring the origins of soil microbial diversity represents a scientific frontier similar to that of space exploration in its scope, described by Curtis and Sloan (2004) as ”an immense and unexplored frontier in science of astronomical dimensions and of astonishing complexity”. The complexity is attributed to soil ecological heterogeneity reflecting interplay of spatio-temporal, physical, and nutritional variables delineating spheres of influence that define microbial habitats and function. Key to microbial life in soil is a flickering aqueous network that defines nutrient diffusional pathways and shapes microbial dispersion and interactions. We propose to develop an individual-based and spatially resolved modeling platform that explicitly considers soil pore structure and aqueous phase configuration and associated biophysical processes forming a virtual soil microcosm. The assembly of these complex ingredients into a computational platform will enable systematic hypotheses testing concerning central questions in microbial ecology that are neither addressed by present ecological theories nor emerge from standard continuum models. Specifically, the project will provide quantitative insights into effects of hydration extremes on survival strategies, the roles of space and diffusional heterogeneity, aspects of dispersion and trophic interactions, self-organization of consortia, and emergence of temporal niches in soil. The research will transform quantitative understanding of soil biophysical processes, a gap that presently limits coherent interpretation of the rapidly growing molecular-based estimates of soil biodiversity, and is essential for guiding future data collection. The research lies at the interface between environmental microbiology and soil physics cutting across disciplinary boundaries and addressing broad issues impacting soil and water quality, the functioning of global bio-geochemical cycles, and the fate of anthropogenic pollutants.
Max ERC Funding
2 196 632 €
Duration
Start date: 2013-02-01, End date: 2018-01-31
Project acronym SPACE TIE
Project Unifying the three pillars of Geodesy using space ties
Researcher (PI) Adrian JÄGGI
Host Institution (HI) UNIVERSITAET BERN
Call Details Consolidator Grant (CoG), PE10, ERC-2018-COG
Summary "Terrestrial Reference Frames (TRFs) are the basis to which all positions on the Earth’s surface and all satellite orbits in the near Earth space have to refer to. The changes in the Earth's shape, rotation, and gravity field, the so-called ""three pillars"" of geodesy, provide the conceptual and observational basis for the TRFs. For today’s TRF realizations, four space geodetic techniques are combined and linked by co-location sites on the Earth’s surface (“Earth’s shape”) and by common Earth orientation parameters (“Earth rotation”). The third pillar (“Earth’s gravity field”) is today only contributing to the TRF determination via its associated center-of-mass. In SPACE TIE we will pave the way to unify the “three pillars” of Geodesy in future TRF realizations. We propose to use two satellite geodetic techniques, namely Global Navigation Satellite Systems (GNSS) and Satellite Laser Ranging (SLR), to connect them by co-location sites in space. These so-called space ties shall be realized on satellites of the currently existing space infrastructure, as well as on satellites due for launch in the near future. This includes the Medium Earth Orbits (MEO) of the GNSS satellites and, in particular, all satellites in Low Earth Orbits (LEO) with GNSS and SLR co-located on-board. To maximize the sensitivity to the Earth’s gravity field, the ultra-precise inter-satellite ranging between LEO satellites of dedicated gravity missions shall be added as a third satellite geodetic technique. One and the same state-of-the-art space geodetic software package will be used to ensure that standards, background models, and processing strategies are consistently applied across all co-location satellites and measurement techniques. The outcome of SPACE TIE will allow it to assess the geometric and gravimetric impact of mass transport in the atmosphere, oceans, and ice caps in a most consistent way to globally quantify the mass exchange between the different components of the system Earth."
Summary
"Terrestrial Reference Frames (TRFs) are the basis to which all positions on the Earth’s surface and all satellite orbits in the near Earth space have to refer to. The changes in the Earth's shape, rotation, and gravity field, the so-called ""three pillars"" of geodesy, provide the conceptual and observational basis for the TRFs. For today’s TRF realizations, four space geodetic techniques are combined and linked by co-location sites on the Earth’s surface (“Earth’s shape”) and by common Earth orientation parameters (“Earth rotation”). The third pillar (“Earth’s gravity field”) is today only contributing to the TRF determination via its associated center-of-mass. In SPACE TIE we will pave the way to unify the “three pillars” of Geodesy in future TRF realizations. We propose to use two satellite geodetic techniques, namely Global Navigation Satellite Systems (GNSS) and Satellite Laser Ranging (SLR), to connect them by co-location sites in space. These so-called space ties shall be realized on satellites of the currently existing space infrastructure, as well as on satellites due for launch in the near future. This includes the Medium Earth Orbits (MEO) of the GNSS satellites and, in particular, all satellites in Low Earth Orbits (LEO) with GNSS and SLR co-located on-board. To maximize the sensitivity to the Earth’s gravity field, the ultra-precise inter-satellite ranging between LEO satellites of dedicated gravity missions shall be added as a third satellite geodetic technique. One and the same state-of-the-art space geodetic software package will be used to ensure that standards, background models, and processing strategies are consistently applied across all co-location satellites and measurement techniques. The outcome of SPACE TIE will allow it to assess the geometric and gravimetric impact of mass transport in the atmosphere, oceans, and ice caps in a most consistent way to globally quantify the mass exchange between the different components of the system Earth."
Max ERC Funding
1 999 563 €
Duration
Start date: 2019-05-01, End date: 2024-04-30
Project acronym SPADE
Project Speleothems paleoclimate: accounting for isotopic disequilibrium
Researcher (PI) Hagit Pnina AFFEK
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Consolidator Grant (CoG), PE10, ERC-2016-COG
Summary Understanding and quantifying the impacts of climate change at the regional and hemispheric scales are particularly difficult with respect to changes in rainfall and temperature patterns that lead to extended droughts and flooding events. Isotopic records in speleothems are increasingly used to determine climate variability on land and for data-model comparisons. However, transferring speleothem records into quantitative climate parameters suffers from a major limitation: speleothem formation processes result in geochemical disequilibrium and there is currently no way to correct for it in paleoclimate data. SPADE will shift the treatment of paleoclimate archives from regarding them as recorders of slow geological processes to consideration of geological material as recording much faster chemical reactions. As such, they cannot be assumed to form at equilibrium. SPADE will create a new framework, based on one classic and two novel isotopic tracers in carbonates (δ18O-Δ17O-Δ47) to quantify disequilibrium in cave records and overcome this underlying limitation. SPADE’s unique approach is based first on laboratory experiments that isolate chemical processes of speleothem formation, to test their respective effects on isotopic disequilibrium. Then speleothem analog experiments and modern cave material are combined to create speleothem specific calibrations for these isotopic proxies. These SPADE results will then be applied to classic paleoclimate records of dryland hydrology, such as Soreq Cave (Israel) and Devils Hole (Nevada). SPADE will address long standing climatic hypotheses regarding the interplay between temperature, amount of rainfall, surface evaporation, moisture sources, and regional climate connections in these drought vulnerable regions, and will make these records much more useful. A detailed understanding of disequilibrium will enable the use of these innovative geochemical tools in speleothems and more broadly, in other paleoclimate carbonate archives.
Summary
Understanding and quantifying the impacts of climate change at the regional and hemispheric scales are particularly difficult with respect to changes in rainfall and temperature patterns that lead to extended droughts and flooding events. Isotopic records in speleothems are increasingly used to determine climate variability on land and for data-model comparisons. However, transferring speleothem records into quantitative climate parameters suffers from a major limitation: speleothem formation processes result in geochemical disequilibrium and there is currently no way to correct for it in paleoclimate data. SPADE will shift the treatment of paleoclimate archives from regarding them as recorders of slow geological processes to consideration of geological material as recording much faster chemical reactions. As such, they cannot be assumed to form at equilibrium. SPADE will create a new framework, based on one classic and two novel isotopic tracers in carbonates (δ18O-Δ17O-Δ47) to quantify disequilibrium in cave records and overcome this underlying limitation. SPADE’s unique approach is based first on laboratory experiments that isolate chemical processes of speleothem formation, to test their respective effects on isotopic disequilibrium. Then speleothem analog experiments and modern cave material are combined to create speleothem specific calibrations for these isotopic proxies. These SPADE results will then be applied to classic paleoclimate records of dryland hydrology, such as Soreq Cave (Israel) and Devils Hole (Nevada). SPADE will address long standing climatic hypotheses regarding the interplay between temperature, amount of rainfall, surface evaporation, moisture sources, and regional climate connections in these drought vulnerable regions, and will make these records much more useful. A detailed understanding of disequilibrium will enable the use of these innovative geochemical tools in speleothems and more broadly, in other paleoclimate carbonate archives.
Max ERC Funding
2 000 000 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym SYMPAC
Project Synthetic metabolic pathways for carbon fixation
Researcher (PI) Ron Milo
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2010-StG_20091118
Summary Carbon fixation is the main pathway for storing energy and accumulating biomass in the living world. It is also the principal reason for humanity s utilization of land and water. Under human cultivation, carbon fixation significantly limits growth. Hence increasing carbon fixation rate is of major importance towards agricultural and energetic sustainability.
Are there limits on the rate of such central metabolic pathways? Attempts to improve the rate of Rubisco, the key enzyme in the Calvin-Benson cycle, have achieved very limited success. In this proposal we try to overcome this bottleneck by systematically exploring the space of carbon fixation pathways that can be assembled from all ~4000 metabolic enzymes known in nature. We computationally compare all possible pathways based on kinetics, energetics and topology. Our initial analysis suggests a new family of synthetic carbon fixation pathways utilizing the most effective carboxylating enzyme, PEPC. We propose to experimentally test these cycles in the most genetically tractable context by constructing an E. coli strain that will depend on carbon fixation as its sole carbon input. Energy will be supplied by compounds that cannot be used as carbon source. Initially, we will devise an autotrophic E. coli strain to use the Calvin-Benson Cycle; in the next stage, we will implement the most promising synthetic cycles. Systematic in vivo comparison will guide the future implementation in natural photosynthetic organisms.
At the basic science level, this proposal revisits and challenges our understanding of central carbon metabolism and growth. Concomitantly, it is an evolutionary experiment on integration of a biological novelty. It will serve as a model for significantly adapting a central metabolic pathway.
Summary
Carbon fixation is the main pathway for storing energy and accumulating biomass in the living world. It is also the principal reason for humanity s utilization of land and water. Under human cultivation, carbon fixation significantly limits growth. Hence increasing carbon fixation rate is of major importance towards agricultural and energetic sustainability.
Are there limits on the rate of such central metabolic pathways? Attempts to improve the rate of Rubisco, the key enzyme in the Calvin-Benson cycle, have achieved very limited success. In this proposal we try to overcome this bottleneck by systematically exploring the space of carbon fixation pathways that can be assembled from all ~4000 metabolic enzymes known in nature. We computationally compare all possible pathways based on kinetics, energetics and topology. Our initial analysis suggests a new family of synthetic carbon fixation pathways utilizing the most effective carboxylating enzyme, PEPC. We propose to experimentally test these cycles in the most genetically tractable context by constructing an E. coli strain that will depend on carbon fixation as its sole carbon input. Energy will be supplied by compounds that cannot be used as carbon source. Initially, we will devise an autotrophic E. coli strain to use the Calvin-Benson Cycle; in the next stage, we will implement the most promising synthetic cycles. Systematic in vivo comparison will guide the future implementation in natural photosynthetic organisms.
At the basic science level, this proposal revisits and challenges our understanding of central carbon metabolism and growth. Concomitantly, it is an evolutionary experiment on integration of a biological novelty. It will serve as a model for significantly adapting a central metabolic pathway.
Max ERC Funding
1 498 792 €
Duration
Start date: 2011-01-01, End date: 2015-12-31
Project acronym THERA
Project Timing of Holocene volcanic eruptions and their radiative aerosol forcing
Researcher (PI) Michael SIGL
Host Institution (HI) UNIVERSITAET BERN
Call Details Consolidator Grant (CoG), PE10, ERC-2018-COG
Summary Volcanic eruptions play a dominant role in driving climate, in ways beyond the established short-term influence on surface air temperatures. In order to mitigate and adapt to the climate effects of future large volcanic eruptions we need to better quantify the risk of these eruptions including 1) the probability of their occurrence and 2) their expected climatic impact. The observational record of the timing of volcanic eruptions, their locations, magnitudes of sulphate aerosol injection is incomplete which limits our understanding of the sensitivity of the Earth system to volcanism and the vulnerability of social and economic systems to the climate impact of past and future eruptions.
The primary goal of this proposal is to extract data on the timing, magnitudes and source locations of all major volcanic eruptions occurring during the Holocene (i.e., the past 12,000 years) to answer the questions: What is the likelihood of a stratospheric sulfur injection as large as that from the colossal eruption of Tambora in 1815 to occur somewhere on the globe within the next 100 years? What is the role of effusive eruptions on past, present and future climate?
This will be achieved by employing novel, precisely dated, high-time resolution aerosol measurements from bipolar ice-core arrays. New tools will be used to constrain source parameters of the eruptions (location, plume injection height) that control their effects on climate. THERA will constrain recurrence rates for one of the largest global-scale natural hazards, while also assessing linkages between volcanic perturbations and key components of the climate systems (e.g., atmospheric circulation, droughts, ice-sheets and sea-level) through interdisciplinary case studies. As a final goal, THERA will generate global-scale, space-and-time resolved stratospheric aerosol properties for climate models to simulate the volcanic influence on Holocene climate evolution.
Summary
Volcanic eruptions play a dominant role in driving climate, in ways beyond the established short-term influence on surface air temperatures. In order to mitigate and adapt to the climate effects of future large volcanic eruptions we need to better quantify the risk of these eruptions including 1) the probability of their occurrence and 2) their expected climatic impact. The observational record of the timing of volcanic eruptions, their locations, magnitudes of sulphate aerosol injection is incomplete which limits our understanding of the sensitivity of the Earth system to volcanism and the vulnerability of social and economic systems to the climate impact of past and future eruptions.
The primary goal of this proposal is to extract data on the timing, magnitudes and source locations of all major volcanic eruptions occurring during the Holocene (i.e., the past 12,000 years) to answer the questions: What is the likelihood of a stratospheric sulfur injection as large as that from the colossal eruption of Tambora in 1815 to occur somewhere on the globe within the next 100 years? What is the role of effusive eruptions on past, present and future climate?
This will be achieved by employing novel, precisely dated, high-time resolution aerosol measurements from bipolar ice-core arrays. New tools will be used to constrain source parameters of the eruptions (location, plume injection height) that control their effects on climate. THERA will constrain recurrence rates for one of the largest global-scale natural hazards, while also assessing linkages between volcanic perturbations and key components of the climate systems (e.g., atmospheric circulation, droughts, ice-sheets and sea-level) through interdisciplinary case studies. As a final goal, THERA will generate global-scale, space-and-time resolved stratospheric aerosol properties for climate models to simulate the volcanic influence on Holocene climate evolution.
Max ERC Funding
1 978 923 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym TRANSCRIPTION_REG
Project A combined experimental and computational approach for quantitative and mechanistic understanding of transcriptional regulation
Researcher (PI) Eran Segal
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2007-StG
Summary The complex functions of a living cell are carried out through the coordinated activity of many genes. Since transcription is a key step in establishing such coordinated activity, much effort was devoted to its study, and tremendous progress was made in identifying many of the transcription factors and regulatory DNA elements involved in the regulation of specific systems. However, very few attempts were made at going beyond these phenomenological and qualitative descriptions. Consequently, we are far from a quantitative and predictive understanding of transcriptional regulation. Through this program, I aim to develop a mechanistic understanding of transcriptional regulation, and for the first time model the entire process. We wish to go much beyond identifying and qualitatively describing the involved components, and arrive at a quantitative understanding of how transcriptional programs are encoded in the DNA sequences. To this end, my team and I will first work to mechanistically understand various building blocks of the transcriptional system, including: mechanisms of activation and repression; binding cooperativity; binding competition; transcription factors and chromatin interplay; architectural features of promoters that are important for its function; and the transcription functions “computed” by promoters. Since existing data are clearly insufficient for addressing such questions, I have opened an experimental lab and began to assemble a multidisciplinary team of scientists whose expertise span the experimental biology, computer science, physics, statistics, and mathematics disciplines, that will work synergistically to generate the appropriate data, analyze it, and use it to construct and experimentally validate models for the above transcriptional building blocks. We will then integrate all the insights gained into unified and quantitative models that should significantly enhance our understanding of the mechanistic workings of transcriptional regulation.
Summary
The complex functions of a living cell are carried out through the coordinated activity of many genes. Since transcription is a key step in establishing such coordinated activity, much effort was devoted to its study, and tremendous progress was made in identifying many of the transcription factors and regulatory DNA elements involved in the regulation of specific systems. However, very few attempts were made at going beyond these phenomenological and qualitative descriptions. Consequently, we are far from a quantitative and predictive understanding of transcriptional regulation. Through this program, I aim to develop a mechanistic understanding of transcriptional regulation, and for the first time model the entire process. We wish to go much beyond identifying and qualitatively describing the involved components, and arrive at a quantitative understanding of how transcriptional programs are encoded in the DNA sequences. To this end, my team and I will first work to mechanistically understand various building blocks of the transcriptional system, including: mechanisms of activation and repression; binding cooperativity; binding competition; transcription factors and chromatin interplay; architectural features of promoters that are important for its function; and the transcription functions “computed” by promoters. Since existing data are clearly insufficient for addressing such questions, I have opened an experimental lab and began to assemble a multidisciplinary team of scientists whose expertise span the experimental biology, computer science, physics, statistics, and mathematics disciplines, that will work synergistically to generate the appropriate data, analyze it, and use it to construct and experimentally validate models for the above transcriptional building blocks. We will then integrate all the insights gained into unified and quantitative models that should significantly enhance our understanding of the mechanistic workings of transcriptional regulation.
Max ERC Funding
1 005 600 €
Duration
Start date: 2008-07-01, End date: 2013-06-30
Project acronym TransgenerationalRNA
Project RNA-Mediated Inheritance of Acquired Traits
Researcher (PI) Oded Rechavi
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS2, ERC-2013-StG
Summary Inheritance of acquired traits is a topic of long-standing interest and controversy. While some of the classic Lamarckian ideas have been dismissed, more recent observations suggest that certain characteristics acquired by an animal during its lifetime might be transmitted to the next generations. Recently I described, for the first time in animals, a biological context in which acquired traits are inherited via small RNA molecules, which ignore the boundary between the soma and the germ line (“The Weizmann Barrier”). Specifically, I showed that the nematode C.elegans inherit an acquired trait, antiviral resistance, through transgenerational transmission of antiviral small RNAs (viRNAs), which mediate RNA interference (RNAi) (Cell, 2011). viRNAs, which protect the worm from viral propagation, pass down to many ensuing generations in a non-Mendelian manner, in the absence of their DNA template, and thus defend RNAi-deficient progeny from viral propagation. Here I suggest defining the rules that govern RNA-mediated transgenerational inheritance of acquired traits and exploring its contribution for the genetics of complex traits. My first efforts will be directed towards elucidating the mechanism behind transgenerational transmission of small RNAs; I established a well-defined system for monitoring transgenerational silencing that should allow unveiling of its genetic and biochemical basis. Second, I will examine whether responses to relevant environmental stresses carry on to the next generations so that the progeny is better prepared to cope with similar conditions. Lastly, I will explore whether sensing of environmental cues by the nervous system drives small RNA biogenesis, which transfer transgenerationally and mediate inheritance of neuronally-encoded traits. While the idea that RNA encodes for “Inherited Memory” sounds heretic at first, my preliminary efforts suggest that inherited small RNAs may indeed transmit information about ancestral acquired experiences.
Summary
Inheritance of acquired traits is a topic of long-standing interest and controversy. While some of the classic Lamarckian ideas have been dismissed, more recent observations suggest that certain characteristics acquired by an animal during its lifetime might be transmitted to the next generations. Recently I described, for the first time in animals, a biological context in which acquired traits are inherited via small RNA molecules, which ignore the boundary between the soma and the germ line (“The Weizmann Barrier”). Specifically, I showed that the nematode C.elegans inherit an acquired trait, antiviral resistance, through transgenerational transmission of antiviral small RNAs (viRNAs), which mediate RNA interference (RNAi) (Cell, 2011). viRNAs, which protect the worm from viral propagation, pass down to many ensuing generations in a non-Mendelian manner, in the absence of their DNA template, and thus defend RNAi-deficient progeny from viral propagation. Here I suggest defining the rules that govern RNA-mediated transgenerational inheritance of acquired traits and exploring its contribution for the genetics of complex traits. My first efforts will be directed towards elucidating the mechanism behind transgenerational transmission of small RNAs; I established a well-defined system for monitoring transgenerational silencing that should allow unveiling of its genetic and biochemical basis. Second, I will examine whether responses to relevant environmental stresses carry on to the next generations so that the progeny is better prepared to cope with similar conditions. Lastly, I will explore whether sensing of environmental cues by the nervous system drives small RNA biogenesis, which transfer transgenerationally and mediate inheritance of neuronally-encoded traits. While the idea that RNA encodes for “Inherited Memory” sounds heretic at first, my preliminary efforts suggest that inherited small RNAs may indeed transmit information about ancestral acquired experiences.
Max ERC Funding
1 500 000 €
Duration
Start date: 2014-02-01, End date: 2019-01-31