Project acronym ICARUS
Project Towards Innovative cost-effective astronomical instrumentation
Researcher (PI) Emmanuel Hugot
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), PE9, ERC-2015-STG
Summary Enabling disruptive technologies has always been crucial to trigger revolutionary science discoveries. The daring challenges in astronomy and astrophysics are extremely demanding in terms of high angular resolution and high contrast imaging, and require extreme stability and image quality. Instruments based on current classical designs tend to get bigger and more complex, and are faced to ever increasing difficulties to meet science requirements.
This proposal has the clear objective to propose breakthrough compact optical architectures for the next generation of giant observatories. The project focus on the niche of active components and is structured in two main research pillars to (I) enable the use of additive manufacturing (3D-printing) to produce affordable deformable mirrors for VIS or NIR observations, (II) pave the road for a common use of curved and deformable detectors. Extensive finite element analysis will allow to cover the parameter space and broad prototyping will demonstrate and characterize the performance of such devices.
Both pillars are extremely challenging, the fields of detectors and optical fabrication being driven by the market. We will then orientate the activities towards a mass production method.
To maximize the impact of this high gain R&D, the pillars are surrounded by two transverse activities: (i) design and optimization of a new zoo of optical systems using active mirrors and flexible detectors, and (ii) build a solid plan of technology transfer to end-user industrial companies, through a patenting and licensing strategy, to maximize the financial return and then perpetuate the activities.
The pathway proposed here is mandatory to develop affordable components in the near future, and will enable compact and high performance instrumentation. These high potential activities will dramatically reduce the complexity of instruments in the era of giant observatories, simplify the operability of systems and offer increased performance.
Summary
Enabling disruptive technologies has always been crucial to trigger revolutionary science discoveries. The daring challenges in astronomy and astrophysics are extremely demanding in terms of high angular resolution and high contrast imaging, and require extreme stability and image quality. Instruments based on current classical designs tend to get bigger and more complex, and are faced to ever increasing difficulties to meet science requirements.
This proposal has the clear objective to propose breakthrough compact optical architectures for the next generation of giant observatories. The project focus on the niche of active components and is structured in two main research pillars to (I) enable the use of additive manufacturing (3D-printing) to produce affordable deformable mirrors for VIS or NIR observations, (II) pave the road for a common use of curved and deformable detectors. Extensive finite element analysis will allow to cover the parameter space and broad prototyping will demonstrate and characterize the performance of such devices.
Both pillars are extremely challenging, the fields of detectors and optical fabrication being driven by the market. We will then orientate the activities towards a mass production method.
To maximize the impact of this high gain R&D, the pillars are surrounded by two transverse activities: (i) design and optimization of a new zoo of optical systems using active mirrors and flexible detectors, and (ii) build a solid plan of technology transfer to end-user industrial companies, through a patenting and licensing strategy, to maximize the financial return and then perpetuate the activities.
The pathway proposed here is mandatory to develop affordable components in the near future, and will enable compact and high performance instrumentation. These high potential activities will dramatically reduce the complexity of instruments in the era of giant observatories, simplify the operability of systems and offer increased performance.
Max ERC Funding
1 747 667 €
Duration
Start date: 2016-08-01, End date: 2021-07-31
Project acronym ICEBERG
Project Discovery of Type 2 Diabetes Targets
Researcher (PI) Juleen Rae Zierath
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Advanced Grant (AdG), LS4, ERC-2008-AdG
Summary This proposal is focused on the identification and biological validation of the metabolic pathways and key regulatory genes that control insulin sensitivity in Type 2 diabetes mellitus (T2DM). We are focusing on skeletal muscle because it is quantitatively the most important tissue involved in maintaining glucose homeostasis under insulin-stimulated conditions and it is a major site of insulin resistance in T2DM. Our central hypothesis is that alterations in insulin signal transduction to glucose transport contribute to the profound impairment in whole body glucose homeostasis and T2DM pathogenesis. Identification of the defects in T2DM can lead to the development of new therapeutic strategies to prevent and cure this disease. The proposal is divided into two main objectives: We will apply: 1) target identification platforms including microarray, proteomics and bioinformatics to identify dysregulated genes in normal glucose tolerant versus T2DM subjects or genetically modified model systems and 2) functional genomics to assign a physiological role of the identified targets in Aim 1 using cellular and whole-body approaches. We will focus on the mitogen-activated protein kinase family, the energy-sensing enzyme AMP-activated protein kinase, and the lipid intermediate metabolizing enzyme diacylglycerol kinase delta. Our previous work indicates that these candidates play a role in the regulation of glucose metabolism, triglyceride storage, and energy homeostasis.
Summary
This proposal is focused on the identification and biological validation of the metabolic pathways and key regulatory genes that control insulin sensitivity in Type 2 diabetes mellitus (T2DM). We are focusing on skeletal muscle because it is quantitatively the most important tissue involved in maintaining glucose homeostasis under insulin-stimulated conditions and it is a major site of insulin resistance in T2DM. Our central hypothesis is that alterations in insulin signal transduction to glucose transport contribute to the profound impairment in whole body glucose homeostasis and T2DM pathogenesis. Identification of the defects in T2DM can lead to the development of new therapeutic strategies to prevent and cure this disease. The proposal is divided into two main objectives: We will apply: 1) target identification platforms including microarray, proteomics and bioinformatics to identify dysregulated genes in normal glucose tolerant versus T2DM subjects or genetically modified model systems and 2) functional genomics to assign a physiological role of the identified targets in Aim 1 using cellular and whole-body approaches. We will focus on the mitogen-activated protein kinase family, the energy-sensing enzyme AMP-activated protein kinase, and the lipid intermediate metabolizing enzyme diacylglycerol kinase delta. Our previous work indicates that these candidates play a role in the regulation of glucose metabolism, triglyceride storage, and energy homeostasis.
Max ERC Funding
2 500 000 €
Duration
Start date: 2009-01-01, End date: 2013-12-31
Project acronym ICYBOB
Project Initial Conditions of YMCs, Birth of OB associations and long term evolution of stellar clusters
Researcher (PI) Clare Louise DOBBS
Host Institution (HI) THE UNIVERSITY OF EXETER
Call Details Consolidator Grant (CoG), PE9, ERC-2018-COG
Summary The goal of this proposal is to establish a new era of stellar cluster evolution research by performing numerical simulations on different scales, and of different stages of a cluster’s life, from the formation of YMCs, the formation and evolution of OB associations, to the evolution of clusters and associations in galaxies. The PI is one of the pioneers of galactic simulations of GMC and star formation was one of the first numericists to perform galactic scale simulations of molecular cloud formation and evolution, and has produced some of the most realistic and sophisticated isolated simulations of galaxies in this field to date. The next challenge is to follow cluster evolution, something which has not yet been attempted numerically. And, with the GaiaAIA instrument set to transform stellar astronomy in our Galaxy, our work will provide a fundamental theoretical counterpart. Key questions we will address include i) how does gas disperse from new clusters and what happens to that gas, ii) how do YMCs form, iii) how do new clustersGiant Molecular Clouds (GMCs) evolve into OB associations, and ivii) how long can clusters survive for as they orbit a galaxy and what causes their destruction.
Summary
The goal of this proposal is to establish a new era of stellar cluster evolution research by performing numerical simulations on different scales, and of different stages of a cluster’s life, from the formation of YMCs, the formation and evolution of OB associations, to the evolution of clusters and associations in galaxies. The PI is one of the pioneers of galactic simulations of GMC and star formation was one of the first numericists to perform galactic scale simulations of molecular cloud formation and evolution, and has produced some of the most realistic and sophisticated isolated simulations of galaxies in this field to date. The next challenge is to follow cluster evolution, something which has not yet been attempted numerically. And, with the GaiaAIA instrument set to transform stellar astronomy in our Galaxy, our work will provide a fundamental theoretical counterpart. Key questions we will address include i) how does gas disperse from new clusters and what happens to that gas, ii) how do YMCs form, iii) how do new clustersGiant Molecular Clouds (GMCs) evolve into OB associations, and ivii) how long can clusters survive for as they orbit a galaxy and what causes their destruction.
Max ERC Funding
1 980 385 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym icyMARS
Project Cold and wet early Mars: Proposing and testing a new theory to understand the early Martian environments
Researcher (PI) Alberto Gonzalez Fairen
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Starting Grant (StG), PE9, ERC-2012-StG_20111012
Summary Geologic evidence indicative of flowing and ponding liquid water on the surface of ancient Mars appears abundantly across most of the Martian landscape, indicating that liquid water was present in variable amounts and for long periods of time on and/or near the surface at different moments of Mars’ early history, the Noachian era. Early Mars appears to have been “wet”. However, the presence of liquid water on the surface of early Mars is difficult to reconcile with the reduced solar luminosity at 3.8 Ga. and before, which would have imposed mean temperatures below freezing all over the planet. Atmospheric greenhouse gases and carbon dioxide ice clouds in the upper troposphere are suggested to provide over freezing temperatures, explaining some of this discrepancy, but these solutions have been probed to face numerous problems. So, it is difficult to explain the early Martian hydrology invoking global “warm” conditions. Here I propose to conduct interdisciplinary investigations in order to define and test a new hypothesis to understand the early environmental traits on Mars: that the young Martian surface was characterized by global mean freezing conditions, as predicted by climate models, and at the same time a vigorous hydrogeological cycle was active during hundreds of millions of years, as confirmed by geomorphological and mineralogical analyses. The aim of this investigation is to comprehensively analyze the triggers, traits and consequences of a cold aqueous environment dominating the Noachian, studying the geomorphological, mineralogical and geochemical evidences that such a hydrological cycle would have left behind, and also proposing new paths for the astrobiological exploration of Mars on the basis of geochemical and geomicrobiological studies in cold aqueous environments. Mission-derived datasets will be used to test hypotheses through paleogeomorphological reconstructions, theoretical modeling and experiments in the laboratory.
Summary
Geologic evidence indicative of flowing and ponding liquid water on the surface of ancient Mars appears abundantly across most of the Martian landscape, indicating that liquid water was present in variable amounts and for long periods of time on and/or near the surface at different moments of Mars’ early history, the Noachian era. Early Mars appears to have been “wet”. However, the presence of liquid water on the surface of early Mars is difficult to reconcile with the reduced solar luminosity at 3.8 Ga. and before, which would have imposed mean temperatures below freezing all over the planet. Atmospheric greenhouse gases and carbon dioxide ice clouds in the upper troposphere are suggested to provide over freezing temperatures, explaining some of this discrepancy, but these solutions have been probed to face numerous problems. So, it is difficult to explain the early Martian hydrology invoking global “warm” conditions. Here I propose to conduct interdisciplinary investigations in order to define and test a new hypothesis to understand the early environmental traits on Mars: that the young Martian surface was characterized by global mean freezing conditions, as predicted by climate models, and at the same time a vigorous hydrogeological cycle was active during hundreds of millions of years, as confirmed by geomorphological and mineralogical analyses. The aim of this investigation is to comprehensively analyze the triggers, traits and consequences of a cold aqueous environment dominating the Noachian, studying the geomorphological, mineralogical and geochemical evidences that such a hydrological cycle would have left behind, and also proposing new paths for the astrobiological exploration of Mars on the basis of geochemical and geomicrobiological studies in cold aqueous environments. Mission-derived datasets will be used to test hypotheses through paleogeomorphological reconstructions, theoretical modeling and experiments in the laboratory.
Max ERC Funding
1 411 200 €
Duration
Start date: 2014-01-01, End date: 2018-12-31
Project acronym iGBMavatars
Project Glioblastoma Subtype Avatar models for Target Discovery and Biology
Researcher (PI) Gaetano GARGIULO
Host Institution (HI) MAX DELBRUECK CENTRUM FUER MOLEKULARE MEDIZIN IN DER HELMHOLTZ-GEMEINSCHAFT (MDC)
Call Details Starting Grant (StG), LS4, ERC-2016-STG
Summary The Glioblastoma Multiforme (GBM) is the most common primary brain tumor and it is incurable. Two major challenges affect GBM clinical management: its heterogeneity (which treatment will best fit this very patient?) and its resistance to available treatments (will the patient benefit in any way from the chosen therapy?). Here we approach these questions with a personalized entry point. First, we aim to create “humanized” experimental models of GBM accurately reflecting patients at molecular level. These GBM Subtype Avatars models (GSA) will be exploited as “targeted patients” in personalized biology and intervention studies. Since GBM exists as molecular subtypes with similar histopathology but mutually exclusive genetic lesions and molecular features, we will generate GSA by targeting mutations recurrently associated with Proneural, Classical or Mesenchymal GBM subtypes into adult human neural stem cells (NSC). Evidence supports that these cells can give rise to high-grade gliomas when engineered with the appropriate genetic lesions. Next, engineered NSC will be orthotopically implanted into immunocompromised rats and the resulting tumors profiled for gene expression, DNA methylation and copy number aberrations. These profiles will be compared to those generated in patient-derived xenografts and biopsies. Second, to identify drug targets favoring patients’ response to the current standard of care, we will exploit GSA for state-of-art genetic screens in vivo. Specifically, we will seek for synthetic lethal interactions between DNA damaging agents and the GSA transcriptome using an in vivo CRISPRi screening approach. Third, to investigate the molecular basis of GBM heterogeneity in GSA models, we will combine genetic and immunophenotypic tracing with gene expression and epigenomic profiling. Identifying tumor-specific vulnerabilities in a dismal disease urging for effective therapies and its molecular fingerprinting convey conceivably rapid Translation in Oncology.
Summary
The Glioblastoma Multiforme (GBM) is the most common primary brain tumor and it is incurable. Two major challenges affect GBM clinical management: its heterogeneity (which treatment will best fit this very patient?) and its resistance to available treatments (will the patient benefit in any way from the chosen therapy?). Here we approach these questions with a personalized entry point. First, we aim to create “humanized” experimental models of GBM accurately reflecting patients at molecular level. These GBM Subtype Avatars models (GSA) will be exploited as “targeted patients” in personalized biology and intervention studies. Since GBM exists as molecular subtypes with similar histopathology but mutually exclusive genetic lesions and molecular features, we will generate GSA by targeting mutations recurrently associated with Proneural, Classical or Mesenchymal GBM subtypes into adult human neural stem cells (NSC). Evidence supports that these cells can give rise to high-grade gliomas when engineered with the appropriate genetic lesions. Next, engineered NSC will be orthotopically implanted into immunocompromised rats and the resulting tumors profiled for gene expression, DNA methylation and copy number aberrations. These profiles will be compared to those generated in patient-derived xenografts and biopsies. Second, to identify drug targets favoring patients’ response to the current standard of care, we will exploit GSA for state-of-art genetic screens in vivo. Specifically, we will seek for synthetic lethal interactions between DNA damaging agents and the GSA transcriptome using an in vivo CRISPRi screening approach. Third, to investigate the molecular basis of GBM heterogeneity in GSA models, we will combine genetic and immunophenotypic tracing with gene expression and epigenomic profiling. Identifying tumor-specific vulnerabilities in a dismal disease urging for effective therapies and its molecular fingerprinting convey conceivably rapid Translation in Oncology.
Max ERC Funding
1 500 000 €
Duration
Start date: 2017-07-01, End date: 2022-06-30
Project acronym IL7sigNETure
Project IL-7/IL-7R signaling networks in health and malignancy
Researcher (PI) João Pedro Taborda Barata
Host Institution (HI) INSTITUTO DE MEDICINA MOLECULAR JOAO LOBO ANTUNES
Call Details Consolidator Grant (CoG), LS4, ERC-2014-CoG
Summary Deregulation of signal transduction is a feature of tumor cells and signaling therapies are gaining importance in the growing arsenal against cancer. However, their full potential can only be achieved once we overcome the limited knowledge on how signaling networks are wired in cancer cells. Interleukin 7 (IL7) and its receptor (IL7R) are essential for normal T-cell development and function. However, they can also promote autoimmunity, chronic inflammation and cancer. We showed that patients with T-cell acute lymphoblastic leukemia (T-ALL), an aggressive hematological cancer, can display IL7R gain-of-function mutations leading to downstream signaling activation and cell transformation. Despite the biological relevance of IL7 and IL7R, the characterization of their signaling effectors remains limited. Here, we propose to move from the single molecule/pathway-centered analysis that has characterized the research on IL7/IL7R signaling, into a ‘holistic’ view of the IL7/IL7R signaling landscape. To do so, we will employ a multidisciplinary strategy, in which data from complementary high throughput analyses, informing on different levels of regulation of the IL7/IL7R signaling network, will be integrated via a systems biology approach, and complemented by cell and molecular biology experimentation and state-of-the-art in vivo models. The knowledge we will generate should have a profound impact on the understanding of the fundamental mechanisms by which IL7/IL7R signaling promotes leukemia and reveal novel targets for fine-tuned therapeutic intervention in T-ALL. Moreover, the scope of insights gained should extend beyond leukemia. Our in-depth, systems-level characterization of IL7/IL7R signaling will constitute a platform with extraordinary potential to illuminate the molecular role of the IL7/IL7R axis in other cancers (e.g. breast and lung) and pathological settings where IL7 has been implicated, such as HIV infection, multiple sclerosis and rheumatoid arthritis.
Summary
Deregulation of signal transduction is a feature of tumor cells and signaling therapies are gaining importance in the growing arsenal against cancer. However, their full potential can only be achieved once we overcome the limited knowledge on how signaling networks are wired in cancer cells. Interleukin 7 (IL7) and its receptor (IL7R) are essential for normal T-cell development and function. However, they can also promote autoimmunity, chronic inflammation and cancer. We showed that patients with T-cell acute lymphoblastic leukemia (T-ALL), an aggressive hematological cancer, can display IL7R gain-of-function mutations leading to downstream signaling activation and cell transformation. Despite the biological relevance of IL7 and IL7R, the characterization of their signaling effectors remains limited. Here, we propose to move from the single molecule/pathway-centered analysis that has characterized the research on IL7/IL7R signaling, into a ‘holistic’ view of the IL7/IL7R signaling landscape. To do so, we will employ a multidisciplinary strategy, in which data from complementary high throughput analyses, informing on different levels of regulation of the IL7/IL7R signaling network, will be integrated via a systems biology approach, and complemented by cell and molecular biology experimentation and state-of-the-art in vivo models. The knowledge we will generate should have a profound impact on the understanding of the fundamental mechanisms by which IL7/IL7R signaling promotes leukemia and reveal novel targets for fine-tuned therapeutic intervention in T-ALL. Moreover, the scope of insights gained should extend beyond leukemia. Our in-depth, systems-level characterization of IL7/IL7R signaling will constitute a platform with extraordinary potential to illuminate the molecular role of the IL7/IL7R axis in other cancers (e.g. breast and lung) and pathological settings where IL7 has been implicated, such as HIV infection, multiple sclerosis and rheumatoid arthritis.
Max ERC Funding
1 988 125 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym IM Targeting CVD
Project Accelerated Atherosclerosis in Patients with Immune Mediated Disorders as a Model to Investigate the Link between Inflammation and Cardiovascular Disease: From Basic Mechanisms to Clinical Application
Researcher (PI) Ziad Mallat
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary Atherosclerosis is a chronic inflammatory disease of the arterial wall with (auto)immune component, initiated in response to modified (phospho)lipids. Despite important advances in our understanding of the inflammatory response in atherosclerosis, the critical pathways responsible for the breakdown of immune tolerance to lipoproteins and other self-antigens remain largely unknown. An important feature of ruptured/thrombosed atherosclerotic lesions is the accumulation of apoptotic, and secondary necrotic, lipid-laden macrophages and smooth muscle cells due to defective efferocytosis (clearance of apoptotic cells). This leads to the formation of a large ‘necrotic’ lipid core, associated with enhanced vascular inflammation. Interestingly, defective efferocytosis has been associated with the development of autoimmunity, and patients with systemic lupus erythematosus who show increased accumulation of apoptotic material are at very high risk of accelerated atherosclerosis and myocardial infarction. We hypothesize that accumulation of apoptotic/secondary necrotic cells due to defective efferocytosis, together with modified lipids, activate critical immuno-inflammatory pathways in macrophages and B cells, and break immune tolerance in atherosclerosis and post-myocardial infarction. This is consistent with the critical role played by defective efferocytosis and macrophage activation in atherosclerotic lesion progression, and with our recent unsuspected data showing a critical role for B cell activation in driving lesion development in several models of atherosclerosis. We also propose that interactions between macrophages and B cells are essential for the perpetuation of the pathogenic immuno-inflammatory response in cardiovascular disease. Finally, we will harness this knowledge for a better identification of patients at risk of cardiovascular complications, and will target these pathways to limit the progression and complications of cardiovascular disease.
Summary
Atherosclerosis is a chronic inflammatory disease of the arterial wall with (auto)immune component, initiated in response to modified (phospho)lipids. Despite important advances in our understanding of the inflammatory response in atherosclerosis, the critical pathways responsible for the breakdown of immune tolerance to lipoproteins and other self-antigens remain largely unknown. An important feature of ruptured/thrombosed atherosclerotic lesions is the accumulation of apoptotic, and secondary necrotic, lipid-laden macrophages and smooth muscle cells due to defective efferocytosis (clearance of apoptotic cells). This leads to the formation of a large ‘necrotic’ lipid core, associated with enhanced vascular inflammation. Interestingly, defective efferocytosis has been associated with the development of autoimmunity, and patients with systemic lupus erythematosus who show increased accumulation of apoptotic material are at very high risk of accelerated atherosclerosis and myocardial infarction. We hypothesize that accumulation of apoptotic/secondary necrotic cells due to defective efferocytosis, together with modified lipids, activate critical immuno-inflammatory pathways in macrophages and B cells, and break immune tolerance in atherosclerosis and post-myocardial infarction. This is consistent with the critical role played by defective efferocytosis and macrophage activation in atherosclerotic lesion progression, and with our recent unsuspected data showing a critical role for B cell activation in driving lesion development in several models of atherosclerosis. We also propose that interactions between macrophages and B cells are essential for the perpetuation of the pathogenic immuno-inflammatory response in cardiovascular disease. Finally, we will harness this knowledge for a better identification of patients at risk of cardiovascular complications, and will target these pathways to limit the progression and complications of cardiovascular disease.
Max ERC Funding
1 499 731 €
Duration
Start date: 2012-04-01, End date: 2017-03-31
Project acronym ImagePlanetFormDiscs
Project Imaging the Dynamical Imprints of Planet Formation in Protoplanetary Discs
Researcher (PI) Stefan Kraus
Host Institution (HI) THE UNIVERSITY OF EXETER
Call Details Starting Grant (StG), PE9, ERC-2014-STG
Summary The gas and dust discs around young stars are thought to be the birthplace of planetary systems and are a key area to study if further progress is to be made on understanding the history of our solar system and our own origins. Once planets have formed in these discs, they dynamically sculpt their environment, for instance by opening tidally-cleared gaps or triggering spiral arms and disc warps. The late stages of this process are likely observed in the “transitional” discs, where regions spanning tens of astronomical units (AU) have been cleared. The aim of this project is to image the planet formation signatures both during the transitional disc and the earlier T Tauri or Herbig Ae/Be stars phase, where the protoplanetary bodies are just starting to carve gaps in the optically thick disc. For this purpose, we will employ the latest generation of near-infrared, mid-infrared, and sub-millimeter interferometric instruments that will allow us to trace a wide range of stellocentric radii, disc scale heights, and dust opacities. We will make use of recent revolutionary advancements in infrared detector technology and equip the CHARA/MIRC 6-telescope beam combiner with a low-read noise camera that will significantly increase the sensitivity of this instrument and enable us to image protoplanetary discs with 2.5 times higher resolution and much higher efficiency than ever before. These quick-look imaging capabilities will enable us to trace time-variable structures in the inner few AU and to investigate their relation to the commonly observed photometric and spectroscopic variability. Our interferometric observations in spectral lines aim to detect the accretion signatures of the young protoplanets themselves. Employing sophisticated radiation hydrodynamics simulations we will achieve an unprecedented global view on protoplanetary disc structure and obtain fundamentally new constraints on theoretical models of planet formation, planet-disc interaction, and disc evolution.
Summary
The gas and dust discs around young stars are thought to be the birthplace of planetary systems and are a key area to study if further progress is to be made on understanding the history of our solar system and our own origins. Once planets have formed in these discs, they dynamically sculpt their environment, for instance by opening tidally-cleared gaps or triggering spiral arms and disc warps. The late stages of this process are likely observed in the “transitional” discs, where regions spanning tens of astronomical units (AU) have been cleared. The aim of this project is to image the planet formation signatures both during the transitional disc and the earlier T Tauri or Herbig Ae/Be stars phase, where the protoplanetary bodies are just starting to carve gaps in the optically thick disc. For this purpose, we will employ the latest generation of near-infrared, mid-infrared, and sub-millimeter interferometric instruments that will allow us to trace a wide range of stellocentric radii, disc scale heights, and dust opacities. We will make use of recent revolutionary advancements in infrared detector technology and equip the CHARA/MIRC 6-telescope beam combiner with a low-read noise camera that will significantly increase the sensitivity of this instrument and enable us to image protoplanetary discs with 2.5 times higher resolution and much higher efficiency than ever before. These quick-look imaging capabilities will enable us to trace time-variable structures in the inner few AU and to investigate their relation to the commonly observed photometric and spectroscopic variability. Our interferometric observations in spectral lines aim to detect the accretion signatures of the young protoplanets themselves. Employing sophisticated radiation hydrodynamics simulations we will achieve an unprecedented global view on protoplanetary disc structure and obtain fundamentally new constraints on theoretical models of planet formation, planet-disc interaction, and disc evolution.
Max ERC Funding
1 648 265 €
Duration
Start date: 2015-07-01, End date: 2020-06-30
Project acronym imbh
Project Do intermediate-mass black holes exist?
Researcher (PI) Peter Gustaaf Jonker
Host Institution (HI) STICHTING NEDERLANDSE WETENSCHAPPELIJK ONDERZOEK INSTITUTEN
Call Details Consolidator Grant (CoG), PE9, ERC-2014-CoG
Summary With this proposed project I will determine whether intermediate-mass black holes (IMBHs) exist. I propose to use ESA's new Gaia mission, the rich Hubble Space Telescope data archive, and state-of-the-art techniques to investigate systems predicted to exist but not yet found hitherto, such as recoiled hyper-compact stellar systems, red-supergiant mass donors to ultra-luminous X-ray sources, and white dwarf tidal disruption events. The latter can only be detected if black holes with masses less than 1E5 Msun are involved. Using these systems and events we can probe the sphere of influence of the IMBH and determine the black hole mass dynamically.
Currently, there are strong indications for the existence of IMBHs, but dynamical evidence, the irrefutable proof of their existence, is still lacking. Whereas the unequivocal detection of an IMBH will be a breakthrough discovery in itself, it has also important consequences for searches of dark matter annihilation signals, it will provide a baseline for the rate predictions of gravitational wave radiation events involving IMBHs, and the properties of a population of IMBHs provides important constraints on the growth of supermassive black holes and galaxies. Finally, if we discover IMBHs in hyper-compact star clusters it validates numerical relativity simulations that predict that merging black holes receive a recoil kick.
My membership of Gaia's Data Processing and Analysis Consortium gives me a distinct advantage in analysing and interpreting Gaia data that, through the superb angular resolution, immediate spectroscopic observations and all-sky coverage, provides unique capabilities ideally suited for answering the question whether IMBHs exist.
My proposed project is the first to recognize the potential of Gaia (WP1&2) as well as the implications of having red supergiant mass donors in some ultra-luminous X-ray sources (WP3) for answering the question on the existence of IMBHs.
Summary
With this proposed project I will determine whether intermediate-mass black holes (IMBHs) exist. I propose to use ESA's new Gaia mission, the rich Hubble Space Telescope data archive, and state-of-the-art techniques to investigate systems predicted to exist but not yet found hitherto, such as recoiled hyper-compact stellar systems, red-supergiant mass donors to ultra-luminous X-ray sources, and white dwarf tidal disruption events. The latter can only be detected if black holes with masses less than 1E5 Msun are involved. Using these systems and events we can probe the sphere of influence of the IMBH and determine the black hole mass dynamically.
Currently, there are strong indications for the existence of IMBHs, but dynamical evidence, the irrefutable proof of their existence, is still lacking. Whereas the unequivocal detection of an IMBH will be a breakthrough discovery in itself, it has also important consequences for searches of dark matter annihilation signals, it will provide a baseline for the rate predictions of gravitational wave radiation events involving IMBHs, and the properties of a population of IMBHs provides important constraints on the growth of supermassive black holes and galaxies. Finally, if we discover IMBHs in hyper-compact star clusters it validates numerical relativity simulations that predict that merging black holes receive a recoil kick.
My membership of Gaia's Data Processing and Analysis Consortium gives me a distinct advantage in analysing and interpreting Gaia data that, through the superb angular resolution, immediate spectroscopic observations and all-sky coverage, provides unique capabilities ideally suited for answering the question whether IMBHs exist.
My proposed project is the first to recognize the potential of Gaia (WP1&2) as well as the implications of having red supergiant mass donors in some ultra-luminous X-ray sources (WP3) for answering the question on the existence of IMBHs.
Max ERC Funding
1 999 975 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym IMMCEPTION
Project Nociception and sensory nerves as regulators of type 2 immunity and skin inflammation
Researcher (PI) Nicolas GAUDENZIO
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS4, ERC-2018-STG
Summary Preserving skin homeostasis depends on complex interactions among structural cells, immune cells, and the environment. Dysregulation of this delicate equilibrium contributes to the development of type 2 immunity-associated skin inflammation (i.e., allergic skin inflammation), including atopic dermatitis (AD). The skin is a complex organ harboring various tissue-resident immune cells (e.g., dendritic cells, mast cells and macrophages) and innervated by a meshwork of sensory nerves, including those involved in nociception (i.e., nociceptors), which respond to injurious or potentially damaging stimuli by transmitting signals to the spinal cord and brain. Despite their role in the transmission of sensation, recent evidences have suggested that nociceptors could be powerful regulators of ongoing immune response.
We wish to use sophisticated mouse models and new in vivo imaging approaches to define the roles of subsets of dermal nociceptors, cationic neuropeptide substance P, dermal mast cells expressing the recently discovered receptor for cationic molecules Mas-related G protein-coupled receptor b2 (i.e., Mrgprb2), in a mouse model of AD that has many pathological, immunological, and gene expression similarities with the corresponding human disorder. We also will define the translational relevance of our mouse studies by performing parallel analyzes of nociceptors and mast cells in the lesional skin of patients from USA and France with clinically-established AD. To accomplish these goals, we have proposed herein a body of work that is solidly based on our preliminary data, with four Aims that will test innovative hypotheses by using informative genetic approaches, as well as new intravital imaging systems we recently developed.
This work thus will address significant gaps in our knowledge about the pathophysiology of AD and has the potential to identify such neuro-immune interactions as a promising new therapeutic target in AD and perhaps other allergic disorders.
Summary
Preserving skin homeostasis depends on complex interactions among structural cells, immune cells, and the environment. Dysregulation of this delicate equilibrium contributes to the development of type 2 immunity-associated skin inflammation (i.e., allergic skin inflammation), including atopic dermatitis (AD). The skin is a complex organ harboring various tissue-resident immune cells (e.g., dendritic cells, mast cells and macrophages) and innervated by a meshwork of sensory nerves, including those involved in nociception (i.e., nociceptors), which respond to injurious or potentially damaging stimuli by transmitting signals to the spinal cord and brain. Despite their role in the transmission of sensation, recent evidences have suggested that nociceptors could be powerful regulators of ongoing immune response.
We wish to use sophisticated mouse models and new in vivo imaging approaches to define the roles of subsets of dermal nociceptors, cationic neuropeptide substance P, dermal mast cells expressing the recently discovered receptor for cationic molecules Mas-related G protein-coupled receptor b2 (i.e., Mrgprb2), in a mouse model of AD that has many pathological, immunological, and gene expression similarities with the corresponding human disorder. We also will define the translational relevance of our mouse studies by performing parallel analyzes of nociceptors and mast cells in the lesional skin of patients from USA and France with clinically-established AD. To accomplish these goals, we have proposed herein a body of work that is solidly based on our preliminary data, with four Aims that will test innovative hypotheses by using informative genetic approaches, as well as new intravital imaging systems we recently developed.
This work thus will address significant gaps in our knowledge about the pathophysiology of AD and has the potential to identify such neuro-immune interactions as a promising new therapeutic target in AD and perhaps other allergic disorders.
Max ERC Funding
1 497 441 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym Immune-senescence
Project Dual targeting of senescence and tumor immunity for cancer therapy
Researcher (PI) Andrea Alimonti
Host Institution (HI) FONDAZIONE PER L'ISTITUTO ONCOLOGICO DI RICERCA (IOR)
Call Details Consolidator Grant (CoG), LS4, ERC-2015-CoG
Summary We have previously demonstrated that cellular senescence opposes tumorigenesis thereby opening up new potential opportunities for cancer treatment. Senescence and tumor immunity in cancer are tightly interconnected. Tumor-infiltrating immune cells promote the clearance of senescent tumor cells thereby contributing to the tumor suppressive function of senescence. Moreover, T lymphocytes can drive senescence in cancers by secreting different cytokines in the tumor microenvironment. We have also recently reported that GR1+ myeloid cells antagonize treatment-induced senescence (TIS) and that compounds that block the tumor recruitment of GR1+ cells enhance TIS. Major objective of this proposal is to characterize the immune landscape of different prostate cancer mouse models in order to develop novel treatment modalities that combine pro-senescence compounds with immunotherapy. Using proteomics and bioinformatics approaches, we will assess how the genetic background of prostate tumors, shapes the tumor microenvironment and immune response during TIS. Next, we will define the mechanisms that regulate the recruitment and activation of myeloid derived suppressive cells, macrophages and B-lymphocytes in Pten deficient prostate tumors by focusing on a novel class of secreted factors identified in these tumors. We will also assess in vivo whether the secretome of tumor cells can transmit senescence to TILs and compounds that interfere with the secretome can prevent immunosenescence. Finally, we will develop monoclonal antibodies directed towards senescent tumors cells that we will use as diagnostic and therapeutic tools. These antibodies will be used as biomarkers to detect senescent tumor cells in prostate cancers and will be tested in pre-clinical trials to assess whether they improve tumor clearance during TIS. Our findings will form the basis for future clinical trials in prostate cancer patients.
Summary
We have previously demonstrated that cellular senescence opposes tumorigenesis thereby opening up new potential opportunities for cancer treatment. Senescence and tumor immunity in cancer are tightly interconnected. Tumor-infiltrating immune cells promote the clearance of senescent tumor cells thereby contributing to the tumor suppressive function of senescence. Moreover, T lymphocytes can drive senescence in cancers by secreting different cytokines in the tumor microenvironment. We have also recently reported that GR1+ myeloid cells antagonize treatment-induced senescence (TIS) and that compounds that block the tumor recruitment of GR1+ cells enhance TIS. Major objective of this proposal is to characterize the immune landscape of different prostate cancer mouse models in order to develop novel treatment modalities that combine pro-senescence compounds with immunotherapy. Using proteomics and bioinformatics approaches, we will assess how the genetic background of prostate tumors, shapes the tumor microenvironment and immune response during TIS. Next, we will define the mechanisms that regulate the recruitment and activation of myeloid derived suppressive cells, macrophages and B-lymphocytes in Pten deficient prostate tumors by focusing on a novel class of secreted factors identified in these tumors. We will also assess in vivo whether the secretome of tumor cells can transmit senescence to TILs and compounds that interfere with the secretome can prevent immunosenescence. Finally, we will develop monoclonal antibodies directed towards senescent tumors cells that we will use as diagnostic and therapeutic tools. These antibodies will be used as biomarkers to detect senescent tumor cells in prostate cancers and will be tested in pre-clinical trials to assess whether they improve tumor clearance during TIS. Our findings will form the basis for future clinical trials in prostate cancer patients.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym ImmunoBile
Project Bile acid, immune-metabolism, lipid and glucose homeostasis
Researcher (PI) Bart STAELS
Host Institution (HI) UNIVERSITE DE LILLE
Call Details Advanced Grant (AdG), LS4, ERC-2015-AdG
Summary The role of chronic inflammation in obesity, metabolic and cardiovascular diseases is increasingly recognized. Bile acids (BA), synthesized in the liver and modified by the gut flora, facilitate lipid absorption in the intestine. BA modulate lipid and glucose homeostasis by activating the nuclear receptor FXR and the GPCR TGR5. Intriguingly, peripheral BA concentrations are elevated in type 2 diabetes (T2D) and FXR mediates the beneficial metabolic response to gastric bypass in mice. The immune system plays an important role in the cross-talk with metabolic tissues, such as liver, intestine and adipose tissues. However, whether BA modulate immune cell function is unknown. Our unpublished results identifying FXR and TGR5 expression in lymphoid cells, prompt us to study their role in the regulation of glucose and lipid metabolism through immune cell modulation. Using reporter mice and specific ligands, we will characterize the immune cells expressing active FXR and TGR5. We will determine their role in metabolism and inflammation by immune cell-specific gene inactivation in models of obesity, T2D and elevated peripheral blood BA concentrations. Mass cytometry, cell sorting and single cell transcriptomic analysis will allow the identification of gene networks regulated by BA and their receptors. As microbiota generate biologically active secondary BA, we will assess the impact of microbiota depletion and subsequent BA acid pool modifications on immune cell populations. Translational studies in humans with altered BA metabolism and pharmacological treatment with anti-diabetic BA sequestrants will allow assessment of alterations in immune functions. This project aims to identify an hitherto unexplored role of BA through modulation of the immune system on T2D, NAFLD and dyslipidemia. Success of the project critically depends on an integrative approach uniquely undertaken in my laboratory through its unique multidisciplinary expertise in basic and translational biology.
Summary
The role of chronic inflammation in obesity, metabolic and cardiovascular diseases is increasingly recognized. Bile acids (BA), synthesized in the liver and modified by the gut flora, facilitate lipid absorption in the intestine. BA modulate lipid and glucose homeostasis by activating the nuclear receptor FXR and the GPCR TGR5. Intriguingly, peripheral BA concentrations are elevated in type 2 diabetes (T2D) and FXR mediates the beneficial metabolic response to gastric bypass in mice. The immune system plays an important role in the cross-talk with metabolic tissues, such as liver, intestine and adipose tissues. However, whether BA modulate immune cell function is unknown. Our unpublished results identifying FXR and TGR5 expression in lymphoid cells, prompt us to study their role in the regulation of glucose and lipid metabolism through immune cell modulation. Using reporter mice and specific ligands, we will characterize the immune cells expressing active FXR and TGR5. We will determine their role in metabolism and inflammation by immune cell-specific gene inactivation in models of obesity, T2D and elevated peripheral blood BA concentrations. Mass cytometry, cell sorting and single cell transcriptomic analysis will allow the identification of gene networks regulated by BA and their receptors. As microbiota generate biologically active secondary BA, we will assess the impact of microbiota depletion and subsequent BA acid pool modifications on immune cell populations. Translational studies in humans with altered BA metabolism and pharmacological treatment with anti-diabetic BA sequestrants will allow assessment of alterations in immune functions. This project aims to identify an hitherto unexplored role of BA through modulation of the immune system on T2D, NAFLD and dyslipidemia. Success of the project critically depends on an integrative approach uniquely undertaken in my laboratory through its unique multidisciplinary expertise in basic and translational biology.
Max ERC Funding
2 500 000 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym IMMUNOBIOTA
Project Host-microbiota interactions across the gut immune system:
lessons from early onset inflammatory bowel diseases
and from gnotobiotic mice
Researcher (PI) Nadine Christine Cerf Spouse Bensussan
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS4, ERC-2013-ADG
Summary "How the immune system controls the partnership established between mammalian hosts and the complex microbial community that colonizes their distal intestine is a fascinating topic with wide implications in physiology and pathology. In order to delineate how the two partners build and maintain mutualistic relationships, we intend to combine bench to bed-site approaches in humans and mechanistic studies in mice.
The first part of the proposal aims at dissecting human host pathways mandatory to maintain homeostatic relationships with the microbiota through the analysis of a cohort of young children with colites of early onset likely caused by Mendelian mutations. In this part, we expect to take advantage from recent development of powerful tools in genome analysis to extend our expertise in human intestinal immunopathology, provide novel basic insight into immuno-regulation in the human gut and delineate new tools and strategies for the diagnosis and care of inflammatory bowel diseases in children.
In the second part of the proposal, we intend to capitalise on our recent results highlighting the key role of Segmented Filamentous Bacterium in the post-natal maturation of the gut immune system in mice and on very recent evidence of a host-specific version of this bacterium in the human microbiota. First, we want to combine analyses in gnotobiotic mice and molecular approaches to provide a comprehensive view of the mechanisms underlying the outstanding immunostimulatory functions of this unusual symbiont. We hope thereby to identify the bacterial attributes that drive the physiological activation of the host immune system and to gain insight into the rules that govern host-microbiota interactions. Second, we intend to put all possible efforts to characterise the human version of Segmented Filamentous bacterium and to analyse how this bacterium may influence the normal or pathological development of the gut immune system in humans."
Summary
"How the immune system controls the partnership established between mammalian hosts and the complex microbial community that colonizes their distal intestine is a fascinating topic with wide implications in physiology and pathology. In order to delineate how the two partners build and maintain mutualistic relationships, we intend to combine bench to bed-site approaches in humans and mechanistic studies in mice.
The first part of the proposal aims at dissecting human host pathways mandatory to maintain homeostatic relationships with the microbiota through the analysis of a cohort of young children with colites of early onset likely caused by Mendelian mutations. In this part, we expect to take advantage from recent development of powerful tools in genome analysis to extend our expertise in human intestinal immunopathology, provide novel basic insight into immuno-regulation in the human gut and delineate new tools and strategies for the diagnosis and care of inflammatory bowel diseases in children.
In the second part of the proposal, we intend to capitalise on our recent results highlighting the key role of Segmented Filamentous Bacterium in the post-natal maturation of the gut immune system in mice and on very recent evidence of a host-specific version of this bacterium in the human microbiota. First, we want to combine analyses in gnotobiotic mice and molecular approaches to provide a comprehensive view of the mechanisms underlying the outstanding immunostimulatory functions of this unusual symbiont. We hope thereby to identify the bacterial attributes that drive the physiological activation of the host immune system and to gain insight into the rules that govern host-microbiota interactions. Second, we intend to put all possible efforts to characterise the human version of Segmented Filamentous bacterium and to analyse how this bacterium may influence the normal or pathological development of the gut immune system in humans."
Max ERC Funding
2 499 551 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym IMMUNODEATH
Project Immunogenic cell death in anticancer therapy
Researcher (PI) Guido Peter Krömer
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS4, ERC-2012-ADG_20120314
Summary We advocate the hypothesis that successful chemotherapeutics can induce a type of tumor cell stress and death that is immunogenic, meaning that the patient’s dying cancer cells serve as a vaccine that stimulates a specific antitumor immune response, which in turn can control (and sometimes even eradicate) residual cancer cells. This is a highly original – and necessarily controversial – “breakthrough” concept since it challenges previous belief that anticancer chemotherapies act solely on the tumor cells, without any significant involvement of the host immune system. Cell death is usually non-immunogenic, and only a small minority of chemotherapeutic agents can induce immunogenic cell death, which - in contrast to classical apoptosis - is preceded by two types of pre-mortem stress, autophagy (which is required for cellular ATP release, an obligatory signal of immunogenicity) and endoplasmic reticulum (ER) stress (which is required for calreticulin [CRT] exposure at the cell surface, another obligatory signal of immunogenicity). Here, we will explore the hypothesis that cancer cell death is only immunogenic if the two pathways of pre-mortem stress, autophagy and ER stress, are simultaneously activated. Thus, we aim at “decoding” the anticancer drug-induced cellular pathways that regulate the immunogenicity of cell death. For this, we will trigger cancer cell death preceded by one or the two types of pre-mortem stress in a “synthetic system” (by genetic manipulation involving inducible transgenes in cancer cells and mice) or by means of selected pharmacological compounds in multiple in vitro and in vivo cancer models, as we monitor the immune-dependent therapeutic response. Moreover, we will investigate the functional links between autophagy, ER stress and immunogenic signaling. Finally, we will explore the translational relevance of these findings on human cancers.
Summary
We advocate the hypothesis that successful chemotherapeutics can induce a type of tumor cell stress and death that is immunogenic, meaning that the patient’s dying cancer cells serve as a vaccine that stimulates a specific antitumor immune response, which in turn can control (and sometimes even eradicate) residual cancer cells. This is a highly original – and necessarily controversial – “breakthrough” concept since it challenges previous belief that anticancer chemotherapies act solely on the tumor cells, without any significant involvement of the host immune system. Cell death is usually non-immunogenic, and only a small minority of chemotherapeutic agents can induce immunogenic cell death, which - in contrast to classical apoptosis - is preceded by two types of pre-mortem stress, autophagy (which is required for cellular ATP release, an obligatory signal of immunogenicity) and endoplasmic reticulum (ER) stress (which is required for calreticulin [CRT] exposure at the cell surface, another obligatory signal of immunogenicity). Here, we will explore the hypothesis that cancer cell death is only immunogenic if the two pathways of pre-mortem stress, autophagy and ER stress, are simultaneously activated. Thus, we aim at “decoding” the anticancer drug-induced cellular pathways that regulate the immunogenicity of cell death. For this, we will trigger cancer cell death preceded by one or the two types of pre-mortem stress in a “synthetic system” (by genetic manipulation involving inducible transgenes in cancer cells and mice) or by means of selected pharmacological compounds in multiple in vitro and in vivo cancer models, as we monitor the immune-dependent therapeutic response. Moreover, we will investigate the functional links between autophagy, ER stress and immunogenic signaling. Finally, we will explore the translational relevance of these findings on human cancers.
Max ERC Funding
2 500 000 €
Duration
Start date: 2013-04-01, End date: 2018-03-31
Project acronym ImmunoFit
Project Harnessing tumor metabolism to overcome immunosupression
Researcher (PI) Massimiliano MAZZONE
Host Institution (HI) VIB
Call Details Consolidator Grant (CoG), LS4, ERC-2017-COG
Summary Anti-cancer immunotherapy has provided patients with a promising treatment. Yet, it has also unveiled that the immunosuppressive tumor microenvironment (TME) hampers the efficiency of this therapeutic option and limits its success. The concept that metabolism is able to shape the immune response has gained general acceptance. Nonetheless, little is known on how the metabolic crosstalk between different tumor compartments contributes to the harsh TME and ultimately impairs T cell fitness within the tumor.
This proposal aims to decipher which metabolic changes in the TME impede proper anti-tumor immunity. Starting from the meta-analysis of public human datasets, corroborated by metabolomics and transcriptomics data from several mouse tumors, we ranked clinically relevant and altered metabolic pathways that correlate with resistance to immunotherapy. Using a CRISPR/Cas9 platform for their functional in vivo selection, we want to identify cancer cell intrinsic metabolic mediators and, indirectly, distinguish those belonging specifically to the stroma. By means of genetic tools and small molecules, we will modify promising metabolic pathways in cancer cells and stromal cells (particularly in tumor-associated macrophages) to harness tumor immunosuppression. In a mirroring approach, we will apply a similar screening tool on cytotoxic T cells to identify metabolic targets that enhance their fitness under adverse growth conditions. This will allow us to manipulate T cells ex vivo and to therapeutically intervene via adoptive T cell transfer. By analyzing the metabolic network and crosstalk within the tumor, this project will shed light on how metabolism contributes to the immunosuppressive TME and T cell maladaptation. The overall goal is to identify druggable metabolic targets that i) reinforce the intrinsic anti-tumor immune response by breaking immunosuppression and ii) promote T cell function in immunotherapeutic settings by rewiring either the TME or the T cell itself.
Summary
Anti-cancer immunotherapy has provided patients with a promising treatment. Yet, it has also unveiled that the immunosuppressive tumor microenvironment (TME) hampers the efficiency of this therapeutic option and limits its success. The concept that metabolism is able to shape the immune response has gained general acceptance. Nonetheless, little is known on how the metabolic crosstalk between different tumor compartments contributes to the harsh TME and ultimately impairs T cell fitness within the tumor.
This proposal aims to decipher which metabolic changes in the TME impede proper anti-tumor immunity. Starting from the meta-analysis of public human datasets, corroborated by metabolomics and transcriptomics data from several mouse tumors, we ranked clinically relevant and altered metabolic pathways that correlate with resistance to immunotherapy. Using a CRISPR/Cas9 platform for their functional in vivo selection, we want to identify cancer cell intrinsic metabolic mediators and, indirectly, distinguish those belonging specifically to the stroma. By means of genetic tools and small molecules, we will modify promising metabolic pathways in cancer cells and stromal cells (particularly in tumor-associated macrophages) to harness tumor immunosuppression. In a mirroring approach, we will apply a similar screening tool on cytotoxic T cells to identify metabolic targets that enhance their fitness under adverse growth conditions. This will allow us to manipulate T cells ex vivo and to therapeutically intervene via adoptive T cell transfer. By analyzing the metabolic network and crosstalk within the tumor, this project will shed light on how metabolism contributes to the immunosuppressive TME and T cell maladaptation. The overall goal is to identify druggable metabolic targets that i) reinforce the intrinsic anti-tumor immune response by breaking immunosuppression and ii) promote T cell function in immunotherapeutic settings by rewiring either the TME or the T cell itself.
Max ERC Funding
1 999 721 €
Duration
Start date: 2018-07-01, End date: 2023-06-30
Project acronym IMMUNOTHROMBOSIS
Project Cross-talk between platelets and immunity - implications for host homeostasis and defense
Researcher (PI) Steffen MASSBERG
Host Institution (HI) LUDWIG-MAXIMILIANS-UNIVERSITAET MUENCHEN
Call Details Advanced Grant (AdG), LS4, ERC-2018-ADG
Summary The overall aim of the IMMUNOTHROMBOSIS project is to clarify the mechanisms underlying the recently identified synergism between thrombosis and inflammation. Thrombus formation and inflammation are vital host responses that ensure homeostasis, but can also drive cardiovascular disease, including myocardial infarction and stroke, the major causes of death in Europe. My group and others discovered, that thrombosis and inflammation are not to be considered separate processes. They are tightly interrelated and synergize in immune defence, but also in inflammatory and thrombotic diseases in a process we termed immunothrombosis. Targeting this synergism has great potential to identify innovative and unconventional strategies to more specifically prevent undesired activation of thrombotic and inflammatory pathways. However, this requires a deeper mechanistic understanding of immunothrombosis. I recently identified two ground-breaking novel immunothrombotic principles: I discovered that platelets have the ability to migrate autonomously, which assists immune cells in fighting pathogens. Further, I revealed that immune cells play a central role in controlling the production of platelets from their megakaryocyte precursors. The physiological and pathophysiological relevance of both processes is unclear. This is the starting point and focus of the IMMUNOTHROMBOSIS project. My aim is to define how platelets use their ability to migrate to support immune cells in protection of vascular integrity (objective 1) and to identify the contribution of platelet migration to different cardiovascular diseases involving immunothrombotic tissue damage (objective 2). Finally, I will clarify how inflammatory responses feedback to the production of thrombotic effectors and dissect inflammatory mechanisms that control platelet production (objective 3). IMMUNOTHROMBOSIS will identify new options for specific prevention or treatment of thrombotic and inflammatory cardiovascular diseases.
Summary
The overall aim of the IMMUNOTHROMBOSIS project is to clarify the mechanisms underlying the recently identified synergism between thrombosis and inflammation. Thrombus formation and inflammation are vital host responses that ensure homeostasis, but can also drive cardiovascular disease, including myocardial infarction and stroke, the major causes of death in Europe. My group and others discovered, that thrombosis and inflammation are not to be considered separate processes. They are tightly interrelated and synergize in immune defence, but also in inflammatory and thrombotic diseases in a process we termed immunothrombosis. Targeting this synergism has great potential to identify innovative and unconventional strategies to more specifically prevent undesired activation of thrombotic and inflammatory pathways. However, this requires a deeper mechanistic understanding of immunothrombosis. I recently identified two ground-breaking novel immunothrombotic principles: I discovered that platelets have the ability to migrate autonomously, which assists immune cells in fighting pathogens. Further, I revealed that immune cells play a central role in controlling the production of platelets from their megakaryocyte precursors. The physiological and pathophysiological relevance of both processes is unclear. This is the starting point and focus of the IMMUNOTHROMBOSIS project. My aim is to define how platelets use their ability to migrate to support immune cells in protection of vascular integrity (objective 1) and to identify the contribution of platelet migration to different cardiovascular diseases involving immunothrombotic tissue damage (objective 2). Finally, I will clarify how inflammatory responses feedback to the production of thrombotic effectors and dissect inflammatory mechanisms that control platelet production (objective 3). IMMUNOTHROMBOSIS will identify new options for specific prevention or treatment of thrombotic and inflammatory cardiovascular diseases.
Max ERC Funding
2 321 416 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym INFANTLEUKEMIA
Project GENOMIC, CELLULAR AND DEVELOPMENTAL RECONSTRUCTION OFINFANT MLL-AF4+ ACUTE LYMPHOBLASTIC LEUKEMIA
Researcher (PI) Pablo Menendez Buján
Host Institution (HI) FUNDACIO INSTITUT DE RECERCA CONTRA LA LEUCEMIA JOSEP CARRERAS
Call Details Consolidator Grant (CoG), LS4, ERC-2014-CoG
Summary Infant cancer is very distinct to adult cancer and it is progressively seen as a developmental disease. An intriguing infant cancer is the t(4;11) acute lymphoblastic leukemia (ALL) characterized by the hallmark rearrangement MLL-AF4 (MA4), and associated with dismal prognosis. The 100% concordance in twins and its prenatal onset suggest an extremely rapid disease progression. Many key issues remain elusive:
Is MA4 leukemogenic?
Which are other relevant oncogenic drivers?
Which is the nature of the cell transformed by MA4?
Which is the leukemia-initiating cell (LIC)?
Does this ALL follow a hierarchical or stochastic cancer model?
How to explain therapy resistance and CNS involvement?
To what extent do genetics vs epigenetics contribute this ALL?
These questions remain a challenge due to: 1) the absence of prospective studies on diagnostic/remission-matched samples, 2) the lack of models which faithfully reproduce the disease and 3) a surprising genomic stability of this ALL.
I hypothesize that a Multilayer-Omics to function approach in patient blasts and early human hematopoietic stem/progenitor cells (HSPC) is required to fully scrutinize the biology underlying this life-threatening leukemia. I will perform genome-wide studies on the mutational landscape, DNA and H3K79 methylation profiles, and transcriptome on a uniquely available, large cohort of diagnostic/remission-matched samples. Omics data integration will provide unprecedented information about oncogenic drivers which must be analyzed in ground-breaking functional assays using patient blasts and early HSPCs carrying a CRISPR/Cas9-mediated locus/allele-specific t(4;11). Serial xenografts combined with exome-seq in paired diagnostic samples and xenografts will identify the LIC and determine whether variegated genetics may underlie clonal functional heterogeneity. This project will provide a precise understanding and a disease model for MA4+ ALL, offering a platform for new treatment strategies.
Summary
Infant cancer is very distinct to adult cancer and it is progressively seen as a developmental disease. An intriguing infant cancer is the t(4;11) acute lymphoblastic leukemia (ALL) characterized by the hallmark rearrangement MLL-AF4 (MA4), and associated with dismal prognosis. The 100% concordance in twins and its prenatal onset suggest an extremely rapid disease progression. Many key issues remain elusive:
Is MA4 leukemogenic?
Which are other relevant oncogenic drivers?
Which is the nature of the cell transformed by MA4?
Which is the leukemia-initiating cell (LIC)?
Does this ALL follow a hierarchical or stochastic cancer model?
How to explain therapy resistance and CNS involvement?
To what extent do genetics vs epigenetics contribute this ALL?
These questions remain a challenge due to: 1) the absence of prospective studies on diagnostic/remission-matched samples, 2) the lack of models which faithfully reproduce the disease and 3) a surprising genomic stability of this ALL.
I hypothesize that a Multilayer-Omics to function approach in patient blasts and early human hematopoietic stem/progenitor cells (HSPC) is required to fully scrutinize the biology underlying this life-threatening leukemia. I will perform genome-wide studies on the mutational landscape, DNA and H3K79 methylation profiles, and transcriptome on a uniquely available, large cohort of diagnostic/remission-matched samples. Omics data integration will provide unprecedented information about oncogenic drivers which must be analyzed in ground-breaking functional assays using patient blasts and early HSPCs carrying a CRISPR/Cas9-mediated locus/allele-specific t(4;11). Serial xenografts combined with exome-seq in paired diagnostic samples and xenografts will identify the LIC and determine whether variegated genetics may underlie clonal functional heterogeneity. This project will provide a precise understanding and a disease model for MA4+ ALL, offering a platform for new treatment strategies.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym InMec
Project Inside mechanisms sustaining cancer stem cells
Researcher (PI) Pier Giuseppe Pelicci
Host Institution (HI) ISTITUTO EUROPEO DI ONCOLOGIA SRL
Call Details Advanced Grant (AdG), LS4, ERC-2013-ADG
Summary The “Cancer Stem Cell (CSC) Hypothesis” postulates that the capacity to maintain tumour growth is owned by rare cancer cells, the CSCs, endowed with self-renewal properties. This hypothesis implies that CSCs must be eliminated to achieve cancer cure. Nevertheless, direct proof is still lacking, and recent findings challenge our concepts of CSCs, showing the limits of the CSC-defining assay (transplantation) and suggesting that CSC-identity might be context-dependent. We found two properties of CSCs self-renewal that are indispensable for the maintenance of an expanding CSC-pool and tumour growth: increased frequency of symmetric divisions, due to inactivation of the p53 tumour suppressor, and increased replicative potential, due to up-regulation of the cell-cycle inhibitor p21. We will now investigate: i) How loss of p53 in tumours leads to expansion of the CSC pool, by testing the hypothesis that p53-loss activates the Myc oncogene which induces CSC-reprogramming of differentiated cancer cells. ii) Whether p53-independent pathways are also implicated, by in vivo shRNA screens of primary tumours or normal progenitors to identify pathways involved, respectively, in CSC self-renewal or inhibition of SC-reprogramming. iii) How p21-induced cell-cycle arrest protects CSCs from self-renewal exhaustion, by investigating regulation of cell-cycle recruitment of quiescent CSCs. iv) Whether activation of p21 in CSCs induces a mutator phenotype, due to its ability to activate DNA repair, by investigating mechanisms of DNA-damage, mutation rates, and relevance of CSC mutations for development of chemoresistance. We will test self-renewal functions in a transplantation-independent assay, based on tumour re-growth in vivo after cytotoxic treatments and “clonal tracking” of re-growing tumours (using barcoded lentiviral libraries). Our long-term goal is the identification of CSC-specific targets that could be used to create the basis for CSC-specific pharmacological intervention.
Summary
The “Cancer Stem Cell (CSC) Hypothesis” postulates that the capacity to maintain tumour growth is owned by rare cancer cells, the CSCs, endowed with self-renewal properties. This hypothesis implies that CSCs must be eliminated to achieve cancer cure. Nevertheless, direct proof is still lacking, and recent findings challenge our concepts of CSCs, showing the limits of the CSC-defining assay (transplantation) and suggesting that CSC-identity might be context-dependent. We found two properties of CSCs self-renewal that are indispensable for the maintenance of an expanding CSC-pool and tumour growth: increased frequency of symmetric divisions, due to inactivation of the p53 tumour suppressor, and increased replicative potential, due to up-regulation of the cell-cycle inhibitor p21. We will now investigate: i) How loss of p53 in tumours leads to expansion of the CSC pool, by testing the hypothesis that p53-loss activates the Myc oncogene which induces CSC-reprogramming of differentiated cancer cells. ii) Whether p53-independent pathways are also implicated, by in vivo shRNA screens of primary tumours or normal progenitors to identify pathways involved, respectively, in CSC self-renewal or inhibition of SC-reprogramming. iii) How p21-induced cell-cycle arrest protects CSCs from self-renewal exhaustion, by investigating regulation of cell-cycle recruitment of quiescent CSCs. iv) Whether activation of p21 in CSCs induces a mutator phenotype, due to its ability to activate DNA repair, by investigating mechanisms of DNA-damage, mutation rates, and relevance of CSC mutations for development of chemoresistance. We will test self-renewal functions in a transplantation-independent assay, based on tumour re-growth in vivo after cytotoxic treatments and “clonal tracking” of re-growing tumours (using barcoded lentiviral libraries). Our long-term goal is the identification of CSC-specific targets that could be used to create the basis for CSC-specific pharmacological intervention.
Max ERC Funding
2 500 000 €
Duration
Start date: 2014-07-01, End date: 2019-06-30
Project acronym INSIGHT
Project An Integrated Network of Glucose Sensing Cells in Glucose Homeostasis
Researcher (PI) Bernard Marie Thorens
Host Institution (HI) UNIVERSITE DE LAUSANNE
Call Details Advanced Grant (AdG), LS4, ERC-2010-AdG_20100317
Summary Glucose sensing cells constantly monitor glucose absorption from food and variations in blood glycemic levels. They control the secretion of GLP-1, insulin and glucagon, and the activity of the autonomic nervous system. These hormonal and nervous signals coordinate glucose utilization by liver, fat and muscle, and endogenous glucose production as well as feeding and energy expenditure. Type 2 diabetes, a disease that afflicts an increasing proportion of the world population, is characterized by insufficient insulin production by pancreatic beta-cells, abnormal secretion of GLP-1 and glucagon, and is often associated with imbalance between feeding and energy expenditure. Type 2 diabetes can thus be considered a disease of glucose sensing. Here, I propose a research program using cell biological, genetic, genomic and physiology techniques to investigate three aspects of this integrated glucose sensing network:
1. The identification of novel molecular pathways activated by GLP-1 and that control adult beta-cell proliferation, glucose competence and apoptosis in order to maintain sufficient insulin secretion capacity.
2. The identification and molecular characterization of brain glucose sensors, which share functional similarities with pancreatic beta-cells, and which control glucose homeostasis and pancreatic islet mass and function.
3. The discovery by unbiased genetic-genomic analysis of loci, genes, and gene networks involved in central hypoglycemia detection and the secretion of glucagon, a process whose deregulation is a major limitation in insulin treatment of both type 1 and type 2 diabetes.
Together these investigations will bring new knowledge on the integrated control of glucose homeostasis that may lead to novel strategies to control diabetes.
Summary
Glucose sensing cells constantly monitor glucose absorption from food and variations in blood glycemic levels. They control the secretion of GLP-1, insulin and glucagon, and the activity of the autonomic nervous system. These hormonal and nervous signals coordinate glucose utilization by liver, fat and muscle, and endogenous glucose production as well as feeding and energy expenditure. Type 2 diabetes, a disease that afflicts an increasing proportion of the world population, is characterized by insufficient insulin production by pancreatic beta-cells, abnormal secretion of GLP-1 and glucagon, and is often associated with imbalance between feeding and energy expenditure. Type 2 diabetes can thus be considered a disease of glucose sensing. Here, I propose a research program using cell biological, genetic, genomic and physiology techniques to investigate three aspects of this integrated glucose sensing network:
1. The identification of novel molecular pathways activated by GLP-1 and that control adult beta-cell proliferation, glucose competence and apoptosis in order to maintain sufficient insulin secretion capacity.
2. The identification and molecular characterization of brain glucose sensors, which share functional similarities with pancreatic beta-cells, and which control glucose homeostasis and pancreatic islet mass and function.
3. The discovery by unbiased genetic-genomic analysis of loci, genes, and gene networks involved in central hypoglycemia detection and the secretion of glucagon, a process whose deregulation is a major limitation in insulin treatment of both type 1 and type 2 diabetes.
Together these investigations will bring new knowledge on the integrated control of glucose homeostasis that may lead to novel strategies to control diabetes.
Max ERC Funding
2 499 421 €
Duration
Start date: 2011-08-01, End date: 2016-07-31
Project acronym INSTINCTIVE DRIVES
Project Orchestration of instinctive drives
Researcher (PI) Denis Burdakov
Host Institution (HI) MEDICAL RESEARCH COUNCIL
Call Details Starting Grant (StG), LS4, ERC-2007-StG
Summary A major goal of 21st century science is to protect human health from the growing mismatch between ancient behavioural instincts and modern socio-economic reality. This is especially vital for basic instinctive drives such as appetite, which lead to overeating when food is readily available. The resulting obesity is responsible for 100,000s of premature deaths per year in Europe and North America, and this number is rapidly rising. Sleep is another powerful instinct which substantially contributes to premature human death, for example from car accidents caused by tiredness. Thus “self-destructive” behaviours caused by inappropriate activation of feeding and sleep drives take a devastating social and economic toll in developed countries, and there is a huge unmet need for effective therapies in this area. To design these therapies, we need to understand the brain mechanisms of instinctive drives. However, brain circuits regulating appetite and sleep have only been delineated in the past few years, and their principles of operation are poorly understood at present. The broad aim of my newly-established laboratory is to fill this gap in knowledge. To understand neural signals controlling instinctive drives, and their relationship to well-being and disease, the following questions must be answered: 1) how do neurons that control appetite and sleep generate their electrical and chemical signals? ) how do these neurons interact with each other? 3) how are these neurons altered in disorders of energy balance and sleep? Our objective for the next five years is to address these key unknowns by focusing on neurons known to be unequivocally important for normal sleep and appetite, the orexin and MCH neurons of the lateral hypothalamus.
Summary
A major goal of 21st century science is to protect human health from the growing mismatch between ancient behavioural instincts and modern socio-economic reality. This is especially vital for basic instinctive drives such as appetite, which lead to overeating when food is readily available. The resulting obesity is responsible for 100,000s of premature deaths per year in Europe and North America, and this number is rapidly rising. Sleep is another powerful instinct which substantially contributes to premature human death, for example from car accidents caused by tiredness. Thus “self-destructive” behaviours caused by inappropriate activation of feeding and sleep drives take a devastating social and economic toll in developed countries, and there is a huge unmet need for effective therapies in this area. To design these therapies, we need to understand the brain mechanisms of instinctive drives. However, brain circuits regulating appetite and sleep have only been delineated in the past few years, and their principles of operation are poorly understood at present. The broad aim of my newly-established laboratory is to fill this gap in knowledge. To understand neural signals controlling instinctive drives, and their relationship to well-being and disease, the following questions must be answered: 1) how do neurons that control appetite and sleep generate their electrical and chemical signals? ) how do these neurons interact with each other? 3) how are these neurons altered in disorders of energy balance and sleep? Our objective for the next five years is to address these key unknowns by focusing on neurons known to be unequivocally important for normal sleep and appetite, the orexin and MCH neurons of the lateral hypothalamus.
Max ERC Funding
1 299 999 €
Duration
Start date: 2008-10-01, End date: 2013-09-30
Project acronym INTEGRATE
Project Central integration of metabolic and hedonic cues in metabolic health
Researcher (PI) Bernard Marie Thorens
Host Institution (HI) UNIVERSITE DE LAUSANNE
Call Details Advanced Grant (AdG), LS4, ERC-2015-AdG
Summary During evolution the brain has selected glucose as a main source of metabolic energy. This has imposed homeostatic and behavioral constraints. First, the glycemic levels must be maintained at a minimum of ~5 mM to ensure constant energy supply to the brain. Second, a high reward value has to be attributed to glucose-containing foods to increase the motivation to obtain them. These homeostatic and hedonic regulations depend on glucose sensing cells and neuronal circuits in the central nervous system. These cells and circuits regulate the activity of the sympathetic and parasympathetic nerves, which control the function of peripheral organs (liver, fat, muscles) and the secretion of glucagon and insulin by pancreatic islet cells. They also attribute a reward value to glucose-containing foods to control food-seeking behavior, a process that involves the mesolimbic dopaminergic system. Here, we will focus on three interrelated aims:
1. Identify the physiological role of glucose sensing neurons of the ventromedial hypothalamic nucleus (VMN, a key feeding and glucoregulatory center) in glucose homeostasis and food preference; identify their cellular diversity and their molecular make-up; and characterize their deregulations in metabolic diseases.
2. Characterize the molecular physiology of glucose sensing neurons of the paraventricular thalamus, which modulate the activity of the mesolimbic dopaminergic system to control motivated sucrose-seeking behavior; determine their control by other interoceptive signals, including from glucose sensing cells of the VMN.
3. Establish new molecular approaches to characterize, at the molecular and functional levels, the impact of early postnatal nutrition on the development and function of central glucose sensing cells in the control of adult animal physiology.
These studies will open-up new perspectives in the understanding of homeostatic and hedonic regulatory pathways, which preserve metabolic health over a lifetime.
Summary
During evolution the brain has selected glucose as a main source of metabolic energy. This has imposed homeostatic and behavioral constraints. First, the glycemic levels must be maintained at a minimum of ~5 mM to ensure constant energy supply to the brain. Second, a high reward value has to be attributed to glucose-containing foods to increase the motivation to obtain them. These homeostatic and hedonic regulations depend on glucose sensing cells and neuronal circuits in the central nervous system. These cells and circuits regulate the activity of the sympathetic and parasympathetic nerves, which control the function of peripheral organs (liver, fat, muscles) and the secretion of glucagon and insulin by pancreatic islet cells. They also attribute a reward value to glucose-containing foods to control food-seeking behavior, a process that involves the mesolimbic dopaminergic system. Here, we will focus on three interrelated aims:
1. Identify the physiological role of glucose sensing neurons of the ventromedial hypothalamic nucleus (VMN, a key feeding and glucoregulatory center) in glucose homeostasis and food preference; identify their cellular diversity and their molecular make-up; and characterize their deregulations in metabolic diseases.
2. Characterize the molecular physiology of glucose sensing neurons of the paraventricular thalamus, which modulate the activity of the mesolimbic dopaminergic system to control motivated sucrose-seeking behavior; determine their control by other interoceptive signals, including from glucose sensing cells of the VMN.
3. Establish new molecular approaches to characterize, at the molecular and functional levels, the impact of early postnatal nutrition on the development and function of central glucose sensing cells in the control of adult animal physiology.
These studies will open-up new perspectives in the understanding of homeostatic and hedonic regulatory pathways, which preserve metabolic health over a lifetime.
Max ERC Funding
2 499 714 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym INTERCLOUDS
Project Using the Magellanic Clouds to Understand the Interaction of Galaxies
Researcher (PI) Maria-Rosa Cioni
Host Institution (HI) LEIBNIZ-INSTITUT FUR ASTROPHYSIK POTSDAM (AIP)
Call Details Consolidator Grant (CoG), PE9, ERC-2015-CoG
Summary The Magellanic Clouds are the nearest gas-rich dwarf satellites of the Milky Way and illustrate a typical example of an early phase of a minor merger event, the collision of galaxies that differ in mass by at least a factor of ten. In spite of their important role in supplementing material to the Milky Way halo and the numerous investigations made in the last decade, there remain several uncertainties. Their origin is still a matter of debate, their satellite status is unclear, their mass is uncertain, their gravitational centres are undefined, their structure depends strongly on stellar populations and is severely shaped by interactions, their orbital history is only vaguely associated to star forming events, and their chemical history rests upon limited data. This proposal aims to remedy this lack of knowledge by providing a comprehensive analysis of the stellar content of the Magellanic Clouds and dissect the substructures that are related to their accretion history and the interaction with the Milky Way. Their internal kinematics and orbital history, establishing their bound/unbound status, will be resolved thanks to the analysis of state-of-the art proper motions from the VMC survey and the Gaia mission, and the development of sophisticated theoretical models. Multi-wavelength photometric observations from ongoing large-scale projects will be analysed together to characterise the stellar population of the Magellanic Clouds as has never been previously attempted, including the effects of separate structural components. New large-scale spectroscopic survey projects in preparation will resolve metallicity dependencies and complete the full six-phase space information (distance, position, and motion). This proposal will have a tremendous impact on our understanding of the consequences of minor mergers, and will offer a firm perspective of the Magellanic Clouds.
Summary
The Magellanic Clouds are the nearest gas-rich dwarf satellites of the Milky Way and illustrate a typical example of an early phase of a minor merger event, the collision of galaxies that differ in mass by at least a factor of ten. In spite of their important role in supplementing material to the Milky Way halo and the numerous investigations made in the last decade, there remain several uncertainties. Their origin is still a matter of debate, their satellite status is unclear, their mass is uncertain, their gravitational centres are undefined, their structure depends strongly on stellar populations and is severely shaped by interactions, their orbital history is only vaguely associated to star forming events, and their chemical history rests upon limited data. This proposal aims to remedy this lack of knowledge by providing a comprehensive analysis of the stellar content of the Magellanic Clouds and dissect the substructures that are related to their accretion history and the interaction with the Milky Way. Their internal kinematics and orbital history, establishing their bound/unbound status, will be resolved thanks to the analysis of state-of-the art proper motions from the VMC survey and the Gaia mission, and the development of sophisticated theoretical models. Multi-wavelength photometric observations from ongoing large-scale projects will be analysed together to characterise the stellar population of the Magellanic Clouds as has never been previously attempted, including the effects of separate structural components. New large-scale spectroscopic survey projects in preparation will resolve metallicity dependencies and complete the full six-phase space information (distance, position, and motion). This proposal will have a tremendous impact on our understanding of the consequences of minor mergers, and will offer a firm perspective of the Magellanic Clouds.
Max ERC Funding
1 985 017 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym INTERSTELLAR
Project The Interstellar Medium of High Redshift Galaxies
Researcher (PI) Andrea FERRARA
Host Institution (HI) SCUOLA NORMALE SUPERIORE
Call Details Advanced Grant (AdG), PE9, ERC-2016-ADG
Summary When and how did the first galaxies form across cosmic history? Were they different from present-day ones? This is only a small subset of key cosmological questions that the combination of deep galaxy observations, theoretical modeling, and powerful simulations envisaged here will allow us to answer for the first time.
Deep galaxy surveys have provided a first valuable characterization of early galaxies in the Epoch of Reionization (redshift z > 6), mostly in terms of their stellar content. However, almost nothing is known about their internal structure and Interstellar Medium (ISM). This is in striking contrast with galaxies at z < 2, for which ISM observations have enabled a much more complete physical description. Hence, a substantial progress in the study of early galaxies must be based on techniques able to probe their ISM. Conversely, ISM studies will help completing the “stellar” picture.
Interstellar will bridge this gap. Its main aim is to understand the internal structure and interstellar medium of galaxies in the Epoch of Reionization by performing theoretical modeling and high fidelity simulations. By post-processing the simulations and calibrating them with local analogs, we will produce mock images/spectra used to (i) interpret available high-redshift observations, and (ii) plan breakthrough experiments with ALMA, JWST and E-ELT.
The advent of ALMA, JWST, E-ELT and advances in computational cosmology make the study of high-z ISM one of the most promising areas of development in cosmology.
The aim will be achieved through 5 objectives distributed among 3 Work Packages (WPs). WP1 is concerned with theoretical work, a preparatory phase for the cosmological simulations performed in WP2. WP2 represents the production phase of the project and will deliver cutting-edge zoom simulations of a sample of high-z galaxies and their ISM. Finally, WP3 is concerned with the exploitation of the numerical results and their integration with observations.
Summary
When and how did the first galaxies form across cosmic history? Were they different from present-day ones? This is only a small subset of key cosmological questions that the combination of deep galaxy observations, theoretical modeling, and powerful simulations envisaged here will allow us to answer for the first time.
Deep galaxy surveys have provided a first valuable characterization of early galaxies in the Epoch of Reionization (redshift z > 6), mostly in terms of their stellar content. However, almost nothing is known about their internal structure and Interstellar Medium (ISM). This is in striking contrast with galaxies at z < 2, for which ISM observations have enabled a much more complete physical description. Hence, a substantial progress in the study of early galaxies must be based on techniques able to probe their ISM. Conversely, ISM studies will help completing the “stellar” picture.
Interstellar will bridge this gap. Its main aim is to understand the internal structure and interstellar medium of galaxies in the Epoch of Reionization by performing theoretical modeling and high fidelity simulations. By post-processing the simulations and calibrating them with local analogs, we will produce mock images/spectra used to (i) interpret available high-redshift observations, and (ii) plan breakthrough experiments with ALMA, JWST and E-ELT.
The advent of ALMA, JWST, E-ELT and advances in computational cosmology make the study of high-z ISM one of the most promising areas of development in cosmology.
The aim will be achieved through 5 objectives distributed among 3 Work Packages (WPs). WP1 is concerned with theoretical work, a preparatory phase for the cosmological simulations performed in WP2. WP2 represents the production phase of the project and will deliver cutting-edge zoom simulations of a sample of high-z galaxies and their ISM. Finally, WP3 is concerned with the exploitation of the numerical results and their integration with observations.
Max ERC Funding
2 151 875 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym INTRAHETEROSEQ
Project Molecular characterization of the role of intra-tumor heterogeneity in cancer progression and metastasis
Researcher (PI) Ignacio VARELA EGOCHEAGA
Host Institution (HI) UNIVERSIDAD DE CANTABRIA
Call Details Starting Grant (StG), LS4, ERC-2014-STG
Summary Cancer is caused by somatically acquired changes in the DNA. Some of these changes fall in “cancer genes”, conferring clonal selective advantage to the cells that carry the mutant alleles. Identifying these genes/pathways is of vital importance for a correct understanding of cancer biology as well as for the diagnosis and treatment of human malignancies. In this respect, the use of genetically modified mice has been extremely useful in the past for characterizing the molecular pathways involved in cancer progression. The remarkable progress made during the last two decades on the genetic modification of mouse genomes offers unique opportunities to investigate different aspects of tumor molecular behavior, impossible to study on human samples.
Recently, the advent of next-generation sequencing technologies has provided new strategies for the systematic genome-wide identification of somatic changes in cancer cell genomes. Using these technologies, we and others have characterized the high intra-tumor heterogeneity observed in some human tumors. Although the exact significance of this heterogeneity is uncertain, it seems to be responsible for key aspects in the management of cancer patients such as metastasis predisposition and tissue specificity or treatment resistance.
Taking advantage of next-generation sequencing, we propose to finely characterize the intra-tumor heterogeneity evolution during the progression of tumors induced in a mouse model of pancreatic cancer, as well as, for the first time, to purify the different cell populations these primary tumors are composed of. A complete genomic and transcriptomic characterization of these populations followed by posterior functional assays will help us to identify the genes/pathways involved in tumor progression as well as metastatic potential and its tissue specificity. This new knowledge could finally contribute to a better understanding of cancer and to the design of more efficient anti-tumor therapies
Summary
Cancer is caused by somatically acquired changes in the DNA. Some of these changes fall in “cancer genes”, conferring clonal selective advantage to the cells that carry the mutant alleles. Identifying these genes/pathways is of vital importance for a correct understanding of cancer biology as well as for the diagnosis and treatment of human malignancies. In this respect, the use of genetically modified mice has been extremely useful in the past for characterizing the molecular pathways involved in cancer progression. The remarkable progress made during the last two decades on the genetic modification of mouse genomes offers unique opportunities to investigate different aspects of tumor molecular behavior, impossible to study on human samples.
Recently, the advent of next-generation sequencing technologies has provided new strategies for the systematic genome-wide identification of somatic changes in cancer cell genomes. Using these technologies, we and others have characterized the high intra-tumor heterogeneity observed in some human tumors. Although the exact significance of this heterogeneity is uncertain, it seems to be responsible for key aspects in the management of cancer patients such as metastasis predisposition and tissue specificity or treatment resistance.
Taking advantage of next-generation sequencing, we propose to finely characterize the intra-tumor heterogeneity evolution during the progression of tumors induced in a mouse model of pancreatic cancer, as well as, for the first time, to purify the different cell populations these primary tumors are composed of. A complete genomic and transcriptomic characterization of these populations followed by posterior functional assays will help us to identify the genes/pathways involved in tumor progression as well as metastatic potential and its tissue specificity. This new knowledge could finally contribute to a better understanding of cancer and to the design of more efficient anti-tumor therapies
Max ERC Funding
1 498 850 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym IntratumoralNiche
Project Defining heterocellular signalling within the intratumoral stem cell niche of colorectal cancer
Researcher (PI) Hugo SNIPPERT
Host Institution (HI) UNIVERSITAIR MEDISCH CENTRUM UTRECHT
Call Details Starting Grant (StG), LS4, ERC-2018-STG
Summary Purpose: Cells in a tumor are highly heterogeneous. The role and consequence of having multiple cell types within a cancer is mostly centered towards the function of cancer stem cells (CSCs) since they are the driving forces of tumor growth. However, the exact signaling cues that support CSC function remain to be understood. For instance, what are the roles of immediate descendant tumor cells in relation to CSC support? Do colorectal tumors make their own niche?
Preliminary data: To study communication between different cell types (heterocellular signaling) in human colorectal cancers (CRCs), my lab developed movieSTAR technology to mark CSCs in patient-derived CRC organoids (PDOs) for high-resolution live imaging of their dynamics and behavior. Although niche factor dependency decreases along the adenoma-carcinoma transition, we identified a strong interdependency between CSCs and other tumor cells in colorectal PDOs of malignant nature.
Hypothesis: We hypothesize a continuous existence of an intratumoral stem cell niche that remains essential for tumor growth and metastasis formation. Which types of heterocellular signaling support CSC function, especially at malignant stages, is unknown.
Approach: This project aims to define heterocellular signaling between CSCs and intratumoral niche cells. Therefore, I) we will combine our expertise in human organoid technology for in-depth characterization of the nature of heterocellular communication within the intratumoral niche, II) high-resolution live imaging of PDOs to interrogate heterogeneity of signaling activities at cellular resolution and in real-time, as well as III) in vivo mouse models for validation and further studies of essential intratumoral signaling pathways.
Innovation: Our integrative use of novel approaches will provide comprehensive insight into intratumoral niche function during tumorigenesis, establishing novel technologies for future cancer research and new concepts to improve cancer therapy.
Summary
Purpose: Cells in a tumor are highly heterogeneous. The role and consequence of having multiple cell types within a cancer is mostly centered towards the function of cancer stem cells (CSCs) since they are the driving forces of tumor growth. However, the exact signaling cues that support CSC function remain to be understood. For instance, what are the roles of immediate descendant tumor cells in relation to CSC support? Do colorectal tumors make their own niche?
Preliminary data: To study communication between different cell types (heterocellular signaling) in human colorectal cancers (CRCs), my lab developed movieSTAR technology to mark CSCs in patient-derived CRC organoids (PDOs) for high-resolution live imaging of their dynamics and behavior. Although niche factor dependency decreases along the adenoma-carcinoma transition, we identified a strong interdependency between CSCs and other tumor cells in colorectal PDOs of malignant nature.
Hypothesis: We hypothesize a continuous existence of an intratumoral stem cell niche that remains essential for tumor growth and metastasis formation. Which types of heterocellular signaling support CSC function, especially at malignant stages, is unknown.
Approach: This project aims to define heterocellular signaling between CSCs and intratumoral niche cells. Therefore, I) we will combine our expertise in human organoid technology for in-depth characterization of the nature of heterocellular communication within the intratumoral niche, II) high-resolution live imaging of PDOs to interrogate heterogeneity of signaling activities at cellular resolution and in real-time, as well as III) in vivo mouse models for validation and further studies of essential intratumoral signaling pathways.
Innovation: Our integrative use of novel approaches will provide comprehensive insight into intratumoral niche function during tumorigenesis, establishing novel technologies for future cancer research and new concepts to improve cancer therapy.
Max ERC Funding
1 500 000 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym INTUMORX
Project Elucidation of intratumoral heterogeneity in Kras-driven cancers
Researcher (PI) Tuomas TAMMELA
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Starting Grant (StG), LS4, ERC-2016-STG
Summary The considerable variability within tissue microenvironments as well as the multiclonality of cancers leads to intratumoral heterogeneity. This increases the probablility of cellular states that promote resistance to therapy and eventually lead to reconstitution of the tumor by treatment-resistant cancer cells, which in some cases have properties of normal tissue stem cells. Wnt signals are important in the maintenance of stem cells in various epithelial tissues, including in lung development and regeneration. We hypothesized that Wnt signals contribute to tumor heterogeneity in genetically engineered KrasG12D; Tp53Δ/Δ (”KP”) mouse lung adenocarcinomas (LUAD). We observed that a subpopulation of LUAD cells exhibited high Wnt reporter activity and had increased tumor forming ability, which could be suppressed by silencing of Wnt signaling pathway components or by small molecule Wnt inhibitors in vitro and in vivo. KP LUAD cells show hierarchical features with two distinct populations, one with increased Wnt reporter activity and another forming a niche that provides the Wnt signal. Lineage-tracing experiments in the autochthonous KP tumors demonstrated that Wnt responder cells have increased tumor propagation ability in vivo. Strikingly, selective ablation of the Wnt responder cells resulted in tumor stasis. CRISPR-based targeting or small molecules targeting Wnt signaling reduced tumor growth and prolonged survival in the autochthonous KP mouse lung cancer model. These results indicate that maintenance of heterogeneity within tumors may be advantageous for the tumor cell population collectively. We propose to elucidate the molecular and cellullar mechanisms that control stem-like and niche cell phenotypes using a combination of novel lentiviral vectors and genetically modified mice in the context of the KP LUAD model. These efforts may lead to novel therapeutic concepts in human lung cancer.
Summary
The considerable variability within tissue microenvironments as well as the multiclonality of cancers leads to intratumoral heterogeneity. This increases the probablility of cellular states that promote resistance to therapy and eventually lead to reconstitution of the tumor by treatment-resistant cancer cells, which in some cases have properties of normal tissue stem cells. Wnt signals are important in the maintenance of stem cells in various epithelial tissues, including in lung development and regeneration. We hypothesized that Wnt signals contribute to tumor heterogeneity in genetically engineered KrasG12D; Tp53Δ/Δ (”KP”) mouse lung adenocarcinomas (LUAD). We observed that a subpopulation of LUAD cells exhibited high Wnt reporter activity and had increased tumor forming ability, which could be suppressed by silencing of Wnt signaling pathway components or by small molecule Wnt inhibitors in vitro and in vivo. KP LUAD cells show hierarchical features with two distinct populations, one with increased Wnt reporter activity and another forming a niche that provides the Wnt signal. Lineage-tracing experiments in the autochthonous KP tumors demonstrated that Wnt responder cells have increased tumor propagation ability in vivo. Strikingly, selective ablation of the Wnt responder cells resulted in tumor stasis. CRISPR-based targeting or small molecules targeting Wnt signaling reduced tumor growth and prolonged survival in the autochthonous KP mouse lung cancer model. These results indicate that maintenance of heterogeneity within tumors may be advantageous for the tumor cell population collectively. We propose to elucidate the molecular and cellullar mechanisms that control stem-like and niche cell phenotypes using a combination of novel lentiviral vectors and genetically modified mice in the context of the KP LUAD model. These efforts may lead to novel therapeutic concepts in human lung cancer.
Max ERC Funding
1 972 905 €
Duration
Start date: 2017-07-01, End date: 2022-06-30
Project acronym INVADERS
Project Mucus-Penetrating Microbiota: Characterization, Mechanism and Therapeutic in Metabolic Disease
Researcher (PI) Benoit CHASSAING
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS4, ERC-2018-STG
Summary Mucus-Penetrating Microbiota: Characterization, Mechanism and Therapeutic in Metabolic Disease
Humanity is facing an epidemic of inter-related metabolic disorders, including obesity, insulin resistance, hyperglycemia, hyperlipidemia, and hepatic steatosis, that altogether have major impact on the promotion of cardiovascular diseases. The increasing incidence of these complex metabolic disorders and their highly morbid, chronic and costly downstream diseases threatens to overwhelm the world’s health care systems and economies, making it a top public health priority in dire need of investigation.
The intestinal tract is inhabited by a large and diverse community of bacteria, collectively referred to as the intestinal microbiota. When stably maintained at an appropriately safe distance from the epithelial cell monolayer, the microbiota provides important benefits to its host. However, disturbance of the microbiota-host relationship, promoted by genetic or non-genetic factors, can alter intestinal homeostasis and drive chronic low-grade intestinal inflammation, ultimately leading to metabolic abnormalities. We previously reported that a ubiquitous class of food additives, emulsifiers, detrimentally impact the microbiota resulting in its encroachment into the mucus layer that associated with low-grade inflammation and development of metabolic disorders.
The central goal of this proposal is to investigate the hypothesis that bacteria that penetrate the inner part of the mucus layer, referred as invaders, promote development of metabolic alterations.
We herein propose to identify mucus-invaders, in preclinical models and clinical conditions, and investigate mechanisms by which they promote inflammatory and metabolic abnormalities. Furthermore, we propose to define original approaches to modulate the intestinal microbiota in order to counteract microbiota encroachment and protect against associated metabolic abnormalities.
Summary
Mucus-Penetrating Microbiota: Characterization, Mechanism and Therapeutic in Metabolic Disease
Humanity is facing an epidemic of inter-related metabolic disorders, including obesity, insulin resistance, hyperglycemia, hyperlipidemia, and hepatic steatosis, that altogether have major impact on the promotion of cardiovascular diseases. The increasing incidence of these complex metabolic disorders and their highly morbid, chronic and costly downstream diseases threatens to overwhelm the world’s health care systems and economies, making it a top public health priority in dire need of investigation.
The intestinal tract is inhabited by a large and diverse community of bacteria, collectively referred to as the intestinal microbiota. When stably maintained at an appropriately safe distance from the epithelial cell monolayer, the microbiota provides important benefits to its host. However, disturbance of the microbiota-host relationship, promoted by genetic or non-genetic factors, can alter intestinal homeostasis and drive chronic low-grade intestinal inflammation, ultimately leading to metabolic abnormalities. We previously reported that a ubiquitous class of food additives, emulsifiers, detrimentally impact the microbiota resulting in its encroachment into the mucus layer that associated with low-grade inflammation and development of metabolic disorders.
The central goal of this proposal is to investigate the hypothesis that bacteria that penetrate the inner part of the mucus layer, referred as invaders, promote development of metabolic alterations.
We herein propose to identify mucus-invaders, in preclinical models and clinical conditions, and investigate mechanisms by which they promote inflammatory and metabolic abnormalities. Furthermore, we propose to define original approaches to modulate the intestinal microbiota in order to counteract microbiota encroachment and protect against associated metabolic abnormalities.
Max ERC Funding
1 850 000 €
Duration
Start date: 2019-10-01, End date: 2024-09-30
Project acronym INVESTIGERFE
Project Investigating the regulation of iron homeostasis by erythroferrone and therapeutic applications
Researcher (PI) Léon Charles KAUTZ
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS4, ERC-2016-STG
Summary The existence of an “erythron-related regulator” that intensifies iron absorption and its release from stores to meet the requirements for red blood cells synthesis was proposed in the 1950s. Delineating this mechanism is of high biomedical importance as the pathway could be targeted to develop novel treatments for iron-restricted anemias that are very frequent but for which current therapies are ineffective (e.g. infections, inflammatory bowel disease, cancer, or chronic kidney disease) and for iron-loading anemias (e.g. thalassemias). We have recently identified the hormone erythroferrone (ERFE) and showed that it could be the long-sought erythroid regulator of iron homeostasis. ERFE suppresses the synthesis of the iron-regulatory hormone hepcidin to facilitate the recovery from anemia but leads to secondary iron overload in β-thalassemia. The potential of ERFE in the treatment of iron disorders is tremendous but understanding its mechanism of action is a prerequisite to envision ERFE-based therapies. The identification of ERFE has opened new research areas and our project will be organized around four axes.
1) Develop an assay to measure ERFE levels in human pathologies. Its contribution is not known and needs to be confirmed.
2) Identify the receptor for ERFE, the signaling pathways triggered by ERFE, and molecules with agonist/antagonist effects, a prerequisite in the development of new therapies.
3) Search for potential other erythroid regulators. We will take advantage of the Erfe-/- mice to determine whether hepcidin could be suppressed by an ERFE-independent mechanism.
4) Study the potential of ERFE manipulation in therapy in the mouse. We will first establish a proof of principle in a mouse model of anemia (B. abortus). The benefits of ERFE antagonization will be addressed in thalassemic mice. We will also examine the role of ERFE in murine models of chronic anemia: chronic kidney disease, inflammatory bowel disease, rheumatoid arthritis and infections.
Summary
The existence of an “erythron-related regulator” that intensifies iron absorption and its release from stores to meet the requirements for red blood cells synthesis was proposed in the 1950s. Delineating this mechanism is of high biomedical importance as the pathway could be targeted to develop novel treatments for iron-restricted anemias that are very frequent but for which current therapies are ineffective (e.g. infections, inflammatory bowel disease, cancer, or chronic kidney disease) and for iron-loading anemias (e.g. thalassemias). We have recently identified the hormone erythroferrone (ERFE) and showed that it could be the long-sought erythroid regulator of iron homeostasis. ERFE suppresses the synthesis of the iron-regulatory hormone hepcidin to facilitate the recovery from anemia but leads to secondary iron overload in β-thalassemia. The potential of ERFE in the treatment of iron disorders is tremendous but understanding its mechanism of action is a prerequisite to envision ERFE-based therapies. The identification of ERFE has opened new research areas and our project will be organized around four axes.
1) Develop an assay to measure ERFE levels in human pathologies. Its contribution is not known and needs to be confirmed.
2) Identify the receptor for ERFE, the signaling pathways triggered by ERFE, and molecules with agonist/antagonist effects, a prerequisite in the development of new therapies.
3) Search for potential other erythroid regulators. We will take advantage of the Erfe-/- mice to determine whether hepcidin could be suppressed by an ERFE-independent mechanism.
4) Study the potential of ERFE manipulation in therapy in the mouse. We will first establish a proof of principle in a mouse model of anemia (B. abortus). The benefits of ERFE antagonization will be addressed in thalassemic mice. We will also examine the role of ERFE in murine models of chronic anemia: chronic kidney disease, inflammatory bowel disease, rheumatoid arthritis and infections.
Max ERC Funding
1 499 235 €
Duration
Start date: 2017-05-01, End date: 2022-04-30
Project acronym iPROTECTION
Project Molecular mechanisms of induced protection against sepsis by DNA damage responses
Researcher (PI) Luis Filipe Ferreira Moita
Host Institution (HI) FUNDACAO CALOUSTE GULBENKIAN
Call Details Consolidator Grant (CoG), LS4, ERC-2014-CoG
Summary Severe sepsis remains a poorly understood systemic inflammatory condition with high mortality rates and limited therapeutic options outside of infection control and organ support measures. Based on our recent discovery that anthracycline drugs prevent organ failure without affecting the bacterial burden in a model of severe sepsis, we propose that strategies aimed at target organ protection have extraordinary potential for the treatment of sepsis and possibly for other inflammation-driven conditions. However, the mechanisms of organ protection and disease tolerance are either unknown or poorly characterized.
The central goal of the current proposal is to identify and characterize novel cytoprotective mechanisms, with a focus on DNA damage response dependent protection activated by anthracyclines as a window into stress-induced genetic programs conferring disease tolerance. To that end, we will carry out a combination of candidate and unbiased approaches for the in vivo identification of ATM-dependent and independent mechanisms of tissue protection. We will validate the leading candidates through adenovirus-mediated delivery of constructs for overexpression (gain-of-function) or shRNA for gene silencing (loss-of-function) to the lung, based on our recent finding that rescuing this organ is essential and perhaps sufficient in anthracycline-induced protection against severe sepsis. The candidates showing the most promise will be characterized using a combination of in vitro and in vivo genetic, biochemical, cell biological and physiological methods.
The results arising from the current proposal are likely not only to inspire the design of transformative therapies for sepsis but also to open a completely new field of opportunity to molecularly understand core surveillance mechanisms of basic cellular processes with a critical role in the homeostasis of organ function and whose activation can ultimately promote quality of life during aging and increase lifespan.
Summary
Severe sepsis remains a poorly understood systemic inflammatory condition with high mortality rates and limited therapeutic options outside of infection control and organ support measures. Based on our recent discovery that anthracycline drugs prevent organ failure without affecting the bacterial burden in a model of severe sepsis, we propose that strategies aimed at target organ protection have extraordinary potential for the treatment of sepsis and possibly for other inflammation-driven conditions. However, the mechanisms of organ protection and disease tolerance are either unknown or poorly characterized.
The central goal of the current proposal is to identify and characterize novel cytoprotective mechanisms, with a focus on DNA damage response dependent protection activated by anthracyclines as a window into stress-induced genetic programs conferring disease tolerance. To that end, we will carry out a combination of candidate and unbiased approaches for the in vivo identification of ATM-dependent and independent mechanisms of tissue protection. We will validate the leading candidates through adenovirus-mediated delivery of constructs for overexpression (gain-of-function) or shRNA for gene silencing (loss-of-function) to the lung, based on our recent finding that rescuing this organ is essential and perhaps sufficient in anthracycline-induced protection against severe sepsis. The candidates showing the most promise will be characterized using a combination of in vitro and in vivo genetic, biochemical, cell biological and physiological methods.
The results arising from the current proposal are likely not only to inspire the design of transformative therapies for sepsis but also to open a completely new field of opportunity to molecularly understand core surveillance mechanisms of basic cellular processes with a critical role in the homeostasis of organ function and whose activation can ultimately promote quality of life during aging and increase lifespan.
Max ERC Funding
1 985 375 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym iPS-ChOp-AF
Project Combining induced pluripotent stem cells, tissue engineering, optogenetic and chemogenetic concepts for the study and treatment of atrial fibrillation
Researcher (PI) Lior GEPSTEIN
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Consolidator Grant (CoG), LS4, ERC-2017-COG
Summary "Cardiac arrhythmias are responsible for significant morbidity and mortality. However, the study and treatment of these rhythm disorders have been hampered by the lack of relevant human cardiac tissue models, specifically those reflecting patient/disease-specific abnormalities, by paucity of methods for long-term electrophysiological analysis of the tissue, and by the inability to perform targeted, high-resolution, reversible, and functional perturbations of the system.
To address these challenges, we propose to combine human induced pluripotent stem cells (hiPSC) and genome-editing (CRISPR) technologies, developmental biology-inspired differentiating systems that yield chamber-specific heart cells, novel tissue engineering strategies, and emerging concepts from the fields of optogenetics and chemogenetics. The resulting experimental models should represent a paradigm shift in the way we study and treat cardiac arrhythmias. To demonstrate the unique potential of this approach, we plan to focus on atrial fibrillation (AF), the most common arrhythmia.
Our specific aims are to:
1. Develop patient/disease-specific hiPSC models of genetic AF and to establish hiPSC differentiation protocols to yield purified atrial cells
2. Utilize the hiPSC-atrial cells and advanced tissue-engineering strategies (hydrogels, 3D printing, decellularization) to establish 2D cell-sheet and 3D tissue models of acquired and inherited AF, in which functional re-entry (""rotors"") can be studied
3. Utilize tools from optogenetics (light-sensitive ion channels and pumps) or chemogenetics (ligand-specific engineered receptors) for targeted manipulation of the system, to gain insights into AF pathogenesis and to develop novel therapies
4. Evaluate the developed optogenetic and chemogenetic treatments in animal models of AF
The results of this project should provide novel mechanistic insights into AF (and other arrhythmias) and open the road for the development of novel therapeutic paradigms."
Summary
"Cardiac arrhythmias are responsible for significant morbidity and mortality. However, the study and treatment of these rhythm disorders have been hampered by the lack of relevant human cardiac tissue models, specifically those reflecting patient/disease-specific abnormalities, by paucity of methods for long-term electrophysiological analysis of the tissue, and by the inability to perform targeted, high-resolution, reversible, and functional perturbations of the system.
To address these challenges, we propose to combine human induced pluripotent stem cells (hiPSC) and genome-editing (CRISPR) technologies, developmental biology-inspired differentiating systems that yield chamber-specific heart cells, novel tissue engineering strategies, and emerging concepts from the fields of optogenetics and chemogenetics. The resulting experimental models should represent a paradigm shift in the way we study and treat cardiac arrhythmias. To demonstrate the unique potential of this approach, we plan to focus on atrial fibrillation (AF), the most common arrhythmia.
Our specific aims are to:
1. Develop patient/disease-specific hiPSC models of genetic AF and to establish hiPSC differentiation protocols to yield purified atrial cells
2. Utilize the hiPSC-atrial cells and advanced tissue-engineering strategies (hydrogels, 3D printing, decellularization) to establish 2D cell-sheet and 3D tissue models of acquired and inherited AF, in which functional re-entry (""rotors"") can be studied
3. Utilize tools from optogenetics (light-sensitive ion channels and pumps) or chemogenetics (ligand-specific engineered receptors) for targeted manipulation of the system, to gain insights into AF pathogenesis and to develop novel therapies
4. Evaluate the developed optogenetic and chemogenetic treatments in animal models of AF
The results of this project should provide novel mechanistic insights into AF (and other arrhythmias) and open the road for the development of novel therapeutic paradigms."
Max ERC Funding
1 988 750 €
Duration
Start date: 2018-03-01, End date: 2023-02-28
Project acronym ISCATAXIA
Project Unraveling the molecular mechanisms leading to cellular dysfunction in diseases linked to defects in mitochondrial iron-sulfur cluster metabolism
Researcher (PI) Hélène Monique Sadoulet Puccio
Host Institution (HI) CENTRE EUROPEEN DE RECHERCHE EN BIOLOGIE ET MEDECINE
Call Details Starting Grant (StG), LS4, ERC-2007-StG
Summary The project aims at unraveling the molecular pathophysiology of recessive ataxias, a heterogeneous set of severely disabling neurodegenerative disorders due to loss of function of proteins involved either in mitochondrial/metabolic pathways or DNA repair. Friedreich ataxia, the most common form, is due to partial loss of function of frataxin, a mitochondrial protein involved in iron-sulfur cluster (ISC) biogenesis. Furthermore, the rare X-linked sideroblastic anemia with cerebellar ataxia is caused by mutation in ABCb7, an ATP-binding cassette transporter of the mitochondrial inner membrane necessary for cytosolic ISC export. ISC are versatile co-factors of proteins involved in electron transport, enzyme catalysis and regulation of gene expression. The synthesis and insertion of ISC into apoproteins involve complex machineries that are still poorly understood in the mammalian cell. The objectives of this proposal are: 1) to elucidate ISC biogenesis and metabolism in the mammalian cell, with an emphasis on the role of frataxin and ABCb7; 2) to better understand the molecular pathways that are involved in neuronal dysfunction due to defects in mitochondrial ISC metabolism. These objectives will be accomplished by a multidisciplinary approach combining molecular and biochemical approaches to study the ISC assembly machineries, bioinformatic and proteomic studies to identify new Fe-S proteins, the development and pathological analysis of animal and cellular models to dissect the molecular mechanisms, and transcriptomic analysis to uncover the common pathways among recessive ataxias. A specific focus of the proposal will be the involvement of DNA damage response pathways in neuronal dysfunction, as several DNA repair enzymes have recently been identified as Fe-S proteins and thus might be directly affected by frataxin and ABCb7 deficiency. This proposal should lead to the identification of different pathways for therapeutic intervention for these devastating disorders.
Summary
The project aims at unraveling the molecular pathophysiology of recessive ataxias, a heterogeneous set of severely disabling neurodegenerative disorders due to loss of function of proteins involved either in mitochondrial/metabolic pathways or DNA repair. Friedreich ataxia, the most common form, is due to partial loss of function of frataxin, a mitochondrial protein involved in iron-sulfur cluster (ISC) biogenesis. Furthermore, the rare X-linked sideroblastic anemia with cerebellar ataxia is caused by mutation in ABCb7, an ATP-binding cassette transporter of the mitochondrial inner membrane necessary for cytosolic ISC export. ISC are versatile co-factors of proteins involved in electron transport, enzyme catalysis and regulation of gene expression. The synthesis and insertion of ISC into apoproteins involve complex machineries that are still poorly understood in the mammalian cell. The objectives of this proposal are: 1) to elucidate ISC biogenesis and metabolism in the mammalian cell, with an emphasis on the role of frataxin and ABCb7; 2) to better understand the molecular pathways that are involved in neuronal dysfunction due to defects in mitochondrial ISC metabolism. These objectives will be accomplished by a multidisciplinary approach combining molecular and biochemical approaches to study the ISC assembly machineries, bioinformatic and proteomic studies to identify new Fe-S proteins, the development and pathological analysis of animal and cellular models to dissect the molecular mechanisms, and transcriptomic analysis to uncover the common pathways among recessive ataxias. A specific focus of the proposal will be the involvement of DNA damage response pathways in neuronal dysfunction, as several DNA repair enzymes have recently been identified as Fe-S proteins and thus might be directly affected by frataxin and ABCb7 deficiency. This proposal should lead to the identification of different pathways for therapeutic intervention for these devastating disorders.
Max ERC Funding
1 449 924 €
Duration
Start date: 2008-07-01, End date: 2013-06-30
Project acronym ISLETMESENCHYME
Project ß-cell Dysfunction in Diabetes: Elucidating the Role of Islet-Associated Mesenchymal Cells
Researcher (PI) Limor Landsman
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS4, ERC-2013-StG
Summary "Glucose homeostasis relies on tightly controlled release of insulin by pancreatic beta-cells. Diabetes, characterized by increased blood glucose levels, is a chronic disease now reaching epidemic proportions. The most common form of this disease is Type 2 diabetes (T2D), which was previously regarded as a disease of insulin resistance. However, work over the past decade
had shifted this paradigm by implicating beta-cell failure as a key factor in this disease. Despite major progress, the cellular and molecular basis of this T2D is far from being elucidated. Here, I present a novel pancreatic cell population, islet-associated mesenchymal cells (isMCs), which are with close contact to beta-cells in both human and mouse pancreata. My preliminary findings revealed that isMCs function to maintain beta-cells maturity and functionality. I therefore hypothesize that impaired isMCs function serve as an underlying cause for diabetes. To test this hypothesis, we will characterize the continuous requirement of isMCs for glucose homeostasis by their specific depletion in vivo. Next, we will link genes associated with T2D to isMCs function, by manipulating their expression and elucidating the effect on beta-cell function. Finally, we will investigate the source of diabetes prevalence found in pancreatic cancer and pancreatitis patients, by identifying how isMCs ability to maintain beta-cell function is affected in these diseases. To this end, we will use transgenic mouse models and culture systems to specifically manipulate cells and genes, and to study the resultant effect on beta-cell phenotype and glucose homeostasis. The implications of this work are far reaching as they will point to isMCs as a new player in glucose regulation, and as a contributor to beta-cell dysfunction in diabetes. Furthermore, the findings of this study will implicate isMCs a novel target for therapeutic approaches to diabetes, a currently unmet medical need."
Summary
"Glucose homeostasis relies on tightly controlled release of insulin by pancreatic beta-cells. Diabetes, characterized by increased blood glucose levels, is a chronic disease now reaching epidemic proportions. The most common form of this disease is Type 2 diabetes (T2D), which was previously regarded as a disease of insulin resistance. However, work over the past decade
had shifted this paradigm by implicating beta-cell failure as a key factor in this disease. Despite major progress, the cellular and molecular basis of this T2D is far from being elucidated. Here, I present a novel pancreatic cell population, islet-associated mesenchymal cells (isMCs), which are with close contact to beta-cells in both human and mouse pancreata. My preliminary findings revealed that isMCs function to maintain beta-cells maturity and functionality. I therefore hypothesize that impaired isMCs function serve as an underlying cause for diabetes. To test this hypothesis, we will characterize the continuous requirement of isMCs for glucose homeostasis by their specific depletion in vivo. Next, we will link genes associated with T2D to isMCs function, by manipulating their expression and elucidating the effect on beta-cell function. Finally, we will investigate the source of diabetes prevalence found in pancreatic cancer and pancreatitis patients, by identifying how isMCs ability to maintain beta-cell function is affected in these diseases. To this end, we will use transgenic mouse models and culture systems to specifically manipulate cells and genes, and to study the resultant effect on beta-cell phenotype and glucose homeostasis. The implications of this work are far reaching as they will point to isMCs as a new player in glucose regulation, and as a contributor to beta-cell dysfunction in diabetes. Furthermore, the findings of this study will implicate isMCs a novel target for therapeutic approaches to diabetes, a currently unmet medical need."
Max ERC Funding
1 105 440 €
Duration
Start date: 2013-10-01, End date: 2018-09-30
Project acronym ISLETVASC
Project Molecular Mechanisms Regulating Pancreatic Islet Vascularization
Researcher (PI) Matthew Poy
Host Institution (HI) MAX DELBRUECK CENTRUM FUER MOLEKULARE MEDIZIN IN DER HELMHOLTZ-GEMEINSCHAFT (MDC)
Call Details Starting Grant (StG), LS4, ERC-2010-StG_20091118
Summary Many reports indicate the number of people with diabetes will exceed 350 million by the year 2030. Both type 1 and type 2 diabetes are characterized by the deterioration and impaired function of pancreatic b-cells. While transplantation is a promising strategy to replace lost tissue, several obstacles remain in the pathway to its clinical application. Whether b-cells are derived from patient samples or differentiated from embryonic stem cells, a major concern facing these strategies is how a recipient will respond to transplanted foreign tissue. Since the native environment for pancreatic islets is comprised of neural and vascular networks, successful integration may depend upon signals received from these neighboring cell types. Using a multidisciplinary approach, the principal investigator plans to elucidate molecular mechanisms underlying the interactions between pancreatic islet cells and their neighboring endothelial cells. Developing an understanding of how these interactions change during the pathogenesis of disease will provide insight into how islet growth and insulin release is dependent upon signals received from adjacent cell types. Emphasis will be placed on genetic mouse models to measure changes in gene expression in both isolated pancreatic b-cells and endothelial cells to identify genes that mediate the interaction between these cell types. In addition, it is of great interest to identify secreted factors that may constitute autocrine or paracrine signalling mechanisms that influence growth and function between these cell types. Furthermore, it will be determined whether current protocols for the differentiation of mouse stem cells into insulin producing cells are improved by restoring the expression of genes which facilitate communication to endothelial cells. This project aims to identify genes essential to the vascular context of pancreatic b-cells to improve transplantation protocols and facilitate the development of therapeutic strategies for diabetes.
Summary
Many reports indicate the number of people with diabetes will exceed 350 million by the year 2030. Both type 1 and type 2 diabetes are characterized by the deterioration and impaired function of pancreatic b-cells. While transplantation is a promising strategy to replace lost tissue, several obstacles remain in the pathway to its clinical application. Whether b-cells are derived from patient samples or differentiated from embryonic stem cells, a major concern facing these strategies is how a recipient will respond to transplanted foreign tissue. Since the native environment for pancreatic islets is comprised of neural and vascular networks, successful integration may depend upon signals received from these neighboring cell types. Using a multidisciplinary approach, the principal investigator plans to elucidate molecular mechanisms underlying the interactions between pancreatic islet cells and their neighboring endothelial cells. Developing an understanding of how these interactions change during the pathogenesis of disease will provide insight into how islet growth and insulin release is dependent upon signals received from adjacent cell types. Emphasis will be placed on genetic mouse models to measure changes in gene expression in both isolated pancreatic b-cells and endothelial cells to identify genes that mediate the interaction between these cell types. In addition, it is of great interest to identify secreted factors that may constitute autocrine or paracrine signalling mechanisms that influence growth and function between these cell types. Furthermore, it will be determined whether current protocols for the differentiation of mouse stem cells into insulin producing cells are improved by restoring the expression of genes which facilitate communication to endothelial cells. This project aims to identify genes essential to the vascular context of pancreatic b-cells to improve transplantation protocols and facilitate the development of therapeutic strategies for diabetes.
Max ERC Funding
1 496 257 €
Duration
Start date: 2010-11-01, End date: 2015-10-31
Project acronym JetNS
Project Relativistic Jets in Astrophysics -Compact binary mergers, Gamma-Ray Bursts, and Beyond
Researcher (PI) Ehud Nakar
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Consolidator Grant (CoG), PE9, ERC-2018-COG
Summary What is the origin of the electromagnetic (EM) counterparts of gravitational waves observed from compact binary mergers? What makes short gamma ray bursts (GRBs)? What are the sources of IceCube’s high-energy neutrinos? Are all core-collapse supernovae exploding via the same mechanism? These are some of the puzzles that have emerged with the rapid progress of time domain astronomy. Relativistic jets in compact binary mergers and GRBs, and their interaction with the surrounding media hold the key to these, and other, seemingly unrelated broad-impact questions. Here I propose a new forefront study of how relativistic jets interact with their surrounding media and of its numerous implications, focusing on compact binary mergers and GRBs.
The goal of this project is to study, first, the jet-media interaction, and the microphysics of the radiation-mediated shocks that it drives. I will then use the results, together with available observations, to learn about compact binary mergers, GRBs and SNe, sheding light on the questions listed above, and probing the nature of relativistic jets in general. Important goals will include: (i) General models for the propagation of relativistic jets in various media types. (ii) Modeling of the EM signal generated by jet-media interaction following compact binary mergers. (iii) Estimates of the neutrino signal from jet-media interaction in GRBs and SNe. (iv) Constraint the role of jets in SN explosions.
This project is timey as it comes at the beginning of a new multi-messenger era where the EM counterparts of GW sources are going to be detected on a regular basis and where the face of transient astrophysics is going to be changed by a range of large scale surveys such as LSST, the SKA, and more. This project will set the theoretical base for understanding numerous known and yet-to be discovered transients that will be detected in the next decade.
Summary
What is the origin of the electromagnetic (EM) counterparts of gravitational waves observed from compact binary mergers? What makes short gamma ray bursts (GRBs)? What are the sources of IceCube’s high-energy neutrinos? Are all core-collapse supernovae exploding via the same mechanism? These are some of the puzzles that have emerged with the rapid progress of time domain astronomy. Relativistic jets in compact binary mergers and GRBs, and their interaction with the surrounding media hold the key to these, and other, seemingly unrelated broad-impact questions. Here I propose a new forefront study of how relativistic jets interact with their surrounding media and of its numerous implications, focusing on compact binary mergers and GRBs.
The goal of this project is to study, first, the jet-media interaction, and the microphysics of the radiation-mediated shocks that it drives. I will then use the results, together with available observations, to learn about compact binary mergers, GRBs and SNe, sheding light on the questions listed above, and probing the nature of relativistic jets in general. Important goals will include: (i) General models for the propagation of relativistic jets in various media types. (ii) Modeling of the EM signal generated by jet-media interaction following compact binary mergers. (iii) Estimates of the neutrino signal from jet-media interaction in GRBs and SNe. (iv) Constraint the role of jets in SN explosions.
This project is timey as it comes at the beginning of a new multi-messenger era where the EM counterparts of GW sources are going to be detected on a regular basis and where the face of transient astrophysics is going to be changed by a range of large scale surveys such as LSST, the SKA, and more. This project will set the theoretical base for understanding numerous known and yet-to be discovered transients that will be detected in the next decade.
Max ERC Funding
1 981 250 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym KATP-DIABETES
Project ATP-sensitive potassium channels: from atomic structure to human disease
Researcher (PI) Frances Mary Ashcroft
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Advanced Grant (AdG), LS4, ERC-2012-ADG_20120314
Summary We are currently experiencing a fast-growing diabetes pandemic. Both type 2 diabetes and rare monogenic forms of diabetes, such as neonatal diabetes, are characterised by impaired insulin secretion. This project seeks to resolve the fundamental mechanisms underlying insulin secretion and its failure in diabetes. We have shown that activating mutations in the ATP-sensitive potassium (KATP) channel cause neonatal diabetes, which has enabled children with this disease to switch from insulin injections to oral sulphonylurea drugs (which block their open KATP channels and stimulate insulin release). The most severe mutations also cause neurological symptoms that, for unknown reasons, are less well treated by sulphonylureas. We aim to: obtain a detailed mechanistic understanding of how nucleotides and drugs regulate KATP channel activity by combining state-of-the-art structural and functional approaches; define how drug therapy affects glucose homeostasis in neonatal diabetes; and explore how activating KATP channel mutations affect glucagon release from pancreatic alpha-cells. We will also investigate how severe KATP channel mutations cause neurological symptoms (such as developmental delay, reduced sensitivity to general anaesthetics and impaired eye movements) and determine how these might be alleviated by drug therapy. While underpinned by my previous work, this project takes my research in new directions, including structural analysis of eukaryotic membrane proteins, stimulus-secretion coupling in other types of islet cell, and neurological studies in humans as well as animal models. It involves a broad multidisciplinary approach, addresses questions of fundamental scientific importance, and has a strong translational element. We expect our studies will be of direct benefit to patients with neonatal or type 2 diabetes.
Summary
We are currently experiencing a fast-growing diabetes pandemic. Both type 2 diabetes and rare monogenic forms of diabetes, such as neonatal diabetes, are characterised by impaired insulin secretion. This project seeks to resolve the fundamental mechanisms underlying insulin secretion and its failure in diabetes. We have shown that activating mutations in the ATP-sensitive potassium (KATP) channel cause neonatal diabetes, which has enabled children with this disease to switch from insulin injections to oral sulphonylurea drugs (which block their open KATP channels and stimulate insulin release). The most severe mutations also cause neurological symptoms that, for unknown reasons, are less well treated by sulphonylureas. We aim to: obtain a detailed mechanistic understanding of how nucleotides and drugs regulate KATP channel activity by combining state-of-the-art structural and functional approaches; define how drug therapy affects glucose homeostasis in neonatal diabetes; and explore how activating KATP channel mutations affect glucagon release from pancreatic alpha-cells. We will also investigate how severe KATP channel mutations cause neurological symptoms (such as developmental delay, reduced sensitivity to general anaesthetics and impaired eye movements) and determine how these might be alleviated by drug therapy. While underpinned by my previous work, this project takes my research in new directions, including structural analysis of eukaryotic membrane proteins, stimulus-secretion coupling in other types of islet cell, and neurological studies in humans as well as animal models. It involves a broad multidisciplinary approach, addresses questions of fundamental scientific importance, and has a strong translational element. We expect our studies will be of direct benefit to patients with neonatal or type 2 diabetes.
Max ERC Funding
2 478 420 €
Duration
Start date: 2013-09-01, End date: 2018-08-31
Project acronym KERNEL
Project Ultimate Angular Resolution Astrophysics with kernel-phase and full-aperture interferometry
Researcher (PI) Frantz Martinache
Host Institution (HI) OBSERVATOIRE DE LA COTE D'AZUR (OCA)
Call Details Consolidator Grant (CoG), PE9, ERC-2015-CoG
Summary Astronomy requires large telescopes to improve the sensitivity and the angular resolution of its observations. Of these qualities, angular resolution is the most difficult to maintain in the optical and near-infrared, since the atmosphere reduces it to that of a 10 cm aperture, regardless of the telescope size. On the one-hand, Adaptive Optics (AO) actively compensates for this effect but the improvement is often partial only. On the other hand, interferometric techniques (most notably sparse aperture masking interferometry) passively allow the extraction of self-calibrating observables, that boost the angular resolution, but severely affect the sensitivity of observations. A framework newly established by the PI of the proposal however now makes it possible to extract generalized self-calibrating observables called kernel-phases from conventional AO-corrected images. The work outlined in this proposal will make it possible to scientifically exploit the high angular resolution imaging capability of this technique, to improve its robustness and to expand its capabilities. The framework offers a very general purpose high angular resolution imaging tool for astronomers as well as wavefront control experts. This proposal is organized in five work-packages of increasing challenge that include: the reinterpretation of existing archival data
with a super-resolution capability, the expansion of its robustness to open up new more challenging use-cases, a special focus on the development of a very high-dynamic range mode, the adaptation of interferometric image reconstruction techniques, and the development of new advanced AO concepts. The consequences of this project will have a major impact on the design and scientific exploitation of future high angular resolution instrumentation on the existing generation of 8-10 meter class telescopes as well as on the upcoming generation of 30-40 meter giants, championned by Europe and its E-ELT.
Summary
Astronomy requires large telescopes to improve the sensitivity and the angular resolution of its observations. Of these qualities, angular resolution is the most difficult to maintain in the optical and near-infrared, since the atmosphere reduces it to that of a 10 cm aperture, regardless of the telescope size. On the one-hand, Adaptive Optics (AO) actively compensates for this effect but the improvement is often partial only. On the other hand, interferometric techniques (most notably sparse aperture masking interferometry) passively allow the extraction of self-calibrating observables, that boost the angular resolution, but severely affect the sensitivity of observations. A framework newly established by the PI of the proposal however now makes it possible to extract generalized self-calibrating observables called kernel-phases from conventional AO-corrected images. The work outlined in this proposal will make it possible to scientifically exploit the high angular resolution imaging capability of this technique, to improve its robustness and to expand its capabilities. The framework offers a very general purpose high angular resolution imaging tool for astronomers as well as wavefront control experts. This proposal is organized in five work-packages of increasing challenge that include: the reinterpretation of existing archival data
with a super-resolution capability, the expansion of its robustness to open up new more challenging use-cases, a special focus on the development of a very high-dynamic range mode, the adaptation of interferometric image reconstruction techniques, and the development of new advanced AO concepts. The consequences of this project will have a major impact on the design and scientific exploitation of future high angular resolution instrumentation on the existing generation of 8-10 meter class telescopes as well as on the upcoming generation of 30-40 meter giants, championned by Europe and its E-ELT.
Max ERC Funding
1 717 811 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym KETJU
Project Post-Newtonian modelling of the dynamics of supermassive black holes in galactic-scale hydrodynamical simulations (KETJU)
Researcher (PI) Peter Hilding JOHANSSON
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Consolidator Grant (CoG), PE9, ERC-2018-COG
Summary Supermassive black holes (SMBHs) with masses in the range ~10^6-10^10 M⊙ are found at the centres of all massive galaxies in the Local Universe. In the ΛCDM picture of structure formation galaxies grow bottom-up through mergers and gas accretion, leading to multiple SMBHs in the same stellar system. Current simulation codes are unable to resolve in a single simulation the full SMBH merging process, which involves dynamical friction, three-body interactions and finally gravitational wave (GW) emission. KETJU will provide a significant breakthrough in SMBH research by following for the first time accurately global galactic-scale dynamical and gaseous astrophysical processes, while simultaneously solving the dynamics of SMBHs, SMBH binaries and surrounding stellar systems at sub-parsec scales. Our code KETJU (the word for 'chain' in Finnish) is built on the GADGET-3 code and it includes regions around every SMBH in which the dynamics of SMBHs and stellar particles is modelled using a non-softened Post-Newtonian algorithmic chain regularisation technique. The remaining simulation particles far from the SMBHs are evolved using softened GADGET-3. Using KETJU we can study at unprecedented accuracy the dynamics of SMBHs to separations of ~10 Schwarzschild radii, the formation of cores in massive galaxies, the formation of nuclear stellar clusters and finally provide a realistic prediction for the amplitude and frequency distribution of the cosmological gravitational wave background. The UH theoretical extragalactic team is ideally suited for this project, as it has an unusually versatile background in modelling the dynamics, feedback and merging of SMBHs. KETJU is also particularly timely, as the spectacular direct detection of GWs in 2016 is paving the way for a new era in gravitational wave astronomy. Future space-borne GW observatories, such as the European Space Agency's LISA, require accurate global GW predictions in order to fully realise their science goals.
Summary
Supermassive black holes (SMBHs) with masses in the range ~10^6-10^10 M⊙ are found at the centres of all massive galaxies in the Local Universe. In the ΛCDM picture of structure formation galaxies grow bottom-up through mergers and gas accretion, leading to multiple SMBHs in the same stellar system. Current simulation codes are unable to resolve in a single simulation the full SMBH merging process, which involves dynamical friction, three-body interactions and finally gravitational wave (GW) emission. KETJU will provide a significant breakthrough in SMBH research by following for the first time accurately global galactic-scale dynamical and gaseous astrophysical processes, while simultaneously solving the dynamics of SMBHs, SMBH binaries and surrounding stellar systems at sub-parsec scales. Our code KETJU (the word for 'chain' in Finnish) is built on the GADGET-3 code and it includes regions around every SMBH in which the dynamics of SMBHs and stellar particles is modelled using a non-softened Post-Newtonian algorithmic chain regularisation technique. The remaining simulation particles far from the SMBHs are evolved using softened GADGET-3. Using KETJU we can study at unprecedented accuracy the dynamics of SMBHs to separations of ~10 Schwarzschild radii, the formation of cores in massive galaxies, the formation of nuclear stellar clusters and finally provide a realistic prediction for the amplitude and frequency distribution of the cosmological gravitational wave background. The UH theoretical extragalactic team is ideally suited for this project, as it has an unusually versatile background in modelling the dynamics, feedback and merging of SMBHs. KETJU is also particularly timely, as the spectacular direct detection of GWs in 2016 is paving the way for a new era in gravitational wave astronomy. Future space-borne GW observatories, such as the European Space Agency's LISA, require accurate global GW predictions in order to fully realise their science goals.
Max ERC Funding
1 953 569 €
Duration
Start date: 2019-07-01, End date: 2024-06-30
Project acronym KIDNEY CANCER
Project Molecular mechanisms underlying control of renal epithelial proliferative homeostasis
Researcher (PI) Ian James Frew
Host Institution (HI) UNIVERSITAT ZURICH
Call Details Starting Grant (StG), LS4, ERC-2010-StG_20091118
Summary This research grant has two major aspects. The first seeks to understand the molecular and cellular basis of the evolution of clear cell renal cell carcinoma(ccRCC), the most frequent form of kidney cancer. We will utilise an integrated approach based on mouse genetics, the use of primary kidney epithelial cell culture systems, genetic screening approaches using RNA interference libraries and analysis of the genetic and molecular changes that arise in human kidney tumours. The rationale behind these studies is that by better understanding the molecular causes of ccRCC it will be possible to identify new molecules or signaling pathways that could serve as appropriate therapeutic targets. The second aspect of this grant relates to the development of a flexible experimental platform that will allow the rapid and simultaneous up- and down-regulation of gene expression in the mouse kidney in a manner in which the affected cells are marked by a luminescent marker. This system will be based on the injection of modified lentiviral gene overexpression and gene knockdown vectors, allowing us to exploit recently-developed genome-wide cDNA libraries and RNA interference libraries. This experimental system should be equally applicable to other organ systems and will allow for the first time a systematic approach to the manipulation of gene expression in living mice, additionally bypassing the time limitations associated with conventional mouse genetic approaches. We aim to develop this system within the biological context of this grant and will combine it with live-animal imaging approaches to generate a series of mouse models of ccRCC. These will ultimately serve as invaluable tools for testing novel therapeutic approaches against this currently untreatable disease.
Summary
This research grant has two major aspects. The first seeks to understand the molecular and cellular basis of the evolution of clear cell renal cell carcinoma(ccRCC), the most frequent form of kidney cancer. We will utilise an integrated approach based on mouse genetics, the use of primary kidney epithelial cell culture systems, genetic screening approaches using RNA interference libraries and analysis of the genetic and molecular changes that arise in human kidney tumours. The rationale behind these studies is that by better understanding the molecular causes of ccRCC it will be possible to identify new molecules or signaling pathways that could serve as appropriate therapeutic targets. The second aspect of this grant relates to the development of a flexible experimental platform that will allow the rapid and simultaneous up- and down-regulation of gene expression in the mouse kidney in a manner in which the affected cells are marked by a luminescent marker. This system will be based on the injection of modified lentiviral gene overexpression and gene knockdown vectors, allowing us to exploit recently-developed genome-wide cDNA libraries and RNA interference libraries. This experimental system should be equally applicable to other organ systems and will allow for the first time a systematic approach to the manipulation of gene expression in living mice, additionally bypassing the time limitations associated with conventional mouse genetic approaches. We aim to develop this system within the biological context of this grant and will combine it with live-animal imaging approaches to generate a series of mouse models of ccRCC. These will ultimately serve as invaluable tools for testing novel therapeutic approaches against this currently untreatable disease.
Max ERC Funding
1 500 000 €
Duration
Start date: 2010-12-01, End date: 2015-11-30
Project acronym KRABNKAP
Project KRAB/KAP1-mediated gene regulation in mammalian physiology and human diseases
Researcher (PI) Didier Trono
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), LS4, ERC-2010-AdG_20100317
Summary This project aims at exploring the roles or KRAB/KAP1-mediated gene regulation in mammalian physiology and the possible impact of its dysfunctions on human health. The proper control of gene expression is paramount to all biological events, and is orchestrated through a sophisticated balance of activating and repressing influences. The mouse and human genomes contain around four hundred genes encoding KRAB-containing zinc finger proteins (KRAB-ZFPs), a family of tetrapod-restricted sequence-specific DNA-binding transcriptional repressors. Even though these KRAB-ZFPs represent the single largest group of transcriptional regulators encoded by higher vertebrates, their functions remain largely unknown. Nevertheless, it has been established that they share an essential cofactor, the histone methyltransferase- and histone deacetylase-recruiting KAP1, and act by triggering the formation of heterochromatin. KAP1 is ubiquitous, and KRAB-ZFPs are present in most if not all cells, albeit along distinctly cell type-, stage- and state-specific patterns, suggesting that KRAB/KAP1 gene regulation influences a very large number of physiological events. A few years ago, we launched a program aimed at addressing this hypothesis through a combination of genetic, functional and molecular studies focused on two paradigmatic organs, the lympho-hematopoietic system and the liver. Our preliminary results confirm that KRAB/KAP1-mediated transcriptional control is a master regulator of mammalian homeostasis. Accordingly, we now propose to dissect the regulatory networks orchestrated by KAP1 and KRAB-ZFPs in these two systems, to identify their gene targets and the mechanisms of their control, and to probe their possible implication in human pathologies targeting these organs.
Summary
This project aims at exploring the roles or KRAB/KAP1-mediated gene regulation in mammalian physiology and the possible impact of its dysfunctions on human health. The proper control of gene expression is paramount to all biological events, and is orchestrated through a sophisticated balance of activating and repressing influences. The mouse and human genomes contain around four hundred genes encoding KRAB-containing zinc finger proteins (KRAB-ZFPs), a family of tetrapod-restricted sequence-specific DNA-binding transcriptional repressors. Even though these KRAB-ZFPs represent the single largest group of transcriptional regulators encoded by higher vertebrates, their functions remain largely unknown. Nevertheless, it has been established that they share an essential cofactor, the histone methyltransferase- and histone deacetylase-recruiting KAP1, and act by triggering the formation of heterochromatin. KAP1 is ubiquitous, and KRAB-ZFPs are present in most if not all cells, albeit along distinctly cell type-, stage- and state-specific patterns, suggesting that KRAB/KAP1 gene regulation influences a very large number of physiological events. A few years ago, we launched a program aimed at addressing this hypothesis through a combination of genetic, functional and molecular studies focused on two paradigmatic organs, the lympho-hematopoietic system and the liver. Our preliminary results confirm that KRAB/KAP1-mediated transcriptional control is a master regulator of mammalian homeostasis. Accordingly, we now propose to dissect the regulatory networks orchestrated by KAP1 and KRAB-ZFPs in these two systems, to identify their gene targets and the mechanisms of their control, and to probe their possible implication in human pathologies targeting these organs.
Max ERC Funding
2 499 996 €
Duration
Start date: 2011-04-01, End date: 2016-03-31
Project acronym KRASHIMPE
Project KRas mutation interactions with host immunity in malignant pleural effusion
Researcher (PI) Georgios Stathopoulos
Host Institution (HI) PANEPISTIMIO PATRON
Call Details Starting Grant (StG), LS4, ERC-2010-StG_20091118
Summary Malignant pleural effusion (MPE) is a significant problem most commonly caused by adenocarcinomas. Although tumors involving the pleura vary in their ability to produce MPE, pathways critical for MPE formation are poorly defined. We have found that mouse tumors harboring mutant (”)KRas produce MPE in mice while tumors without ”KRas do not. LLC and MC38 lung and colon adenocarcinomas, potent inducers of MPE in syngeneic mice, harbor ”KRas that drives constitutive Ras and alternative nuclear factor (NF)-ºB signaling, inflammatory gene expression, and recruitment of specific myeloid cells to the pleural space. In contrast, mouse B16 melanoma and AE17 mesothelioma have wtKRas, lack constitutive Ras/alternative NF-º’ signaling, and are incapable of forming MPE. RNAi-mediated silencing of KRas in MC38 tumors abrogated MPE formation and Ras/alternative NF-º’ activation, while these phenomena were reconstituted in B16 tumors after KRas overexpression. We hypothesize that Ras-activating mutations drive the inflammatory phenotype of adenocarcinomas critical for MPE formation, which is characterized by Ras/alternative NF-ºB activation, inflammatory signalling to host vasculature/immune system, and recruitment of specific myeloid cells, and results in endothelial proliferation/leakiness. To test this hypothesis, we propose to: 1) define the relationship between Ras-activating mutations (RAM) and MPE formation; 2) identify tumor cell Ras-dependent signalling pathways and gene expression signature critical for MPE formation; 3) investigate the host response to tumor cells with RAM that results in MPE; and 4) target Ras and dependent signalling pathways as potential therapy for MPE. Studies will be performed using delivery of mouse/human tumors with/without RAM into the pleura of syngeneic/immunocompromized mice and are likely to yield new insights into the mechanisms of pleural tumor progression and to identify novel approaches to treatment of cancer patients with MPE.
Summary
Malignant pleural effusion (MPE) is a significant problem most commonly caused by adenocarcinomas. Although tumors involving the pleura vary in their ability to produce MPE, pathways critical for MPE formation are poorly defined. We have found that mouse tumors harboring mutant (”)KRas produce MPE in mice while tumors without ”KRas do not. LLC and MC38 lung and colon adenocarcinomas, potent inducers of MPE in syngeneic mice, harbor ”KRas that drives constitutive Ras and alternative nuclear factor (NF)-ºB signaling, inflammatory gene expression, and recruitment of specific myeloid cells to the pleural space. In contrast, mouse B16 melanoma and AE17 mesothelioma have wtKRas, lack constitutive Ras/alternative NF-º’ signaling, and are incapable of forming MPE. RNAi-mediated silencing of KRas in MC38 tumors abrogated MPE formation and Ras/alternative NF-º’ activation, while these phenomena were reconstituted in B16 tumors after KRas overexpression. We hypothesize that Ras-activating mutations drive the inflammatory phenotype of adenocarcinomas critical for MPE formation, which is characterized by Ras/alternative NF-ºB activation, inflammatory signalling to host vasculature/immune system, and recruitment of specific myeloid cells, and results in endothelial proliferation/leakiness. To test this hypothesis, we propose to: 1) define the relationship between Ras-activating mutations (RAM) and MPE formation; 2) identify tumor cell Ras-dependent signalling pathways and gene expression signature critical for MPE formation; 3) investigate the host response to tumor cells with RAM that results in MPE; and 4) target Ras and dependent signalling pathways as potential therapy for MPE. Studies will be performed using delivery of mouse/human tumors with/without RAM into the pleura of syngeneic/immunocompromized mice and are likely to yield new insights into the mechanisms of pleural tumor progression and to identify novel approaches to treatment of cancer patients with MPE.
Max ERC Funding
1 995 000 €
Duration
Start date: 2011-04-01, End date: 2016-03-31
Project acronym LEAP
Project Large European Array for Pulsars
Researcher (PI) Michael Kramer
Host Institution (HI) THE UNIVERSITY OF MANCHESTER
Call Details Advanced Grant (AdG), PE9, ERC-2008-AdG
Summary In general relativity and other relativistic theories of gravity, space and time are combined to form ``space-time'' which is curved in the presence of mass. As masses move, for instance like the two components in a binary system, ripples in space-time are created that propagate through the Universe, very much like waves caused by a stone falling into a pond. These ``gravitational waves'' (GWs) are known to exist from the effect that they have on a system of two orbiting stars. After inferring their existence indirectly, the next great challenge is the {\em direct} detection of GWs. While this is the aim of a number of gravitational wave detectors around the world, a detection has not been made. Fortunately, a method exists that allows us today to detect GWs directly, in a frequency range that is much lower but complementary to those covered by ground-based detectors. This method utilises the radio astronomical observations of a special type of star known as radio pulsars. We propose an experiment to achieve the ground-breaking goal of GW detection with the help of an innovative approach. At the heart of this approach, named LEAP, lies the goal to combine the collective power of Europe's biggest radio-telescopes to form the biggest fully-steerable telescope on Earth, providing a ``leap'' in our sensitivity to go beyond the threshold that delivers the first direct detection of GWs. While the rewards for a successful detection of GWs are immense, we demonstrate that this is possible by harvesting the experience and resources uniquely available in Europe.
Summary
In general relativity and other relativistic theories of gravity, space and time are combined to form ``space-time'' which is curved in the presence of mass. As masses move, for instance like the two components in a binary system, ripples in space-time are created that propagate through the Universe, very much like waves caused by a stone falling into a pond. These ``gravitational waves'' (GWs) are known to exist from the effect that they have on a system of two orbiting stars. After inferring their existence indirectly, the next great challenge is the {\em direct} detection of GWs. While this is the aim of a number of gravitational wave detectors around the world, a detection has not been made. Fortunately, a method exists that allows us today to detect GWs directly, in a frequency range that is much lower but complementary to those covered by ground-based detectors. This method utilises the radio astronomical observations of a special type of star known as radio pulsars. We propose an experiment to achieve the ground-breaking goal of GW detection with the help of an innovative approach. At the heart of this approach, named LEAP, lies the goal to combine the collective power of Europe's biggest radio-telescopes to form the biggest fully-steerable telescope on Earth, providing a ``leap'' in our sensitivity to go beyond the threshold that delivers the first direct detection of GWs. While the rewards for a successful detection of GWs are immense, we demonstrate that this is possible by harvesting the experience and resources uniquely available in Europe.
Max ERC Funding
2 455 285 €
Duration
Start date: 2009-01-01, End date: 2014-09-30
Project acronym LEDA
Project The challenging quest for low-mass dark structures
Researcher (PI) Simona VEGETTI
Host Institution (HI) MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN EV
Call Details Starting Grant (StG), PE9, ERC-2017-STG
Summary Using strong gravitational lensing, I will constrain with my unique modelling technique and acquired knowledge the properties of dark matter and potentially revise the current standard paradigm for the formation of all structures which is at the core of modern cosmology and galaxy formation theories.
Numerical simulations of cosmic structure formation have shown that the amount of mass in low-mass objects depends strongly on the assumed nature of dark matter. My goal is to constrain the nature of dark matter by measuring the dark matter mass function down to ~10^6 M_sol, where the predictions from different currently viable dark matter models differ by large factors.
To this end, I will use the gravitational imaging technique, an advanced modelling tool that I have developed and pioneered, and state-of-the-art strong gravitational lensing data for 12 systems observed with cm- and mm-interferometers. At present, this is the only observational probe of low-mass structure in the dark matter distribution beyond the Local Universe.
This will represent an important milestone in our understanding of the dark Universe and will provide a key observational test of the Cold Dark Matter model in a regime that has not been probed before. This ERC project will challenge our standard model for small-scale structure formation and will distinguish between “warm” and “cold” hypothesis for the nature of dark matter. This ERC project will have significant implications for the fields of cosmology and galaxy formation.
I am in a unique position to achieve the scientific goal here proposed. I have extended experience in studying gravitational lenses and low mass dark structures. I have an unmatched gravitational lens modelling code and high quality data. With this ERC I will build upon my previous successes and create a top-class research group for studying dark matter with gravitational lensing.
Summary
Using strong gravitational lensing, I will constrain with my unique modelling technique and acquired knowledge the properties of dark matter and potentially revise the current standard paradigm for the formation of all structures which is at the core of modern cosmology and galaxy formation theories.
Numerical simulations of cosmic structure formation have shown that the amount of mass in low-mass objects depends strongly on the assumed nature of dark matter. My goal is to constrain the nature of dark matter by measuring the dark matter mass function down to ~10^6 M_sol, where the predictions from different currently viable dark matter models differ by large factors.
To this end, I will use the gravitational imaging technique, an advanced modelling tool that I have developed and pioneered, and state-of-the-art strong gravitational lensing data for 12 systems observed with cm- and mm-interferometers. At present, this is the only observational probe of low-mass structure in the dark matter distribution beyond the Local Universe.
This will represent an important milestone in our understanding of the dark Universe and will provide a key observational test of the Cold Dark Matter model in a regime that has not been probed before. This ERC project will challenge our standard model for small-scale structure formation and will distinguish between “warm” and “cold” hypothesis for the nature of dark matter. This ERC project will have significant implications for the fields of cosmology and galaxy formation.
I am in a unique position to achieve the scientific goal here proposed. I have extended experience in studying gravitational lenses and low mass dark structures. I have an unmatched gravitational lens modelling code and high quality data. With this ERC I will build upon my previous successes and create a top-class research group for studying dark matter with gravitational lensing.
Max ERC Funding
1 359 688 €
Duration
Start date: 2018-02-01, End date: 2023-01-31
Project acronym LEGA-C
Project The Physics of Galaxies 7 Gyr Ago
Researcher (PI) Arjen Van der wel
Host Institution (HI) UNIVERSITEIT GENT
Call Details Consolidator Grant (CoG), PE9, ERC-2015-CoG
Summary Over the past decade, redshift
surveys and multi-wavelength imaging campaigns have drawn up an
empirical picture of how many stars had formed in which types of
galaxies over the history of the universe. However, we have yet to
unravel the individual pathways along which galaxies evolve, and the
physical processes that drive them. Continuing with the previous
approach -- larger and deeper photometric samples -- is not adequate
to achieve this goal. A change of focus is required.
In this ERC project I will embark on a new way to address the question
of galaxy evolution. I will do so as Principle Investigator of the
recently approved LEGA-C observing program that has been allocated 128
nights of observation time over the next 4 years with ESO's flagship
facility the Very Large Telescope. This new survey will produce for
2500 distant (at z~1) galaxies with, for the first time,
sufficient resolution and S/N to measure ages and chemical
compositions of their stellar populations as well as internal velocity
dispersions and dynamical masses. This will provide an entirely new
physical description of the galaxy population 7 Gyr ago, with which I
will finally be able solve long-standing questions in galaxy formation
that were out of reach before: what is the star-formation history of
individual galaxies, why and how is star-formation ``quenched'' in
many galaxies, and to what extent do galaxies grow subsequently
through merging afterward?
LEGA-C is worldwide the largest spectroscopic survey of distant
galaxies to date, and ERC funding will be absolutely critical in
harvesting this unparallelled database. I am seeking to extend my
research group to realize the scientific potential of this substantial
investment (6.5M Eur) of observational resources by the European
astronomy community. Timing of the execution of the VLT program is
perfectly matched with the timeline of this ERC program.
Summary
Over the past decade, redshift
surveys and multi-wavelength imaging campaigns have drawn up an
empirical picture of how many stars had formed in which types of
galaxies over the history of the universe. However, we have yet to
unravel the individual pathways along which galaxies evolve, and the
physical processes that drive them. Continuing with the previous
approach -- larger and deeper photometric samples -- is not adequate
to achieve this goal. A change of focus is required.
In this ERC project I will embark on a new way to address the question
of galaxy evolution. I will do so as Principle Investigator of the
recently approved LEGA-C observing program that has been allocated 128
nights of observation time over the next 4 years with ESO's flagship
facility the Very Large Telescope. This new survey will produce for
2500 distant (at z~1) galaxies with, for the first time,
sufficient resolution and S/N to measure ages and chemical
compositions of their stellar populations as well as internal velocity
dispersions and dynamical masses. This will provide an entirely new
physical description of the galaxy population 7 Gyr ago, with which I
will finally be able solve long-standing questions in galaxy formation
that were out of reach before: what is the star-formation history of
individual galaxies, why and how is star-formation ``quenched'' in
many galaxies, and to what extent do galaxies grow subsequently
through merging afterward?
LEGA-C is worldwide the largest spectroscopic survey of distant
galaxies to date, and ERC funding will be absolutely critical in
harvesting this unparallelled database. I am seeking to extend my
research group to realize the scientific potential of this substantial
investment (6.5M Eur) of observational resources by the European
astronomy community. Timing of the execution of the VLT program is
perfectly matched with the timeline of this ERC program.
Max ERC Funding
1 884 875 €
Duration
Start date: 2016-04-01, End date: 2021-03-31
Project acronym LENSNOVA
Project Cosmic Fireworks Première: Unravelling Enigmas of Type Ia Supernova Progenitor and Cosmology through Strong Lensing
Researcher (PI) Sherry Hsuan SUYU
Host Institution (HI) MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN EV
Call Details Consolidator Grant (CoG), PE9, ERC-2017-COG
Summary The LENSNOVA project proposes a breakthrough both in cosmology and stellar physics, capitalising on 10 years of experience in the field of strong lensing time delays. Type Ia supernovae (SNe Ia) can now be watched on prime seats through strong lensing, where multiple images of the same SN appear at different times/locations around a foreground lens galaxy. After the first SN appears, the next SN occurrence (of the same SN event) can be predicted and observed in its entirety for the first time in history and with unprecedented temporal sampling. Observations of the beginning of SN explosions are key to revealing SN progenitors, that have been under debate for decades. Strongly lensed SNe Ia also allow an independent measurement of the Hubble constant (H0) that sets the cosmic expansion rate. The independent measurement is important to ascertain the possible need of new physics beyond the standard cosmological model, given the tensions in current H0 measurements. Capitalising on the PI’s expertise in strong lensing and success in predicting the reappearance of the first strongly lensed core-collapse SN, the objectives of LENSNOVA are to (1) place the best constraints on SNe Ia progenitors, and (2) pioneer the use of strongly lensed SNe Ia as a new cosmological probe. This will shed light on the natures of SNe Ia progenitors and dark energy, two of the greatest puzzles in the present era.
The advent of the Large Synoptic Survey Telescope and the Euclid mission makes LENSNOVA particularly timely for building the first sample of ~5 strongly lensed SNe Ia. Accurate predictions of the time delays in lensed SNe Ia will allow multiwavelength observations of the reappearances of SNe, especially in the earliest days of explosion for the first time, to place the best constraints on SN progenitors. These lensed SNe Ia will also potentially yield an H0 measurement with 2% uncertainty. LENSNOVA has thus the potential to revolutionise both fields of stellar physics and cosmology.
Summary
The LENSNOVA project proposes a breakthrough both in cosmology and stellar physics, capitalising on 10 years of experience in the field of strong lensing time delays. Type Ia supernovae (SNe Ia) can now be watched on prime seats through strong lensing, where multiple images of the same SN appear at different times/locations around a foreground lens galaxy. After the first SN appears, the next SN occurrence (of the same SN event) can be predicted and observed in its entirety for the first time in history and with unprecedented temporal sampling. Observations of the beginning of SN explosions are key to revealing SN progenitors, that have been under debate for decades. Strongly lensed SNe Ia also allow an independent measurement of the Hubble constant (H0) that sets the cosmic expansion rate. The independent measurement is important to ascertain the possible need of new physics beyond the standard cosmological model, given the tensions in current H0 measurements. Capitalising on the PI’s expertise in strong lensing and success in predicting the reappearance of the first strongly lensed core-collapse SN, the objectives of LENSNOVA are to (1) place the best constraints on SNe Ia progenitors, and (2) pioneer the use of strongly lensed SNe Ia as a new cosmological probe. This will shed light on the natures of SNe Ia progenitors and dark energy, two of the greatest puzzles in the present era.
The advent of the Large Synoptic Survey Telescope and the Euclid mission makes LENSNOVA particularly timely for building the first sample of ~5 strongly lensed SNe Ia. Accurate predictions of the time delays in lensed SNe Ia will allow multiwavelength observations of the reappearances of SNe, especially in the earliest days of explosion for the first time, to place the best constraints on SN progenitors. These lensed SNe Ia will also potentially yield an H0 measurement with 2% uncertainty. LENSNOVA has thus the potential to revolutionise both fields of stellar physics and cosmology.
Max ERC Funding
1 992 500 €
Duration
Start date: 2018-06-01, End date: 2023-05-31
Project acronym LEUKEMIABARRIER
Project The Leukemia-Initiating Cell: Genetic Determinants, Escape Mechanisms and Ontogenic Influence
Researcher (PI) David Bryder
Host Institution (HI) LUNDS UNIVERSITET
Call Details Consolidator Grant (CoG), LS4, ERC-2013-CoG
Summary Acute myeloid leukemia (AML) is the most common malignant myeloid disorder in adults and strongly associated in incidence to advanced age. AML arises from immature hematopoietic progenitor cells via a sequential multistep process, but the nature of these steps remains to a large extent unknown. Therefore, while significant efforts have previously been invested in characterizing the molecular properties of late-stage AML, as diagnosed in patients, less information is available on the events that underlie leukemia initiation and progression. This includes the identity of potential mechanisms that restrict or eradicate developing leukemic cells; hurdles evaded at some point in time for AML to occur.
We have developed an inducible transgenic mouse model of AML that, when combined with high-resolution cell fractionation of primitive hematopoietic progenitor cells, offers a unique opportunity to track development of AML from the very first stages of cancer development. Using this, I propose to: 1) Identify and functionally validate molecular determinants that underlie why only some hematopoietic progenitor cells progress into AML, 2) To explore the extent and identity of immune surveillance/editing that accompany progression into AML, and 3) By building on my previous work on hematopoietic aging, to explore AML progression in the context of aging.
I anticipate the LEUKEMIABARRIER project to generate novel basic knowledge, not excluding with clinical relevance, with the potential to open up several new fields for further studies. This includes identification of novel cell-intrinsic regulators and immune responses, their underlying mechanisms, and their relationship to the increased incidence of AML with age.
Summary
Acute myeloid leukemia (AML) is the most common malignant myeloid disorder in adults and strongly associated in incidence to advanced age. AML arises from immature hematopoietic progenitor cells via a sequential multistep process, but the nature of these steps remains to a large extent unknown. Therefore, while significant efforts have previously been invested in characterizing the molecular properties of late-stage AML, as diagnosed in patients, less information is available on the events that underlie leukemia initiation and progression. This includes the identity of potential mechanisms that restrict or eradicate developing leukemic cells; hurdles evaded at some point in time for AML to occur.
We have developed an inducible transgenic mouse model of AML that, when combined with high-resolution cell fractionation of primitive hematopoietic progenitor cells, offers a unique opportunity to track development of AML from the very first stages of cancer development. Using this, I propose to: 1) Identify and functionally validate molecular determinants that underlie why only some hematopoietic progenitor cells progress into AML, 2) To explore the extent and identity of immune surveillance/editing that accompany progression into AML, and 3) By building on my previous work on hematopoietic aging, to explore AML progression in the context of aging.
I anticipate the LEUKEMIABARRIER project to generate novel basic knowledge, not excluding with clinical relevance, with the potential to open up several new fields for further studies. This includes identification of novel cell-intrinsic regulators and immune responses, their underlying mechanisms, and their relationship to the increased incidence of AML with age.
Max ERC Funding
1 999 714 €
Duration
Start date: 2014-07-01, End date: 2019-06-30
Project acronym LeukemiaEnviron
Project SIGNALING PROPENSITY IN THE MICROENVIRONMENT OF B CELL CHRONIC LYMPHOCYTIC LEUKEMIA
Researcher (PI) Marek Mraz
Host Institution (HI) Masarykova univerzita
Call Details Starting Grant (StG), LS4, ERC-2018-STG
Summary B cell chronic lymphocytic leukemia (CLL) is the most frequent leukemia in adults. CLL cells are characterized by their universal dependency on pro-survival and pro-proliferative signals from immune niches. To achieve this they constantly re-circulate between blood and lymph nodes, which is inhibited by novel microenvironment-targeting therapies such as “BCR inhibitors”. We aim to reveal how the malignant B cells change the propensity of their signalling pathways in response to the different microenvironments such as peripheral blood vs lymph node to obtain the proliferative signals. This is of major relevance for CLL, but also transferable to the biology of some other B cell malignancies and/or normal B cells. We analyzed the “finger print” of microenvironmental interactions in many CLL samples at various times during the disease course or during therapy. The obtained data led us to hypothesize on the mechanisms of regulation of signalling propensity of two pathways that are responsible for proliferation and survival of CLL cells, namely B Cell Receptor (BCR) signalling and signals from T-cells mediated by CD40/IL4. In aim 1 we hypothesize that CD20 is one of the key proteins involved in CLL cell activation, and influences BCR and interleukin signalling (see figure). This has important therapeutic implication since CD20 is used as a therapeutic target for 20 years (rituximab), but its function in CLL/normal B cells is unknown. In aim 2 we hypothesize that miR-29 acts a key regulator of T-cell signalling from CD40 and down-stream NFkB activation (see figure). This represents the first example of miRNAs‘ role in the propensity of T-cell interaction, and could be also utilized therapeutically. In aim 3 we will integrate our data on microenvironmental signaling (aim 1+2) and develop a first mouse model for PDX that would allow stable engraftment of primary CLL cells. Currently, CLL is non-transplantable to any animal model which complicates studies of its biology.
Summary
B cell chronic lymphocytic leukemia (CLL) is the most frequent leukemia in adults. CLL cells are characterized by their universal dependency on pro-survival and pro-proliferative signals from immune niches. To achieve this they constantly re-circulate between blood and lymph nodes, which is inhibited by novel microenvironment-targeting therapies such as “BCR inhibitors”. We aim to reveal how the malignant B cells change the propensity of their signalling pathways in response to the different microenvironments such as peripheral blood vs lymph node to obtain the proliferative signals. This is of major relevance for CLL, but also transferable to the biology of some other B cell malignancies and/or normal B cells. We analyzed the “finger print” of microenvironmental interactions in many CLL samples at various times during the disease course or during therapy. The obtained data led us to hypothesize on the mechanisms of regulation of signalling propensity of two pathways that are responsible for proliferation and survival of CLL cells, namely B Cell Receptor (BCR) signalling and signals from T-cells mediated by CD40/IL4. In aim 1 we hypothesize that CD20 is one of the key proteins involved in CLL cell activation, and influences BCR and interleukin signalling (see figure). This has important therapeutic implication since CD20 is used as a therapeutic target for 20 years (rituximab), but its function in CLL/normal B cells is unknown. In aim 2 we hypothesize that miR-29 acts a key regulator of T-cell signalling from CD40 and down-stream NFkB activation (see figure). This represents the first example of miRNAs‘ role in the propensity of T-cell interaction, and could be also utilized therapeutically. In aim 3 we will integrate our data on microenvironmental signaling (aim 1+2) and develop a first mouse model for PDX that would allow stable engraftment of primary CLL cells. Currently, CLL is non-transplantable to any animal model which complicates studies of its biology.
Max ERC Funding
1 499 990 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym LIDA
Project "Light on the Dark: Probing Dark Matter, Dark Energy and Dark Ages"
Researcher (PI) Jean-Paul Kneib
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), PE9, ERC-2011-ADG_20110209
Summary "Despite impressive progress in cosmology over the last decade, our understanding of the universe is still limited particularly in some Dark Areas: Dark Matter, Dark Energy & Dark Ages, where progress is particularly difficult. I propose to build a focused research group in a unique environment at the crossroad of astrophysics, cosmology and fundamental physics. The goal is to shade new “Light on the DArk” (LIDA) using new analysis techniques and new observations coming from two complementary surveys.
The first survey will target the fifty most massive strong lensing galaxy clusters and will shade new lights: i) on the nature of the Dark Matter through constraints coming from: the detailed mass distribution, the importance of substructures, and possible measurement of Dark Matter particle decay or self-annihilation, ii) on the most distant galaxies and the identification of the nature of the sources participating in the cosmic-reionization and ending the Dark Ages, iii) on the nature of the Dark Energy, through the accurate modeling of numerous multiple images in the best lensing clusters.
Second, with wide field imaging and spectroscopic surveys of the galaxy distribution we will probe new cosmological tests: i) the combination of galaxy-galaxy weak lensing and galaxy clustering in the framework of the halo model, ii) the measurement of the angle distribution of galaxy pairs, and thanks to the huge volume probed by the BOSS survey, iii) the test of the isotropy principle at various location of the surveyed volume. Finally we will prepare for the next generation of cosmological surveys by conducting an emission line galaxies redshift survey (e-BOSS) similar as those planned by the future facilities such as BigBOSS and EUCLID.
The LIDA project can started now, and will benefit from: new data collected by both space and ground-based telescopes over the next five years, as well as an active and vibrant work location in one of the best ranked university in Europe."
Summary
"Despite impressive progress in cosmology over the last decade, our understanding of the universe is still limited particularly in some Dark Areas: Dark Matter, Dark Energy & Dark Ages, where progress is particularly difficult. I propose to build a focused research group in a unique environment at the crossroad of astrophysics, cosmology and fundamental physics. The goal is to shade new “Light on the DArk” (LIDA) using new analysis techniques and new observations coming from two complementary surveys.
The first survey will target the fifty most massive strong lensing galaxy clusters and will shade new lights: i) on the nature of the Dark Matter through constraints coming from: the detailed mass distribution, the importance of substructures, and possible measurement of Dark Matter particle decay or self-annihilation, ii) on the most distant galaxies and the identification of the nature of the sources participating in the cosmic-reionization and ending the Dark Ages, iii) on the nature of the Dark Energy, through the accurate modeling of numerous multiple images in the best lensing clusters.
Second, with wide field imaging and spectroscopic surveys of the galaxy distribution we will probe new cosmological tests: i) the combination of galaxy-galaxy weak lensing and galaxy clustering in the framework of the halo model, ii) the measurement of the angle distribution of galaxy pairs, and thanks to the huge volume probed by the BOSS survey, iii) the test of the isotropy principle at various location of the surveyed volume. Finally we will prepare for the next generation of cosmological surveys by conducting an emission line galaxies redshift survey (e-BOSS) similar as those planned by the future facilities such as BigBOSS and EUCLID.
The LIDA project can started now, and will benefit from: new data collected by both space and ground-based telescopes over the next five years, as well as an active and vibrant work location in one of the best ranked university in Europe."
Max ERC Funding
2 499 935 €
Duration
Start date: 2012-09-01, End date: 2017-08-31
Project acronym LifeWithoutInsulin
Project Metabolic actions of brain leptin receptors signaling in type 1 diabetes
Researcher (PI) Roberto Coppari
Host Institution (HI) UNIVERSITE DE GENEVE
Call Details Consolidator Grant (CoG), LS4, ERC-2013-CoG
Summary An established dogma is that insulin is absolutely required for survival. This notion has been supported by the fact that the sole life-saving intervention available to the millions affected by type 1 diabetes mellitus (T1DM; an illness caused by pancreatic β-cell loss and hence insulin deficiency) is insulin therapy. This treatment however does not restore normal metabolic homeostasis. In fact, the life-expectancy and -quality of T1DM people is worse compared to normal subjects. In part, this is due to challenging morbidities of T1DM, as for example heart disease and hypoglycemia, both of which are thought to be caused by insulin therapy itself. Indeed, owing to insulin’s lipogenic actions, this treatment likely contributes to the ectopic lipid deposition (i.e.: in non-adipose tissues) and extremely high incidence of coronary artery disease seen in T1DM subjects. Also, due to insulin’s potent, fast-acting, glycemia-lowering action, this therapy significantly increases the risk of hypoglycemia; a disabling and life threatening event. Because insulin therapy does not restore metabolic homeostasis in T1DM subjects, better intervention is urgently needed. To these ends, we and others have shown that the hyperglycemic and lethal consequences of insulin deficiency can be rescued by administration of the adipocyte-secreted hormone leptin. Not only these results challenge an established view, they also raise a fundamental biological and medical question: what are the mechanisms by which leptin improves hyperglycemia and permits survival in the context of insulin deficiency? This proposal aims at identifying the critical cellular and molecular components underlying the beneficial effects of leptin in the context of insulin deficiency. Once identified, manipulation of these components has the potential to improve life-expectancy and -quality of the millions affected by insulin deficiency (e.g.: T1DM and also some late-stage type 2 diabetics).
Summary
An established dogma is that insulin is absolutely required for survival. This notion has been supported by the fact that the sole life-saving intervention available to the millions affected by type 1 diabetes mellitus (T1DM; an illness caused by pancreatic β-cell loss and hence insulin deficiency) is insulin therapy. This treatment however does not restore normal metabolic homeostasis. In fact, the life-expectancy and -quality of T1DM people is worse compared to normal subjects. In part, this is due to challenging morbidities of T1DM, as for example heart disease and hypoglycemia, both of which are thought to be caused by insulin therapy itself. Indeed, owing to insulin’s lipogenic actions, this treatment likely contributes to the ectopic lipid deposition (i.e.: in non-adipose tissues) and extremely high incidence of coronary artery disease seen in T1DM subjects. Also, due to insulin’s potent, fast-acting, glycemia-lowering action, this therapy significantly increases the risk of hypoglycemia; a disabling and life threatening event. Because insulin therapy does not restore metabolic homeostasis in T1DM subjects, better intervention is urgently needed. To these ends, we and others have shown that the hyperglycemic and lethal consequences of insulin deficiency can be rescued by administration of the adipocyte-secreted hormone leptin. Not only these results challenge an established view, they also raise a fundamental biological and medical question: what are the mechanisms by which leptin improves hyperglycemia and permits survival in the context of insulin deficiency? This proposal aims at identifying the critical cellular and molecular components underlying the beneficial effects of leptin in the context of insulin deficiency. Once identified, manipulation of these components has the potential to improve life-expectancy and -quality of the millions affected by insulin deficiency (e.g.: T1DM and also some late-stage type 2 diabetics).
Max ERC Funding
1 999 500 €
Duration
Start date: 2014-06-01, End date: 2019-05-31
Project acronym LIGHTNING
Project Charge separation, lightning and radio emission in low-mass objects
Researcher (PI) Christiane Helling
Host Institution (HI) THE UNIVERSITY COURT OF THE UNIVERSITY OF ST ANDREWS
Call Details Starting Grant (StG), PE9, ERC-2010-StG_20091028
Summary This project will investigate the hypothesis that dust clouds are a major source of charge separation and discharge processes in very low mass, extrasolar objects like M-dwarfs, Brown-Dwarfs, and planets. The aim is to model charging, dust formation and sedimentation in dusty media to understand how the atmospheric ionisation mechanisms change at the border from stars to planets in the M-dwarf--Brown-Dwarf transition region where radio emission starst to exceed X-ray emission, and to investigate the physics and the occurrence of intra-cloud lightning outside our solar system. Lightning is suggested to have triggered the occurrence of life on Earth.
Dusty media are generally very common on Earth and in space, for example in volcano plumes that influence the local climate on Earth, on Mars where it blocks Mars-Rover's wheels, in dust-clouds in Brown Dwarfs and planets which determine their chemistry and their detectability, or in planet-forming disks. All have in common that dust of mixed composition is abundant in a turbulent environment in a variety of sizes. This project will perform a characterisation of dusty astrophysical plasma, systemically study charge separation processes and draw comparison to known scenarios in volcanos and Martian plasmas. The project determines stellar parameter and dust cloud characteristics (e.g. cloud height) for which dust cloud charging becomes important, and under which conditions lightning can occur. A charge conservation model will be coupled to a non-equilibrium chemistry to search for discharge-related molecules and for pre-biotic molecules that might occur during lightning. Applications to standard model atmospheres will be carried out to study the influence on the spectral energy distribution and the object's albedo. The long-term aim of this project is to solve the dust and charge conservation equations together with the magnetic field equations in order to study the development of radio emission in low-mass objects.
Summary
This project will investigate the hypothesis that dust clouds are a major source of charge separation and discharge processes in very low mass, extrasolar objects like M-dwarfs, Brown-Dwarfs, and planets. The aim is to model charging, dust formation and sedimentation in dusty media to understand how the atmospheric ionisation mechanisms change at the border from stars to planets in the M-dwarf--Brown-Dwarf transition region where radio emission starst to exceed X-ray emission, and to investigate the physics and the occurrence of intra-cloud lightning outside our solar system. Lightning is suggested to have triggered the occurrence of life on Earth.
Dusty media are generally very common on Earth and in space, for example in volcano plumes that influence the local climate on Earth, on Mars where it blocks Mars-Rover's wheels, in dust-clouds in Brown Dwarfs and planets which determine their chemistry and their detectability, or in planet-forming disks. All have in common that dust of mixed composition is abundant in a turbulent environment in a variety of sizes. This project will perform a characterisation of dusty astrophysical plasma, systemically study charge separation processes and draw comparison to known scenarios in volcanos and Martian plasmas. The project determines stellar parameter and dust cloud characteristics (e.g. cloud height) for which dust cloud charging becomes important, and under which conditions lightning can occur. A charge conservation model will be coupled to a non-equilibrium chemistry to search for discharge-related molecules and for pre-biotic molecules that might occur during lightning. Applications to standard model atmospheres will be carried out to study the influence on the spectral energy distribution and the object's albedo. The long-term aim of this project is to solve the dust and charge conservation equations together with the magnetic field equations in order to study the development of radio emission in low-mass objects.
Max ERC Funding
1 500 000 €
Duration
Start date: 2011-03-01, End date: 2017-02-28
Project acronym LIPIDARRAY
Project Development and application of global lipidomic arrays to inflammatory vascular disease
Researcher (PI) Valerie O'donnell
Host Institution (HI) CARDIFF UNIVERSITY
Call Details Advanced Grant (AdG), LS4, ERC-2013-ADG
Summary How lipids are regulated on a global scale during vascular inflammation is not known. Thus, a major challenge exists to describe and catalog the total lipidome, in particular enabling the identification of new biologically active lipids, and description of changes. This is analogous to ‘omics’ of DNA, RNA and protein, but instead describing diversity of lipids in tissue samples. Importantly, this would encompass not only knowns, but also the vast number of unknowns that have not yet been catalogued in any study so far. Here, new systems biology approaches that can be applied to many other diseases or samples, and integrated with transcriptomic or proteomic analyses will be developed. These would be used to characterize the global lipidome during differentiation of immune cells, and in ex vivo samples from genomically-characterized inflammatory vascular disease. I hypothesize that development and application of “global lipidomic arrays” will define how lipids are regulated during vascular cell differentiation and inflammation, will identify new markers, and open up new therapeutic strategies.
These aims go beyond the current state of the art, and will be achieved by the following objectives that include novel interdisciplinary concepts and approaches:
1. Develop analytical methodologies using Fourier transform mass spectrometry and bioinformatics.
2. Develop approaches for structural identification, using high resolution MSn, high sensitivity NMR, and new computational methodologies.
3. Define the size and diversity of the mammalian cellular lipidome in human platelets (validation).
4. Characterize the global lipidome in (i) monocytes during differentiation from stem /yolk cells to resident, inflammatory or foam cells, (ii) plasma from samples genomically characterized for 14 separate risk alleles for cardiovascular and Alzheimer’s disease.
5. Develop an open access web-based resource for storage and curation of the results to allow others to mine the data.
Summary
How lipids are regulated on a global scale during vascular inflammation is not known. Thus, a major challenge exists to describe and catalog the total lipidome, in particular enabling the identification of new biologically active lipids, and description of changes. This is analogous to ‘omics’ of DNA, RNA and protein, but instead describing diversity of lipids in tissue samples. Importantly, this would encompass not only knowns, but also the vast number of unknowns that have not yet been catalogued in any study so far. Here, new systems biology approaches that can be applied to many other diseases or samples, and integrated with transcriptomic or proteomic analyses will be developed. These would be used to characterize the global lipidome during differentiation of immune cells, and in ex vivo samples from genomically-characterized inflammatory vascular disease. I hypothesize that development and application of “global lipidomic arrays” will define how lipids are regulated during vascular cell differentiation and inflammation, will identify new markers, and open up new therapeutic strategies.
These aims go beyond the current state of the art, and will be achieved by the following objectives that include novel interdisciplinary concepts and approaches:
1. Develop analytical methodologies using Fourier transform mass spectrometry and bioinformatics.
2. Develop approaches for structural identification, using high resolution MSn, high sensitivity NMR, and new computational methodologies.
3. Define the size and diversity of the mammalian cellular lipidome in human platelets (validation).
4. Characterize the global lipidome in (i) monocytes during differentiation from stem /yolk cells to resident, inflammatory or foam cells, (ii) plasma from samples genomically characterized for 14 separate risk alleles for cardiovascular and Alzheimer’s disease.
5. Develop an open access web-based resource for storage and curation of the results to allow others to mine the data.
Max ERC Funding
2 969 345 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym LIPIDOLIV
Project Role of the transcription factor ChREBP and its associated proteins in the development and progression of NAFLD
Researcher (PI) Renaud, Stéphane Dentin
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS4, ERC-2013-StG
Summary "Changes in lifestyle have resulted in a dramatic epidemic of type 2 diabetes and obesity. Among associated complications, Nonalcoholic Fatty Liver Disease (NAFLD) is emerging as the most common chronic liver disease and is gaining increasing recognition as a component of the epidemic of obesity. NAFLD is generally asymptomatic, although a minority of patients may present progressive liver injury with complications of Nonalcoholic steatohepatitis (NASH), cirrhosis and HCC. Excessive accumulation of fatty acids (FA) stored as triglycerides (TGs) in hepatocytes is the hallmark of NAFLD, which is strongly associated with insulin resistance (IR). However, despite the existing correlation between fatty liver and insulin resistance, it remains unclear whether insulin resistance causes the excessive accumulation of TG in the liver, or whether the increase in TG itself or other lipid intermediates such as diacylglycerols (DAG) and/or ceramides may trigger the development of hepatic or systemic insulin resistance. While some studies support the concept that intrahepatic accumulation of lipids precedes insulin resistance, others suggest that hepatic TG may in fact protect the liver from lipotoxicity by buffering the accumulation of FA. Such discrepancy might be explained since different pools of lipids exist within cells and only certain pools regulate insulin signaling. Consistent with such hypothesis, recent results by our group strongly support that specific FA species may influence hepatic TG storage, insulin signaling and/or inflammation. Therefore, the global aim of our proposal is to better understand the regulation of hepatic fatty acid synthesis and partitioning in addition to its impact on the detailed lipid profile. Using state-of-art technology and key genetically modified mouse models combined with original nutritional approaches and lipidomic analysis, our project aims at providing new information on the molecular basis of the pathogenesis of NAFLD."
Summary
"Changes in lifestyle have resulted in a dramatic epidemic of type 2 diabetes and obesity. Among associated complications, Nonalcoholic Fatty Liver Disease (NAFLD) is emerging as the most common chronic liver disease and is gaining increasing recognition as a component of the epidemic of obesity. NAFLD is generally asymptomatic, although a minority of patients may present progressive liver injury with complications of Nonalcoholic steatohepatitis (NASH), cirrhosis and HCC. Excessive accumulation of fatty acids (FA) stored as triglycerides (TGs) in hepatocytes is the hallmark of NAFLD, which is strongly associated with insulin resistance (IR). However, despite the existing correlation between fatty liver and insulin resistance, it remains unclear whether insulin resistance causes the excessive accumulation of TG in the liver, or whether the increase in TG itself or other lipid intermediates such as diacylglycerols (DAG) and/or ceramides may trigger the development of hepatic or systemic insulin resistance. While some studies support the concept that intrahepatic accumulation of lipids precedes insulin resistance, others suggest that hepatic TG may in fact protect the liver from lipotoxicity by buffering the accumulation of FA. Such discrepancy might be explained since different pools of lipids exist within cells and only certain pools regulate insulin signaling. Consistent with such hypothesis, recent results by our group strongly support that specific FA species may influence hepatic TG storage, insulin signaling and/or inflammation. Therefore, the global aim of our proposal is to better understand the regulation of hepatic fatty acid synthesis and partitioning in addition to its impact on the detailed lipid profile. Using state-of-art technology and key genetically modified mouse models combined with original nutritional approaches and lipidomic analysis, our project aims at providing new information on the molecular basis of the pathogenesis of NAFLD."
Max ERC Funding
1 500 000 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym LIPINTOECTION
Project Lipoproteins and angiogenesis: A new look at an old problem
Researcher (PI) Karina Yaniv
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS4, ERC-2013-StG
Summary Cardiovascular diseases (CVDs) are the leading cause of death throughout the world. Elevated Low Density Lipoprotein (LDL) is a well-known cardiovascular risk factor and endothelial (ECs)-lipoprotein (LIPs) interactions underlie the initiation and progression of atherogenesis, thrombosis and other CVDs.
The endothelium is a monolayer of cells that segregates the vascular contents from adjacent tissues. In spite of being continually exposed to LIPs, ECs were long thought of as inert barriers, through which lipids are exchanged between plasma and surrounding tissues. In contrast to this view, recent work from my laboratory has uncovered a deleterious role of ApoB-LIPs as direct inhibitors of angiogenesis in the developing embryo. These findings present only the tip of the iceberg, and the underlying cellular and molecular mechanisms are largely unknown. In this proposal, we will tackle this question by undertaking three independent but complementary approaches, aimed at characterizing LIP-EC interactions at a different level: cellular, molecular or pathological. We will explore these facets in zebrafish and mammals, utilizing live imaging, sophisticated lipidomic and biochemical analyses, as well as tumor xenografts and genetic mutants. An important and unique aspect of our approach is the focus on in vivo dynamics, in contrast to the extensive body of literature on LIP effects on cultured ECs. When completed this proposal will have shed light on a little explored, but critical aspect in the etiology of CVDs. Furthermore it will provide answers to important unresolved questions: What are the signaling pathways activated in ECs upon LIP binding? How are LIPs transported within ECs? Does ApoB possess additional functions beyond that of cholesterol carrier? Can high LIPs levels inhibit tumor angiogenesis and metastasis? In a broader sense, a deeper understanding of the effects of LIPs on ECs will be valuable for identifying new targets for therapeutic intervention.
Summary
Cardiovascular diseases (CVDs) are the leading cause of death throughout the world. Elevated Low Density Lipoprotein (LDL) is a well-known cardiovascular risk factor and endothelial (ECs)-lipoprotein (LIPs) interactions underlie the initiation and progression of atherogenesis, thrombosis and other CVDs.
The endothelium is a monolayer of cells that segregates the vascular contents from adjacent tissues. In spite of being continually exposed to LIPs, ECs were long thought of as inert barriers, through which lipids are exchanged between plasma and surrounding tissues. In contrast to this view, recent work from my laboratory has uncovered a deleterious role of ApoB-LIPs as direct inhibitors of angiogenesis in the developing embryo. These findings present only the tip of the iceberg, and the underlying cellular and molecular mechanisms are largely unknown. In this proposal, we will tackle this question by undertaking three independent but complementary approaches, aimed at characterizing LIP-EC interactions at a different level: cellular, molecular or pathological. We will explore these facets in zebrafish and mammals, utilizing live imaging, sophisticated lipidomic and biochemical analyses, as well as tumor xenografts and genetic mutants. An important and unique aspect of our approach is the focus on in vivo dynamics, in contrast to the extensive body of literature on LIP effects on cultured ECs. When completed this proposal will have shed light on a little explored, but critical aspect in the etiology of CVDs. Furthermore it will provide answers to important unresolved questions: What are the signaling pathways activated in ECs upon LIP binding? How are LIPs transported within ECs? Does ApoB possess additional functions beyond that of cholesterol carrier? Can high LIPs levels inhibit tumor angiogenesis and metastasis? In a broader sense, a deeper understanding of the effects of LIPs on ECs will be valuable for identifying new targets for therapeutic intervention.
Max ERC Funding
1 399 970 €
Duration
Start date: 2013-12-01, End date: 2018-11-30
Project acronym LipoCheX
Project The Role of Lipolysis in the Pathogenesis of
Cancer-associated Cachexia
Researcher (PI) Rudolf Zechner
Host Institution (HI) UNIVERSITAET GRAZ
Call Details Advanced Grant (AdG), LS4, ERC-2013-ADG
Summary Cachexia is a complex syndrome characterized by massive loss of body weight due to uncontrolled loss of adipose tissue and skeletal muscle. The wasting occurs during late stages of many unrelated chronic diseases and frequently leads to the death of affected individuals. Cachexia is most common in cancer, where an estimated 25% of patients die from cancer-associated cachexia (CAC) rather than from the cancer. Despite the tremendous impact of CAC on morbidity and mortality, the underlying molecular mechanisms are poorly understood.
Recently, we demonstrated that lipase-catalyzed triacylglycerol (TG) catabolism is required for the pathogenesis of CAC. Mice lacking adipose triglyceride lipase, the rate-limiting enzyme for TG hydrolysis (lipolysis), were completely protected from loss of both adipose tissue and muscle in two forms of cancer. This implies an essential role of the lipolytic process in the pathogenesis of CAC. Here we propose to elucidate the causal role of lipases and their coregulators in CAC development. We will determine mechanisms involved and pursue novel treatment strategies.
Our objectives are to:
- Investigate how different cancers in mice regulate tissue-specific lipolysis;
- Elucidate the functional role of lipases and their coregulators in the pathogenesis of CAC;
- Assess whether pharmacological inhibition of specific lipases prevents or delays CAC;
- Study the effects of cancer-induced lipolysis on energy dissipating pathways and epigenetic control.
The project enters a largely unexplored field: the role of lipid metabolism in the pathogenesis of CAC. The work will heavily rely on the characterization of induced mutant mouse models with CAC and require extensive collaboration with experts in pathology and large-scale systems analytics. The results are expected to yield new mechanisms of disease development and provide novel therapeutic targets to prevent the devastating and prevalent consequences of CAC.
Summary
Cachexia is a complex syndrome characterized by massive loss of body weight due to uncontrolled loss of adipose tissue and skeletal muscle. The wasting occurs during late stages of many unrelated chronic diseases and frequently leads to the death of affected individuals. Cachexia is most common in cancer, where an estimated 25% of patients die from cancer-associated cachexia (CAC) rather than from the cancer. Despite the tremendous impact of CAC on morbidity and mortality, the underlying molecular mechanisms are poorly understood.
Recently, we demonstrated that lipase-catalyzed triacylglycerol (TG) catabolism is required for the pathogenesis of CAC. Mice lacking adipose triglyceride lipase, the rate-limiting enzyme for TG hydrolysis (lipolysis), were completely protected from loss of both adipose tissue and muscle in two forms of cancer. This implies an essential role of the lipolytic process in the pathogenesis of CAC. Here we propose to elucidate the causal role of lipases and their coregulators in CAC development. We will determine mechanisms involved and pursue novel treatment strategies.
Our objectives are to:
- Investigate how different cancers in mice regulate tissue-specific lipolysis;
- Elucidate the functional role of lipases and their coregulators in the pathogenesis of CAC;
- Assess whether pharmacological inhibition of specific lipases prevents or delays CAC;
- Study the effects of cancer-induced lipolysis on energy dissipating pathways and epigenetic control.
The project enters a largely unexplored field: the role of lipid metabolism in the pathogenesis of CAC. The work will heavily rely on the characterization of induced mutant mouse models with CAC and require extensive collaboration with experts in pathology and large-scale systems analytics. The results are expected to yield new mechanisms of disease development and provide novel therapeutic targets to prevent the devastating and prevalent consequences of CAC.
Max ERC Funding
2 499 446 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym LIPOMET
Project Dietary Influences on Metastasis: How, When, and Why
Researcher (PI) Salvador Aznar Benitah
Host Institution (HI) FUNDACIO INSTITUT DE RECERCA BIOMEDICA (IRB BARCELONA)
Call Details Advanced Grant (AdG), LS4, ERC-2017-ADG
Summary We have recently identified metastasis-initiating cells (MICs) in several types of tumors (Nature, 2017)1.
Intriguingly, MICs: (i) are exclusive in their ability to generate metastases when transplanted; (ii) express the
fatty acid channel CD36 and have a unique lipid metabolic signature; (iii) are exquisitely sensitive to the
levels of fat in circulation, thus providing a link between the predisposition of metastasis and dietary fat; (iv)
are highly sensitive to CD36 inhibition, which almost completely abolishes their metastatic potential.
We still do not know how MICs promote metastasis or how MICs are influenced by dietary fat. In
particular: (A) where are MICs located within the tumor, and does this location influence their behavior?
How and where do they attach and expand at metastatic sites? (B) Why are MICs so sensitive to specific
dietary lipids, and how do these lipids promote metastasis at the molecular and cellular levels? (C) Is the
prolonged consumption of a high-fat diet a risk factor for developing metastatic tumors? If so, what are the
underlying genetic and epigenetic causes for this effect? Can we revert these causes?
To answer these questions, we will combine state-of-the-art in vivo functional models of metastasis, with
quantitative metabolomics and proteomics, epigenetic and geographical position (3D) single-cell
transcriptomic studies, as well as integrative computational analyses, using preclinical models and patientderived
carcinomas of melanoma, oral cancer and breast cancer.
We expect our project to provide fundamental insights into the mechanisms of metastasis, and how they are
influenced by diet. This is highly relevant as 1) large quantities of fatty acids are typically consumed in
Western diets; and 2) metastasis is the leading cause of cancer-related deaths. We also tackle a timely
medical unmet need by exploring the therapeutic anti-metastatic potential of targeting fatty acid metabolism
in cancer patients.
Summary
We have recently identified metastasis-initiating cells (MICs) in several types of tumors (Nature, 2017)1.
Intriguingly, MICs: (i) are exclusive in their ability to generate metastases when transplanted; (ii) express the
fatty acid channel CD36 and have a unique lipid metabolic signature; (iii) are exquisitely sensitive to the
levels of fat in circulation, thus providing a link between the predisposition of metastasis and dietary fat; (iv)
are highly sensitive to CD36 inhibition, which almost completely abolishes their metastatic potential.
We still do not know how MICs promote metastasis or how MICs are influenced by dietary fat. In
particular: (A) where are MICs located within the tumor, and does this location influence their behavior?
How and where do they attach and expand at metastatic sites? (B) Why are MICs so sensitive to specific
dietary lipids, and how do these lipids promote metastasis at the molecular and cellular levels? (C) Is the
prolonged consumption of a high-fat diet a risk factor for developing metastatic tumors? If so, what are the
underlying genetic and epigenetic causes for this effect? Can we revert these causes?
To answer these questions, we will combine state-of-the-art in vivo functional models of metastasis, with
quantitative metabolomics and proteomics, epigenetic and geographical position (3D) single-cell
transcriptomic studies, as well as integrative computational analyses, using preclinical models and patientderived
carcinomas of melanoma, oral cancer and breast cancer.
We expect our project to provide fundamental insights into the mechanisms of metastasis, and how they are
influenced by diet. This is highly relevant as 1) large quantities of fatty acids are typically consumed in
Western diets; and 2) metastasis is the leading cause of cancer-related deaths. We also tackle a timely
medical unmet need by exploring the therapeutic anti-metastatic potential of targeting fatty acid metabolism
in cancer patients.
Max ERC Funding
2 370 625 €
Duration
Start date: 2018-08-01, End date: 2023-07-31
Project acronym LITHIUM
Project From planetary birth with aperture masking interferometry to nulling with Lithium Niobate technology
Researcher (PI) Sylvestre Mathieu André Lacour
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Starting Grant (StG), PE9, ERC-2014-STG
Summary Observing the process of planetary accretion is crucial to inform models of planet formation. Most of the key action is expected to happen in the gaps of protostellar disks – a spatial realm over which aperture masking interferometry has demonstrated a unique ability to deliver incisive imaging. Masking offers twin advantages of higher dynamic range at the diffraction limit (lambda/D) than differential imaging, while at the same time giving nearly complete Fourier coverage compared to long baseline interferometry. The founding objective of this proposal is to create expertise and technology to understand the astrophysical phenomena so far only glimpsed in faint detections in stellar gaps such as those published in T Cha (Huelamo et al. 2011), HD142527 (Biller et al. 2012) and FL Cha (Cieza et al. 2013). But the central goal of this project is to further advance the experimental technique. Reaching even higher dynamic range for fainter detections is essential for probing planetary birth. The way to improve the dynamic range is clear: increase the accuracy of the primary closure phase observable. To do so, we will follow two paths. The first will use laboratory experimentations to analyse and understand the sources of bias to the closure phase. The resulting end-product will be better software offered to the community, and better techniques for a next generation of aperture masking devices. The second path is to amplify the closure phase signal by combining nulling with closure phase (Lacour et al. 2014). This second path is the most challenging, but will be an important breakthrough to the field. Nulling is to aperture masking what coronagraphy is to classical imaging. Without a first level of nulling, the aperture masking technique will always be limited by the photon noise due to the stellar light. We propose to build on our experience of Lithium Niobate integrated optics devices to bring aperture masking to a new level of performance in high dynamic range imaging.
Summary
Observing the process of planetary accretion is crucial to inform models of planet formation. Most of the key action is expected to happen in the gaps of protostellar disks – a spatial realm over which aperture masking interferometry has demonstrated a unique ability to deliver incisive imaging. Masking offers twin advantages of higher dynamic range at the diffraction limit (lambda/D) than differential imaging, while at the same time giving nearly complete Fourier coverage compared to long baseline interferometry. The founding objective of this proposal is to create expertise and technology to understand the astrophysical phenomena so far only glimpsed in faint detections in stellar gaps such as those published in T Cha (Huelamo et al. 2011), HD142527 (Biller et al. 2012) and FL Cha (Cieza et al. 2013). But the central goal of this project is to further advance the experimental technique. Reaching even higher dynamic range for fainter detections is essential for probing planetary birth. The way to improve the dynamic range is clear: increase the accuracy of the primary closure phase observable. To do so, we will follow two paths. The first will use laboratory experimentations to analyse and understand the sources of bias to the closure phase. The resulting end-product will be better software offered to the community, and better techniques for a next generation of aperture masking devices. The second path is to amplify the closure phase signal by combining nulling with closure phase (Lacour et al. 2014). This second path is the most challenging, but will be an important breakthrough to the field. Nulling is to aperture masking what coronagraphy is to classical imaging. Without a first level of nulling, the aperture masking technique will always be limited by the photon noise due to the stellar light. We propose to build on our experience of Lithium Niobate integrated optics devices to bring aperture masking to a new level of performance in high dynamic range imaging.
Max ERC Funding
1 851 881 €
Duration
Start date: 2015-03-01, End date: 2020-02-29
Project acronym LIVERCANCERMECHANISM
Project Uncovering the mechanisms of inflammation induced liver tissue destruction and carcinogenesis
Researcher (PI) Mathias Heikenwaelder
Host Institution (HI) HELMHOLTZ ZENTRUM MUENCHEN DEUTSCHES FORSCHUNGSZENTRUM FUER GESUNDHEIT UND UMWELT GMBH
Call Details Starting Grant (StG), LS4, ERC-2010-StG_20091118
Summary Hepatocellular carcinoma (HCC) is caused by chronic hepatitis and is the third most common cause of cancer-related death worldwide, with a rising incidence in first world countries. To date no effective therapies other than liver transplantation are available for this disease.
Previous studies have provided evidence that inflammatory signalling pathways (e.g. the NF-b pathway) are crucial modulators of liver cancer development. However, the exact mechanisms driving hepatitis-induced liver damage and cancer formation remain elusive. Among others, aberrant expression of cytotoxic cytokines is thought to be critically involved.
We have recently shown that the inflammatory cytokines lymphotoxin (LT) and are specifically upregulated in livers of patients suffering from hepatitis C and B virus-induced liver inflammation or HCC and that liver specific expression of LT and in mice (AlbLT) suffices to induce inflammation-induced liver cancer development. We could further demonstrate that this depended on the presence of functional lymphocytes.
My proposal is pillared by three main approaches that all aim to elucidate the exact cellular and molecular mechanisms underlying chronic liver damage and HCC development in humans as well as in mouse models of inflammation- or carcinogen-induced liver cancer.
First, we will identify the particular immune cell type(s) (e.g. B- or T-lymphocytes; macrophages; NK-T cells) involved in HCC development. Although inflammatory signalling and immune cells appear to be important in HCC development it remains elusive, which immune cell type(s) contribute to inflammation induced liver cancer development.
Secondly, we will investigate how inflammatory signalling pathways induce chromosomal aberrations. It is known that inflammatory signalling cascades cause chromosomal aberrations; however, the detailed mechanisms by which this occurs are not fully understood. Additionally, we will determine how inflammatory signalling influences the pathways involved in DNA repair, replication and chromosomal segregation culminating in chromosomal aberrations and cancer.
Finally, we will examine the role of oval cells in liver cancer formation. Oval cells, which are putative liver-cancer stem cells, differentiate into either hepatocytes or cholangiocytes, proliferate under inflammatory conditions, and are found within HCC. However, their exact functional role in liver carcinogenesis is unknown. We will biochemically characterize proliferating and HCC-associated ovals cells in mouse models of inflammation-induced HCC and in diseased human liver tissues. This will pave the way for the development of the first genetic tools to deplete or express genes in an oval cell-specific manner.
The new scientific knowledge gained by these studies investigating how immune cells and inflammatory signalling induce chronic liver damage and cancer on a mechanistic level, and how oval cells contribute to HCC will provide the basis for future novel pharmacological approaches to treating inflammatory liver diseases and HCC in humans.
Summary
Hepatocellular carcinoma (HCC) is caused by chronic hepatitis and is the third most common cause of cancer-related death worldwide, with a rising incidence in first world countries. To date no effective therapies other than liver transplantation are available for this disease.
Previous studies have provided evidence that inflammatory signalling pathways (e.g. the NF-b pathway) are crucial modulators of liver cancer development. However, the exact mechanisms driving hepatitis-induced liver damage and cancer formation remain elusive. Among others, aberrant expression of cytotoxic cytokines is thought to be critically involved.
We have recently shown that the inflammatory cytokines lymphotoxin (LT) and are specifically upregulated in livers of patients suffering from hepatitis C and B virus-induced liver inflammation or HCC and that liver specific expression of LT and in mice (AlbLT) suffices to induce inflammation-induced liver cancer development. We could further demonstrate that this depended on the presence of functional lymphocytes.
My proposal is pillared by three main approaches that all aim to elucidate the exact cellular and molecular mechanisms underlying chronic liver damage and HCC development in humans as well as in mouse models of inflammation- or carcinogen-induced liver cancer.
First, we will identify the particular immune cell type(s) (e.g. B- or T-lymphocytes; macrophages; NK-T cells) involved in HCC development. Although inflammatory signalling and immune cells appear to be important in HCC development it remains elusive, which immune cell type(s) contribute to inflammation induced liver cancer development.
Secondly, we will investigate how inflammatory signalling pathways induce chromosomal aberrations. It is known that inflammatory signalling cascades cause chromosomal aberrations; however, the detailed mechanisms by which this occurs are not fully understood. Additionally, we will determine how inflammatory signalling influences the pathways involved in DNA repair, replication and chromosomal segregation culminating in chromosomal aberrations and cancer.
Finally, we will examine the role of oval cells in liver cancer formation. Oval cells, which are putative liver-cancer stem cells, differentiate into either hepatocytes or cholangiocytes, proliferate under inflammatory conditions, and are found within HCC. However, their exact functional role in liver carcinogenesis is unknown. We will biochemically characterize proliferating and HCC-associated ovals cells in mouse models of inflammation-induced HCC and in diseased human liver tissues. This will pave the way for the development of the first genetic tools to deplete or express genes in an oval cell-specific manner.
The new scientific knowledge gained by these studies investigating how immune cells and inflammatory signalling induce chronic liver damage and cancer on a mechanistic level, and how oval cells contribute to HCC will provide the basis for future novel pharmacological approaches to treating inflammatory liver diseases and HCC in humans.
Max ERC Funding
1 212 190 €
Duration
Start date: 2010-10-01, End date: 2015-09-30
Project acronym LOCALSTAR
Project Modelling star formation in the local universe
Researcher (PI) Clare Dobbs
Host Institution (HI) THE UNIVERSITY OF EXETER
Call Details Starting Grant (StG), PE9, ERC-2011-StG_20101014
Summary The goal of this proposal is to revolutionize our understanding of star formation in nearby galaxies, using numerical simulations. Traditionally, research in star formation has considered the contraction of a giant molecular cloud (GMC), or more commonly a star forming core, under gravity. However there has been relatively little research on molecular clouds themselves, even though they provide the initial conditions for star formation, and thus determine the main assumptions for theories of star formation. The proposed research will focus on the scales of giant molecular clouds, the clouds of molecular hydrogen (H2) where most star formation takes place in nearby galaxies. These objects link galactic scale physics with the small scale physics of star formation. Only now are the computational resources becoming available to study the interstellar medium (ISM) numerically on galactic scales, and model the complex processes involved in GMC and star formation. Simultaneously observational programs (e.g. ALMA, Herschel, CARMA) are starting to resolve GMCs in nearby galaxies. Our research will involve performing calculations on scales from individual GMCs to interacting galaxies, and comparing to forthcoming observations to answer some of the most fundamental questions in star formation, such as why star formation is inefficient, how do GMCs form and what are their lifetimes.
Summary
The goal of this proposal is to revolutionize our understanding of star formation in nearby galaxies, using numerical simulations. Traditionally, research in star formation has considered the contraction of a giant molecular cloud (GMC), or more commonly a star forming core, under gravity. However there has been relatively little research on molecular clouds themselves, even though they provide the initial conditions for star formation, and thus determine the main assumptions for theories of star formation. The proposed research will focus on the scales of giant molecular clouds, the clouds of molecular hydrogen (H2) where most star formation takes place in nearby galaxies. These objects link galactic scale physics with the small scale physics of star formation. Only now are the computational resources becoming available to study the interstellar medium (ISM) numerically on galactic scales, and model the complex processes involved in GMC and star formation. Simultaneously observational programs (e.g. ALMA, Herschel, CARMA) are starting to resolve GMCs in nearby galaxies. Our research will involve performing calculations on scales from individual GMCs to interacting galaxies, and comparing to forthcoming observations to answer some of the most fundamental questions in star formation, such as why star formation is inefficient, how do GMCs form and what are their lifetimes.
Max ERC Funding
1 169 586 €
Duration
Start date: 2011-11-01, End date: 2017-05-31
Project acronym LOCATE
Project "Locomotion, hunting and habitat utilisation among large African carnivores and their prey"
Researcher (PI) Alan Martin Wilson
Host Institution (HI) THE ROYAL VETERINARY COLLEGE
Call Details Advanced Grant (AdG), LS4, ERC-2012-ADG_20120314
Summary "I propose to make the first detailed measurements of the mechanics and energetic cost of locomotor activity including hunting of five large terrestrial carnivore species and their five predominant prey on the southern African savannah. We will refine and use our own innovative design of dynamically responsive tracking collar with high accuracy GPS, accelerometers, gyroscopes, and solar charged batteries to capture round the clock activity, manoeuvring and agility. We will use a geopointing camera gimble on aircraft and UAV to track collared animals and record locomotion of adjacent uncollared animals to obtain high resolution aerial video of hunting. Hunts will be overlaid on a terrain and vegetation map generated using full waveform LiDAR laser scanning and aerial photography to understand the impact of vegetation type, density and surface profile (camber and incline) on locomotion and hunt outcome. We will make detailed measurements of musculoskeletal anatomy and muscle physiology to determine tradeoffs between power and economy for each group and the extent to which hunt performance and outcome can be simply attributed to musculoskeletal physiology and fatigue in the different species.
The technological innovation and integration, its application to wild animals and the massive potential for its application in field biology is all ground breaking. This would be the first investigation of hunting dynamics where every hunt, day and night of a predator is captured and analysed along with prey dynamics (in a subset) to understand what environmental factors influence outcome. It will be the first study to evaluate the extreme dynamics of highly motivated non-domestic species, their anatomy and muscle physiology and the locomotor determinants of successful and unsuccessful hunts. These data and the detailed tracking data will enable studies of terrain utilisation, intra- and inter-species conflict and the impact of vegetation change on species success and survival."
Summary
"I propose to make the first detailed measurements of the mechanics and energetic cost of locomotor activity including hunting of five large terrestrial carnivore species and their five predominant prey on the southern African savannah. We will refine and use our own innovative design of dynamically responsive tracking collar with high accuracy GPS, accelerometers, gyroscopes, and solar charged batteries to capture round the clock activity, manoeuvring and agility. We will use a geopointing camera gimble on aircraft and UAV to track collared animals and record locomotion of adjacent uncollared animals to obtain high resolution aerial video of hunting. Hunts will be overlaid on a terrain and vegetation map generated using full waveform LiDAR laser scanning and aerial photography to understand the impact of vegetation type, density and surface profile (camber and incline) on locomotion and hunt outcome. We will make detailed measurements of musculoskeletal anatomy and muscle physiology to determine tradeoffs between power and economy for each group and the extent to which hunt performance and outcome can be simply attributed to musculoskeletal physiology and fatigue in the different species.
The technological innovation and integration, its application to wild animals and the massive potential for its application in field biology is all ground breaking. This would be the first investigation of hunting dynamics where every hunt, day and night of a predator is captured and analysed along with prey dynamics (in a subset) to understand what environmental factors influence outcome. It will be the first study to evaluate the extreme dynamics of highly motivated non-domestic species, their anatomy and muscle physiology and the locomotor determinants of successful and unsuccessful hunts. These data and the detailed tracking data will enable studies of terrain utilisation, intra- and inter-species conflict and the impact of vegetation change on species success and survival."
Max ERC Funding
3 079 643 €
Duration
Start date: 2013-06-01, End date: 2018-05-31
Project acronym LODESTONE
Project LODESTONE: Unifying the Radio Spectrum to Map the Magnetic Universe
Researcher (PI) Anna Scaife
Host Institution (HI) THE UNIVERSITY OF MANCHESTER
Call Details Starting Grant (StG), PE9, ERC-2012-StG_20111012
Summary Clusters of galaxies are a unique cosmological probe. Beyond the potential wells of individual clusters the vast filamentary cosmic web which binds them together contains two thirds of the baryonic matter in the Universe and represents a rich scientific resource. In order to use such resources correctly it is essential to understand the energetics, dynamics and redshift dependence of the intra/inter-cluster media. Absolutely fundamental to this understanding is a detailed knowledge of the strength, morphology and evolution of their magnetic fields. In addition, using clusters of galaxies and cosmic large scale structure as a laboratory in which to probe these fields is also the key to unlocking the more fundamental long standing problems of magnetic evolution and structure, and ultimately to determining the very origins of cosmic magnetism itself. To tackle these fundamental questions I will use a combination of two different observational approaches, effectively unifying the radio spectrum from meter to millimeter wavelengths: Rotation Measure Synthesis and the resolved Sunyaev–Zel’dovich Effect. Individually these approaches both offer unique insights; combined they provide an almost complete picture of the internal baryonic dynamics of clusters and large scale structure. I will use a set of leading and next generation radio telescopes around the globe. These telescopes span the radio spectrum, but are united by my leading role in the magnetism and/or cluster science case for each one. I will build an infrastructure which combines their operations to achieve a scientific outcome much greater than the individual instruments can produce independently. This will include development of world leading image reconstruction techniques; novel approaches to data combining; and state of the art analysis methods - as well as building a program which integrate disparate communities in astronomy. This program is called LODESTONE, to reflect the aligning nature of magnetism.
Summary
Clusters of galaxies are a unique cosmological probe. Beyond the potential wells of individual clusters the vast filamentary cosmic web which binds them together contains two thirds of the baryonic matter in the Universe and represents a rich scientific resource. In order to use such resources correctly it is essential to understand the energetics, dynamics and redshift dependence of the intra/inter-cluster media. Absolutely fundamental to this understanding is a detailed knowledge of the strength, morphology and evolution of their magnetic fields. In addition, using clusters of galaxies and cosmic large scale structure as a laboratory in which to probe these fields is also the key to unlocking the more fundamental long standing problems of magnetic evolution and structure, and ultimately to determining the very origins of cosmic magnetism itself. To tackle these fundamental questions I will use a combination of two different observational approaches, effectively unifying the radio spectrum from meter to millimeter wavelengths: Rotation Measure Synthesis and the resolved Sunyaev–Zel’dovich Effect. Individually these approaches both offer unique insights; combined they provide an almost complete picture of the internal baryonic dynamics of clusters and large scale structure. I will use a set of leading and next generation radio telescopes around the globe. These telescopes span the radio spectrum, but are united by my leading role in the magnetism and/or cluster science case for each one. I will build an infrastructure which combines their operations to achieve a scientific outcome much greater than the individual instruments can produce independently. This will include development of world leading image reconstruction techniques; novel approaches to data combining; and state of the art analysis methods - as well as building a program which integrate disparate communities in astronomy. This program is called LODESTONE, to reflect the aligning nature of magnetism.
Max ERC Funding
1 928 369 €
Duration
Start date: 2013-01-01, End date: 2017-12-31
Project acronym LOFAR
Project Searching for The Origin of Cosmic Rays and Neutrinos with LOFAR
Researcher (PI) Stijn Buitink
Host Institution (HI) VRIJE UNIVERSITEIT BRUSSEL
Call Details Starting Grant (StG), PE9, ERC-2014-STG
Summary The origin of cosmic rays remains one of the largest mysteries in astrophysics. Innovative and accurate radio measurements of cosmic rays and neutrinos with LOFAR promise to provide new answers.
It is generally believed that ultra-high-energy cosmic rays are produced in extragalactic sources like gamma- ray bursts or active galactic nuclei, while the lower energy cosmic rays come from our own Galaxy. At what energy this transition takes place is still unknown. Here we focus on disentangling Galactic and extragalactic components by studying the mass composition between 10^17 and 10^18 eV, a regime that is also crucial for understanding the origin of the extraterrestrial neutrinos detected by IceCube.
We do this with LOFAR, the first radio telescope that can detect individual cosmic rays with hundreds of antennas. This incredible level of detail allowed us to finally understand the complicated radiation mechanism and to perform the first-ever accurate mass analysis based on radio measurements. Our first data reveal a strong proton component below 10^18 eV, suggesting an early transition to an extragalactic component. With upgrades to our detector and techniques we will be able to improve our sample size by an order of magnitude, resolve more mass components, and identify the origin of high-energy cosmic rays and neutrinos.
The technique may be scaled up to higher energies, measured at the Pierre Auger Observatory, where mass information is needed to correlate cosmic rays with their astrophysical sources and to confirm the nature of the cutoff at ~10^19.6 eV.
We can even search for particles beyond the GZK limit. With the Westerbork telescope we have already set the best limit on cosmic rays and neutrinos above 10^23 eV. With LOFAR we will achieve a much better sensitivity at lower energies, also probing for new physics, like the decays of cosmic strings predicted by supersymmetric theories.
Summary
The origin of cosmic rays remains one of the largest mysteries in astrophysics. Innovative and accurate radio measurements of cosmic rays and neutrinos with LOFAR promise to provide new answers.
It is generally believed that ultra-high-energy cosmic rays are produced in extragalactic sources like gamma- ray bursts or active galactic nuclei, while the lower energy cosmic rays come from our own Galaxy. At what energy this transition takes place is still unknown. Here we focus on disentangling Galactic and extragalactic components by studying the mass composition between 10^17 and 10^18 eV, a regime that is also crucial for understanding the origin of the extraterrestrial neutrinos detected by IceCube.
We do this with LOFAR, the first radio telescope that can detect individual cosmic rays with hundreds of antennas. This incredible level of detail allowed us to finally understand the complicated radiation mechanism and to perform the first-ever accurate mass analysis based on radio measurements. Our first data reveal a strong proton component below 10^18 eV, suggesting an early transition to an extragalactic component. With upgrades to our detector and techniques we will be able to improve our sample size by an order of magnitude, resolve more mass components, and identify the origin of high-energy cosmic rays and neutrinos.
The technique may be scaled up to higher energies, measured at the Pierre Auger Observatory, where mass information is needed to correlate cosmic rays with their astrophysical sources and to confirm the nature of the cutoff at ~10^19.6 eV.
We can even search for particles beyond the GZK limit. With the Westerbork telescope we have already set the best limit on cosmic rays and neutrinos above 10^23 eV. With LOFAR we will achieve a much better sensitivity at lower energies, also probing for new physics, like the decays of cosmic strings predicted by supersymmetric theories.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-06-01, End date: 2020-05-31
Project acronym LOFAR-AUGER
Project From Black Holes to Ultra-High Energy Cosmic Rays: Exploring the Extremes of the Universe with Low-Frequency Radio Interferometry
Researcher (PI) Heino Falcke
Host Institution (HI) STICHTING KATHOLIEKE UNIVERSITEIT
Call Details Advanced Grant (AdG), PE9, ERC-2008-AdG
Summary Black holes (BHs) and ultra-high energy cosmic rays (UHECRs) are two extremes of the universe that link particle physics and astrophysics. BHs are the most efficient power generators in the universe while UHECRs are the most energetic particles ever detected. As we showed previously, a major fraction of the power of BHs is channeled into radio-emitting plasma jets, which are also efficient particle accelerators. Are BHs also responsible for UHECRs? This long-standing question could be answered soon, through the dawn of cosmic ray astronomy. The giant Auger observatory has now shown for the first time that the arrival directions of UHECRs are non-isotropic, potentially pointing back to their sources of origin. BHs turned out to be major suspects, but other sources could still also be responsible. To address this conclusively and to establish cosmic ray astronomy as a productive new field in the coming years, we need to increase statistics, expand current observatories, and have complementary all-sky radio surveys available to identify sources, since radio emission traces particle acceleration sites. Here, techniques pioneered by the Low-Frequency Array (LOFAR) promise major advances. First of all, working on LOFAR we uncovered a new technique to detect UHECRs with radio antennas and verified it experimentally. The technique promises to increase the number of high-quality events by almost an order of magnitude and provides much improved energy and direction resolution. We now want to implement this technique in Auger, combining LOFAR and AUGER know-how. Secondly, LOFAR and soon other SKA pathfinders will significantly improve all-sky radio surveys with high sensitivity, resolution, and image quality. Hence, we will use LOFAR to understand the astrophysics of UHECR source candidates and compile a radio-based catalog thereof. We start with jets from BHs and move later to other sources. Together this will allow us to identify UHECR sources and study them in detail.
Summary
Black holes (BHs) and ultra-high energy cosmic rays (UHECRs) are two extremes of the universe that link particle physics and astrophysics. BHs are the most efficient power generators in the universe while UHECRs are the most energetic particles ever detected. As we showed previously, a major fraction of the power of BHs is channeled into radio-emitting plasma jets, which are also efficient particle accelerators. Are BHs also responsible for UHECRs? This long-standing question could be answered soon, through the dawn of cosmic ray astronomy. The giant Auger observatory has now shown for the first time that the arrival directions of UHECRs are non-isotropic, potentially pointing back to their sources of origin. BHs turned out to be major suspects, but other sources could still also be responsible. To address this conclusively and to establish cosmic ray astronomy as a productive new field in the coming years, we need to increase statistics, expand current observatories, and have complementary all-sky radio surveys available to identify sources, since radio emission traces particle acceleration sites. Here, techniques pioneered by the Low-Frequency Array (LOFAR) promise major advances. First of all, working on LOFAR we uncovered a new technique to detect UHECRs with radio antennas and verified it experimentally. The technique promises to increase the number of high-quality events by almost an order of magnitude and provides much improved energy and direction resolution. We now want to implement this technique in Auger, combining LOFAR and AUGER know-how. Secondly, LOFAR and soon other SKA pathfinders will significantly improve all-sky radio surveys with high sensitivity, resolution, and image quality. Hence, we will use LOFAR to understand the astrophysics of UHECR source candidates and compile a radio-based catalog thereof. We start with jets from BHs and move later to other sources. Together this will allow us to identify UHECR sources and study them in detail.
Max ERC Funding
3 460 000 €
Duration
Start date: 2009-01-01, End date: 2013-12-31
Project acronym LOFARCORE
Project Unravelling the Cosmic Reionization History
Researcher (PI) Antonius Gerardus De Bruyn
Host Institution (HI) STICHTING ASTRON, NETHERLANDS INSTITUTE FOR RADIO ASTRONOMY
Call Details Advanced Grant (AdG), PE9, ERC-2013-ADG
Summary "One of the frontier cosmological research themes of the past decade has been the Epoch of Reionization (EoR). This era marked the start of the formation of visible baryonic structures: stars, (dwarf-)galaxies, clusters and the cosmic web. Some top level questions surrounding COsmic REionization are: ""When did it start ?”, “How long did it take ?”, “What were the main ionizing sources ?”, and “What did the Universe look like during this crucial evolutionary phase ?"" Simulations of reionization suggest how it may have proceeded, but hard observational facts on the EoR are few and far between.
At the start of the EoR all hydrogen was cold and neutral. At redshift z=6 the transition to an ionized Universe was completed. The 21cm line, redshifted to much longer wavelength, is a key diagnostic during this phase. LOFAR, the European LOw Frequency Array, will be the premier instrument to explore this epoch, simultaneously over the full redshift range from z=6.5-11.5 ! The required angular resolution is also well matched to the 2.5 km CORE of LOFAR. Detecting redshifted HI from the EoR is generally acknowledged as the most difficult radio astronomical project ever attempted. As PI of the LOFAR EoR Key Science Project and as LOFAR Calibration Project scientist, I have worked hard for 10 years to prepare LOFAR and myself for this endeavour.
In November 2012 LOFAR commenced a long (> 3-5 years) observing campaign, generating several PB of data per year. A new phase, with extraordinary challenges, still lies ahead. ERC funding for the LOFARCORE team will permit me to ramp up the execution of this program, and in a timely manner (we have competition from US, Australian and Indian groups), solve problems along the way, and analyze and publish what will be ground-breaking results on the COsmic REionization history. The LOFAR EoR project will be the pathfinder for even more daunting future explorations, using SKA, of the phases preceeding the EoR, i.e. Cosmic Dawn."
Summary
"One of the frontier cosmological research themes of the past decade has been the Epoch of Reionization (EoR). This era marked the start of the formation of visible baryonic structures: stars, (dwarf-)galaxies, clusters and the cosmic web. Some top level questions surrounding COsmic REionization are: ""When did it start ?”, “How long did it take ?”, “What were the main ionizing sources ?”, and “What did the Universe look like during this crucial evolutionary phase ?"" Simulations of reionization suggest how it may have proceeded, but hard observational facts on the EoR are few and far between.
At the start of the EoR all hydrogen was cold and neutral. At redshift z=6 the transition to an ionized Universe was completed. The 21cm line, redshifted to much longer wavelength, is a key diagnostic during this phase. LOFAR, the European LOw Frequency Array, will be the premier instrument to explore this epoch, simultaneously over the full redshift range from z=6.5-11.5 ! The required angular resolution is also well matched to the 2.5 km CORE of LOFAR. Detecting redshifted HI from the EoR is generally acknowledged as the most difficult radio astronomical project ever attempted. As PI of the LOFAR EoR Key Science Project and as LOFAR Calibration Project scientist, I have worked hard for 10 years to prepare LOFAR and myself for this endeavour.
In November 2012 LOFAR commenced a long (> 3-5 years) observing campaign, generating several PB of data per year. A new phase, with extraordinary challenges, still lies ahead. ERC funding for the LOFARCORE team will permit me to ramp up the execution of this program, and in a timely manner (we have competition from US, Australian and Indian groups), solve problems along the way, and analyze and publish what will be ground-breaking results on the COsmic REionization history. The LOFAR EoR project will be the pathfinder for even more daunting future explorations, using SKA, of the phases preceeding the EoR, i.e. Cosmic Dawn."
Max ERC Funding
2 952 628 €
Duration
Start date: 2014-04-01, End date: 2018-06-30
Project acronym LONGHEART
Project Exploring selected long non-coding RNAs as diagnostics and therapeutic targets for heart failure
Researcher (PI) Thomas Thum
Host Institution (HI) MEDIZINISCHE HOCHSCHULE HANNOVER
Call Details Consolidator Grant (CoG), LS4, ERC-2014-CoG
Summary Despite clinical advances, diseases of the cardiovascular system are the most common cause of morbidity and mortality in the EU with currently 50 million people suffering from heart failure. These important challenges call for a better understanding of underlying mechanisms to enable development of innovative, effective diagnostic and therapeutic strategies for heart failure. Cardiac stress such as myocardial infarction or hypertension leads to cellular “remodeling” of the left ventricle resulting in heart failure. Protein-coding genes originate from only 1.5% of the genome, whereas the larger remaining portion is often transcribed to non-coding RNAs, of which functional importance is still ill understood. We pioneered a role of small microRNAs as diagnostics and therapeutic targets for heart failure (Nature, 2008; Nature Comm, 2012, J Clin Invest, 2014). We now will focus on the larger fraction of long non-coding RNAs (lncRNAs) and their functional roles, as well as diagnostic and therapeutic use in heart failure. The proposal has the following interconnected objectives: a) identify novel functional relevant cardiac remodeling-associated lncRNAs; b) characterise key lncRNA cardiac targetomes; c) investigate lncRNA-paracrine mechanisms and the diagnostic and prognostic potential of cardiac-derived extracellular lncRNAs using large clinical cohorts; and d) discover their therapeutic potential to prevent cardiac remodeling in clinically relevant animal models. Innovative molecular and cell-based methods, a unique lncRNA-shRNA library, genetic animal models and availability of large clinical biobanks will form the basis for a successful strategy. LONGHEART will lead to ground-breaking new insight into the role of lncRNAs in the heart. These findings will firmly establish lncRNA-based mechanisms to identify fundamentally novel diagnostic and therapeutic entry points for a most serious clinical important disorder in dire need for new diagnostic and therapeutic paradigms.
Summary
Despite clinical advances, diseases of the cardiovascular system are the most common cause of morbidity and mortality in the EU with currently 50 million people suffering from heart failure. These important challenges call for a better understanding of underlying mechanisms to enable development of innovative, effective diagnostic and therapeutic strategies for heart failure. Cardiac stress such as myocardial infarction or hypertension leads to cellular “remodeling” of the left ventricle resulting in heart failure. Protein-coding genes originate from only 1.5% of the genome, whereas the larger remaining portion is often transcribed to non-coding RNAs, of which functional importance is still ill understood. We pioneered a role of small microRNAs as diagnostics and therapeutic targets for heart failure (Nature, 2008; Nature Comm, 2012, J Clin Invest, 2014). We now will focus on the larger fraction of long non-coding RNAs (lncRNAs) and their functional roles, as well as diagnostic and therapeutic use in heart failure. The proposal has the following interconnected objectives: a) identify novel functional relevant cardiac remodeling-associated lncRNAs; b) characterise key lncRNA cardiac targetomes; c) investigate lncRNA-paracrine mechanisms and the diagnostic and prognostic potential of cardiac-derived extracellular lncRNAs using large clinical cohorts; and d) discover their therapeutic potential to prevent cardiac remodeling in clinically relevant animal models. Innovative molecular and cell-based methods, a unique lncRNA-shRNA library, genetic animal models and availability of large clinical biobanks will form the basis for a successful strategy. LONGHEART will lead to ground-breaking new insight into the role of lncRNAs in the heart. These findings will firmly establish lncRNA-based mechanisms to identify fundamentally novel diagnostic and therapeutic entry points for a most serious clinical important disorder in dire need for new diagnostic and therapeutic paradigms.
Max ERC Funding
1 816 250 €
Duration
Start date: 2015-06-01, End date: 2020-05-31
Project acronym LSD1
Project The lysine-specific demethylase1 (LSD1) in physiology and pathology
Researcher (PI) Roland Schuele
Host Institution (HI) UNIVERSITAETSKLINIKUM FREIBURG
Call Details Advanced Grant (AdG), LS4, ERC-2012-ADG_20120314
Summary "The identification of the first histone demethylase lysine-specific demethylase 1 (LSD1) established not only the concept of reversible histone methylation in epigenetic regulation but also translated this fundamentally novel biological observation into understanding the molecular mechanisms regulation stemness, differentiation, proliferation, and pathological growth. To unravel in an unbiased and comprehensive manner the biological function of LSD1 in physiology and pathology, we developed LSD1-deficient and LSD1-transgenic mouse models. LSD1-transgenic animals develop prostate tumours demonstrating that increased expression of LSD1 suffices for oncogenic growth in vivo. In addition, LSD1-transgenic animals exhibit a metabolic shift towards overt obesity in adulthood. LSD1-deficiency causes early embryonic lethality around day 7.5 of development. However, deletion of LSD1 is not essential for the development of the embryo proper until the onset of gastrulation, suggesting that the early embryonic lethality is caused by trophoblast defects. Indeed, our data demonstrate that LSD1 is crucial for maintaining trophoblast stem cells in their niche and required for the specification of trophoblast stem cell fate during initial steps of differentiation. To identify the underlying mechanisms that allow LSD1 to control a wide range of biological systems such as trophoblast stem cell fate in the early embryo, obesity, and prostate tumourigenesis in the adult, we propose to a) identify LSD1-associated protein complexes and b) LSD1 target genes establishing these phenotypes in the mouse. In addition, we shall uncover c) signalling pathways that modify LSD1 in these phenotypes allowing us to explore the therapeutic potential of targeting these signalling pathways."
Summary
"The identification of the first histone demethylase lysine-specific demethylase 1 (LSD1) established not only the concept of reversible histone methylation in epigenetic regulation but also translated this fundamentally novel biological observation into understanding the molecular mechanisms regulation stemness, differentiation, proliferation, and pathological growth. To unravel in an unbiased and comprehensive manner the biological function of LSD1 in physiology and pathology, we developed LSD1-deficient and LSD1-transgenic mouse models. LSD1-transgenic animals develop prostate tumours demonstrating that increased expression of LSD1 suffices for oncogenic growth in vivo. In addition, LSD1-transgenic animals exhibit a metabolic shift towards overt obesity in adulthood. LSD1-deficiency causes early embryonic lethality around day 7.5 of development. However, deletion of LSD1 is not essential for the development of the embryo proper until the onset of gastrulation, suggesting that the early embryonic lethality is caused by trophoblast defects. Indeed, our data demonstrate that LSD1 is crucial for maintaining trophoblast stem cells in their niche and required for the specification of trophoblast stem cell fate during initial steps of differentiation. To identify the underlying mechanisms that allow LSD1 to control a wide range of biological systems such as trophoblast stem cell fate in the early embryo, obesity, and prostate tumourigenesis in the adult, we propose to a) identify LSD1-associated protein complexes and b) LSD1 target genes establishing these phenotypes in the mouse. In addition, we shall uncover c) signalling pathways that modify LSD1 in these phenotypes allowing us to explore the therapeutic potential of targeting these signalling pathways."
Max ERC Funding
2 488 800 €
Duration
Start date: 2013-04-01, End date: 2018-03-31
Project acronym LUCKY STAR
Project Exploring the outer solar system beyond Neptune using stellar occultations
Researcher (PI) Bruno SICARDY
Host Institution (HI) SORBONNE UNIVERSITE
Call Details Advanced Grant (AdG), PE9, ERC-2014-ADG
Summary The solar system beyond Neptune’s contains largely unaltered material from the primordial circum-solar disk. It also kept the memory of the early planetary migrations, and thus contains essential information on the origin and evolution of our planetary system.
Here I propose to study the Trans-Neptunian Objects (TNOs) using the stellar occultation technique. It consists in observing the passage of remote TNOs in front of those “Lucky Stars”, that reveal shapes, atmosphere and rings of bodies from sub-km to thousand-km in size. Very few teams in the world master this method. The European-led network that I coordinate is now leader in predictions, instrumentation, observations and analysis related to stellar occultations, with innovative approaches and unprecedented results.
In the last decade, our group led the field by discovering rings around the asteroid-like object Chariklo, detecting sub-km TNOs and drastic variations of Pluto’s atmospheric pressure. Based on those noteworthy discoveries and unique skills of ours, I will coordinate the following work packages:
(1) Rings around small bodies - Understand the newly found Chariklo’s rings, tackle the theory of rings’ origins and evolutions around small bodies, discover new ring systems around other bodies.
(2) Very small, sub-km TNOs and Oort Cloud objects - Constrain the collisional history of our early outer solar system, and possibly detect Oort Cloud objects.
(3) Pluto’s atmosphere – Explore Pluto’s atmosphere and its atypical seasonal cycle, search for atmospheres around other TNOs.
(4) Explore specific, large TNOs – Provide their sizes, shapes, albedos and densities.
These programs are timely in view of NASA/New Horizons Pluto flyby in July 2015, and the ESA/GAIA mission expected to provide a greatly improved astrometric catalog release in 2016.
Most of the budget will be dedicated to human power to conduct observations and their analysis, plus the associated travel and telescope time expenses.
Summary
The solar system beyond Neptune’s contains largely unaltered material from the primordial circum-solar disk. It also kept the memory of the early planetary migrations, and thus contains essential information on the origin and evolution of our planetary system.
Here I propose to study the Trans-Neptunian Objects (TNOs) using the stellar occultation technique. It consists in observing the passage of remote TNOs in front of those “Lucky Stars”, that reveal shapes, atmosphere and rings of bodies from sub-km to thousand-km in size. Very few teams in the world master this method. The European-led network that I coordinate is now leader in predictions, instrumentation, observations and analysis related to stellar occultations, with innovative approaches and unprecedented results.
In the last decade, our group led the field by discovering rings around the asteroid-like object Chariklo, detecting sub-km TNOs and drastic variations of Pluto’s atmospheric pressure. Based on those noteworthy discoveries and unique skills of ours, I will coordinate the following work packages:
(1) Rings around small bodies - Understand the newly found Chariklo’s rings, tackle the theory of rings’ origins and evolutions around small bodies, discover new ring systems around other bodies.
(2) Very small, sub-km TNOs and Oort Cloud objects - Constrain the collisional history of our early outer solar system, and possibly detect Oort Cloud objects.
(3) Pluto’s atmosphere – Explore Pluto’s atmosphere and its atypical seasonal cycle, search for atmospheres around other TNOs.
(4) Explore specific, large TNOs – Provide their sizes, shapes, albedos and densities.
These programs are timely in view of NASA/New Horizons Pluto flyby in July 2015, and the ESA/GAIA mission expected to provide a greatly improved astrometric catalog release in 2016.
Most of the budget will be dedicated to human power to conduct observations and their analysis, plus the associated travel and telescope time expenses.
Max ERC Funding
2 423 750 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym LUNELY
Project ALK as a common target for the pathogenesis and therapy in lymphoma, lung carcinoma and neuroblastoma
Researcher (PI) Roberto Chiarle
Host Institution (HI) UNIVERSITA DEGLI STUDI DI TORINO
Call Details Starting Grant (StG), LS4, ERC-2009-StG
Summary The Anaplastic Lymphoma Kinase (ALK) has been discovered as the result of chromosomal translocations in Anaplastic Large Cell Lymphomas (ALCL) (Chiarle et al Nat Rev Cancer. 2008, 8:11). In ALCL, the role of the ALK oncogenic translocations has been established in vitro and in transgenic mouse models. Recent findings have shown ALK translocations, mutations or amplifications in other types of solid cancers, such as lung carcinoma (Soda et al. Nature. 2007, 448:561) and neuroblastoma (Mossè et al. Nature 2008, 455: 930). However, the role of ALK gene mutations in these solid tumours remains largely undetermined. This lack of knowledge is even worse given the fact that a therapy that targets ALK in these tumours could be feasible. Aim 1. Targeting of ALK in ALCL lymphomas. ALCL ALK positive lymphomas will be tested for small molecule inhibitors of the activity of ALK. In addition, a combination of gene silencing, such as small interfering RNA (siRNA), and vaccination against ALK will be validated as selective ALK therapies. Aim 2. Characterization of the role of ALK in lung cancer through the generation of mouse models. We propose to characterize the pathogenetic role of ALK in lung cancer by in vitro studies and by generating mouse models for ALK positive lung cancers. These mouse models will be fundamental to validate the innovative therapies against ALK positive lung carcinoma. Aim 3. Validation of ALK as an oncogene and a therapeutic target in neuroblastoma. We plan to develop mouse models of neuroblastoma to investigate the pathogenetic role of ALK in the onset and maintenance of neuroblastoma in vivo. These mouse model of neuroblastoma will be used for the validation of ALK specific therapies. Overall, the proposed project will define the role of ALK in lymphoma, neuroblastoma and lungcancer and validate its potential use as a a target for therapy in those tumours. The impact of these novel therapies will be of great value in these deadly tumours.
Summary
The Anaplastic Lymphoma Kinase (ALK) has been discovered as the result of chromosomal translocations in Anaplastic Large Cell Lymphomas (ALCL) (Chiarle et al Nat Rev Cancer. 2008, 8:11). In ALCL, the role of the ALK oncogenic translocations has been established in vitro and in transgenic mouse models. Recent findings have shown ALK translocations, mutations or amplifications in other types of solid cancers, such as lung carcinoma (Soda et al. Nature. 2007, 448:561) and neuroblastoma (Mossè et al. Nature 2008, 455: 930). However, the role of ALK gene mutations in these solid tumours remains largely undetermined. This lack of knowledge is even worse given the fact that a therapy that targets ALK in these tumours could be feasible. Aim 1. Targeting of ALK in ALCL lymphomas. ALCL ALK positive lymphomas will be tested for small molecule inhibitors of the activity of ALK. In addition, a combination of gene silencing, such as small interfering RNA (siRNA), and vaccination against ALK will be validated as selective ALK therapies. Aim 2. Characterization of the role of ALK in lung cancer through the generation of mouse models. We propose to characterize the pathogenetic role of ALK in lung cancer by in vitro studies and by generating mouse models for ALK positive lung cancers. These mouse models will be fundamental to validate the innovative therapies against ALK positive lung carcinoma. Aim 3. Validation of ALK as an oncogene and a therapeutic target in neuroblastoma. We plan to develop mouse models of neuroblastoma to investigate the pathogenetic role of ALK in the onset and maintenance of neuroblastoma in vivo. These mouse model of neuroblastoma will be used for the validation of ALK specific therapies. Overall, the proposed project will define the role of ALK in lymphoma, neuroblastoma and lungcancer and validate its potential use as a a target for therapy in those tumours. The impact of these novel therapies will be of great value in these deadly tumours.
Max ERC Funding
1 010 000 €
Duration
Start date: 2009-11-01, End date: 2014-04-30
Project acronym LYMPHIMMUNE
Project Flow in the tumor microenvironment: Linking mechanobiology with immunology
Researcher (PI) Melody Ann Swartz
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), LS4, ERC-2012-ADG_20120314
Summary Tumors often engage the lymphatic system to invade and metastasize. The tumor-draining lymph node (dLN) may be an immune privileged site that protects the tumor from host immunity, and lymph flow draining tumors is often increased, enhancing communication between the tumor and the sentinel node. In addition to increasing transport of tumor antigens and regulatory cytokines to the lymph node, increased lymph flow in the tumor margin causes mechanical stress-induced changes in stromal cells that stiffen the matrix and alter the immune microenvironment of the tumor. In this proposed project, we will investigate the interplay between lymphatic drainage and flow-induced mechanotransduction in the tumor stroma that may synergize to promote tumor immune escape by appropriating lymphatic mechanisms of peripheral tolerance. We will address the hypothesis that lymphatic drainage and flow-induced mechanotransduction in the tumor stroma synergistically promote tumor immune escape by altering the immune microenvironment, and that targeting lymphatic drainage from the tumor may represent a new avenue for tumor immunotherapy. For the latter, we will develop strategies to limit or block lymphatic flow in the tumor microenvironment and characterize their ability to improve the efficacy of tumor immunotherapy by dampening local immunosuppression in the tumor stroma and tumor-draining lymph node (dLN). We will combine in vivo mouse models and intravital imaging with engineered in vitro microenvironments and nanoparticle-based targeting strategies in three broad aims designed to constitute several PhD and postdoctoral projects.
Summary
Tumors often engage the lymphatic system to invade and metastasize. The tumor-draining lymph node (dLN) may be an immune privileged site that protects the tumor from host immunity, and lymph flow draining tumors is often increased, enhancing communication between the tumor and the sentinel node. In addition to increasing transport of tumor antigens and regulatory cytokines to the lymph node, increased lymph flow in the tumor margin causes mechanical stress-induced changes in stromal cells that stiffen the matrix and alter the immune microenvironment of the tumor. In this proposed project, we will investigate the interplay between lymphatic drainage and flow-induced mechanotransduction in the tumor stroma that may synergize to promote tumor immune escape by appropriating lymphatic mechanisms of peripheral tolerance. We will address the hypothesis that lymphatic drainage and flow-induced mechanotransduction in the tumor stroma synergistically promote tumor immune escape by altering the immune microenvironment, and that targeting lymphatic drainage from the tumor may represent a new avenue for tumor immunotherapy. For the latter, we will develop strategies to limit or block lymphatic flow in the tumor microenvironment and characterize their ability to improve the efficacy of tumor immunotherapy by dampening local immunosuppression in the tumor stroma and tumor-draining lymph node (dLN). We will combine in vivo mouse models and intravital imaging with engineered in vitro microenvironments and nanoparticle-based targeting strategies in three broad aims designed to constitute several PhD and postdoctoral projects.
Max ERC Funding
2 217 582 €
Duration
Start date: 2013-05-01, End date: 2016-12-31
Project acronym LymphMap
Project Navigating lymphatic formation and function in health and disease
Researcher (PI) Karina YANIV
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS4, ERC-2018-COG
Summary For many years, lymphatic vessels have been viewed as inert fluid conduits whose open structure allows for passive flow of antigens, proteins and cells from peripheral tissues to lymphoid organs. Yet, recent discoveries highlighting novel functions and heterogeneous origins of the lymphatic endothelium, call for reevaluation of the passive lymphatic-vessel paradigm. During the past decade, we have used the zebrafish (ZF) to detail the cellular and molecular events underlying the development of the lymphatic system. Our discoveries have greatly contributed to our understanding of the origins, specification and mechanisms of formation of lymphatic endothelial cells (LECs) in the developing embryo. In line with our past achievements, we now aim towards novel directions- to transform the adult ZF into an equally convenient model for the study of lymphatic diversity. The overall goal of LymphMap is to reveal the multiple regulatory levels that coordinate the formation and functionality of lymphatic vessels in health and disease. To this end, we will carry out a comprehensive research program characterizing four distinct aspects of lymphatic biology:
1.Cellular origins and molecular signature of LECs
2.Formation and specialization of organotypic lymphatics
3.Lymphatic vessels during organ regeneration
4.Lymphatic involvement in human disease
Our experimental strategy involves the combination of high-resolution imaging, global expression profiling and regeneration models in adult ZF, with analyses of human-derived LECs in various clinical settings. The important and unique aspects of our approach are the focus on in vivo dynamics, and the cross-organ comparative analysis, which will likely provide the much-needed knowledge on lymphatic diversity in health and disease. When completed, we anticipate that this work will be part of a new paradigm – no longer perceiving lymphatics as passive bystanders, but rather as orchestrators of tissue morphogenesis and regeneration.
Summary
For many years, lymphatic vessels have been viewed as inert fluid conduits whose open structure allows for passive flow of antigens, proteins and cells from peripheral tissues to lymphoid organs. Yet, recent discoveries highlighting novel functions and heterogeneous origins of the lymphatic endothelium, call for reevaluation of the passive lymphatic-vessel paradigm. During the past decade, we have used the zebrafish (ZF) to detail the cellular and molecular events underlying the development of the lymphatic system. Our discoveries have greatly contributed to our understanding of the origins, specification and mechanisms of formation of lymphatic endothelial cells (LECs) in the developing embryo. In line with our past achievements, we now aim towards novel directions- to transform the adult ZF into an equally convenient model for the study of lymphatic diversity. The overall goal of LymphMap is to reveal the multiple regulatory levels that coordinate the formation and functionality of lymphatic vessels in health and disease. To this end, we will carry out a comprehensive research program characterizing four distinct aspects of lymphatic biology:
1.Cellular origins and molecular signature of LECs
2.Formation and specialization of organotypic lymphatics
3.Lymphatic vessels during organ regeneration
4.Lymphatic involvement in human disease
Our experimental strategy involves the combination of high-resolution imaging, global expression profiling and regeneration models in adult ZF, with analyses of human-derived LECs in various clinical settings. The important and unique aspects of our approach are the focus on in vivo dynamics, and the cross-organ comparative analysis, which will likely provide the much-needed knowledge on lymphatic diversity in health and disease. When completed, we anticipate that this work will be part of a new paradigm – no longer perceiving lymphatics as passive bystanders, but rather as orchestrators of tissue morphogenesis and regeneration.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-02-01, End date: 2024-01-31
Project acronym LYMPHOONCOGENOMICS
Project Deciphering the Oncogenic Lesions and Pathways of B and T Cell Cancers
Researcher (PI) Ludovic Gérard Marie Deriano
Host Institution (HI) INSTITUT PASTEUR
Call Details Starting Grant (StG), LS4, ERC-2012-StG_20111109
Summary Lymphoid cancers are among the most common human malignancies and characteristically harbour genomic aberrations. Although databases will soon be flooded with genome sequences from thousands of tumours, interpretation and validation of these data is limited by the genetic variability and uncontrolled environmental exposures inherent to human studies. There is therefore a growing need in complementary approaches that would provide understanding of how and which genomic aberrations underlie tumour genesis. It has become apparent that the generation of DNA double strand breaks during the process of antigen receptor diversification, including RAG1/2 protein-generated DNA breaks during V(D)J recombination, are key intermediates in the appearance of lymphoid neoplasm. In this project, we propose to elucidate the molecular mechanisms, the genomic lesions and the oncogenic pathways underlying B and T cell cancers using a combination of mouse models and cutting-edge cytogenetic, genomic and transcriptomic technologies. We aim (i) to define the major genomic lesions and oncogenic pathways underlying the rapid onset of T-cell lymphomas in a unique mouse model that carries exacerbated RAG-induced genomic instability; (ii) to decipher the genomic lesions and pathways underlying B versus T lymphoma tropism; (iii) to investigate the functional interactions between RAG- and DNA damage response / DNA repair-complexes in suppressing genomic instability and lymphomagenesis; and (iv) to identify and validate lesions and genes underlying human B and T cell cancers using a cross-species oncogenomic comparative approach. In doing so, this study will also 1) increase our understanding of the fundamental mechanisms maintaining genome stability during lymphocyte development; 2) greatly improve interpretation and validation of whole genome sequencing analysis of human leukaemia and lymphoma; and 3) provide well-characterized animal models for developing and testing targeted therapeutics.
Summary
Lymphoid cancers are among the most common human malignancies and characteristically harbour genomic aberrations. Although databases will soon be flooded with genome sequences from thousands of tumours, interpretation and validation of these data is limited by the genetic variability and uncontrolled environmental exposures inherent to human studies. There is therefore a growing need in complementary approaches that would provide understanding of how and which genomic aberrations underlie tumour genesis. It has become apparent that the generation of DNA double strand breaks during the process of antigen receptor diversification, including RAG1/2 protein-generated DNA breaks during V(D)J recombination, are key intermediates in the appearance of lymphoid neoplasm. In this project, we propose to elucidate the molecular mechanisms, the genomic lesions and the oncogenic pathways underlying B and T cell cancers using a combination of mouse models and cutting-edge cytogenetic, genomic and transcriptomic technologies. We aim (i) to define the major genomic lesions and oncogenic pathways underlying the rapid onset of T-cell lymphomas in a unique mouse model that carries exacerbated RAG-induced genomic instability; (ii) to decipher the genomic lesions and pathways underlying B versus T lymphoma tropism; (iii) to investigate the functional interactions between RAG- and DNA damage response / DNA repair-complexes in suppressing genomic instability and lymphomagenesis; and (iv) to identify and validate lesions and genes underlying human B and T cell cancers using a cross-species oncogenomic comparative approach. In doing so, this study will also 1) increase our understanding of the fundamental mechanisms maintaining genome stability during lymphocyte development; 2) greatly improve interpretation and validation of whole genome sequencing analysis of human leukaemia and lymphoma; and 3) provide well-characterized animal models for developing and testing targeted therapeutics.
Max ERC Funding
1 498 940 €
Duration
Start date: 2013-02-01, End date: 2019-01-31
Project acronym LYMPHORG
Project Organ-specific mechanisms of lymphatic vascular development and specialisation
Researcher (PI) Taija Marianna Makinen
Host Institution (HI) UPPSALA UNIVERSITET
Call Details Consolidator Grant (CoG), LS4, ERC-2014-CoG
Summary Lymphatic vasculature maintains tissue fluid homeostasis and has important emerging roles in inflammation, immunity, lipid metabolism, blood pressure regulation and cancer metastasis. Lymphatic vessels are specialised to fulfil the functional needs of different organs while diseases associated with lymphatic dysfunction frequently affect vessels of specific tissues. How functional specialisation of vessels is achieved and what underlies tissue-specific vessel failure is not understood. I hypothesise that organ-specific manifestation of lymphatic dysfunction in disease is due to vascular bed-specific differences in vessel formation. In this project my aim is to identify genes and mechanisms required for organ-specific lymphatic development. Building on our recent discovery of a previously unknown progenitor cell type that is required for lymphatic development in an organ-specific manner I set out to identify the origin and function of lymphatic endothelial progenitor cells (LEPC) during development and assess their potential for therapeutic lymphatic regeneration. Towards this aim, we will identify organ-specific origins of lymphatic vasculature using lineage tracing and determine genetic signatures of lymphatic endothelial progenitors by mRNA sequencing. Cells and tissues from normal and mutant mice that show organ-specific lymphatic defects will be analysed. To identify molecular and cellular mechanisms of LEPC derived vessel formation, we will functionally characterise LEPC signature genes using mouse models and visualise vessel development by in vivo two-photon microscopy. The function and therapeutic potential of LEPCs and LEPC derived vessels will be assessed using mouse models of tolerance, inflammation, obesity and lymphoedema. This work will provide novel insights into organ-specific mechanisms of vascular morphogenesis and identify a progenitor cell that may be expoited to restore lymphatic function in disorders associated with lymphatic vessel failure.
Summary
Lymphatic vasculature maintains tissue fluid homeostasis and has important emerging roles in inflammation, immunity, lipid metabolism, blood pressure regulation and cancer metastasis. Lymphatic vessels are specialised to fulfil the functional needs of different organs while diseases associated with lymphatic dysfunction frequently affect vessels of specific tissues. How functional specialisation of vessels is achieved and what underlies tissue-specific vessel failure is not understood. I hypothesise that organ-specific manifestation of lymphatic dysfunction in disease is due to vascular bed-specific differences in vessel formation. In this project my aim is to identify genes and mechanisms required for organ-specific lymphatic development. Building on our recent discovery of a previously unknown progenitor cell type that is required for lymphatic development in an organ-specific manner I set out to identify the origin and function of lymphatic endothelial progenitor cells (LEPC) during development and assess their potential for therapeutic lymphatic regeneration. Towards this aim, we will identify organ-specific origins of lymphatic vasculature using lineage tracing and determine genetic signatures of lymphatic endothelial progenitors by mRNA sequencing. Cells and tissues from normal and mutant mice that show organ-specific lymphatic defects will be analysed. To identify molecular and cellular mechanisms of LEPC derived vessel formation, we will functionally characterise LEPC signature genes using mouse models and visualise vessel development by in vivo two-photon microscopy. The function and therapeutic potential of LEPCs and LEPC derived vessels will be assessed using mouse models of tolerance, inflammation, obesity and lymphoedema. This work will provide novel insights into organ-specific mechanisms of vascular morphogenesis and identify a progenitor cell that may be expoited to restore lymphatic function in disorders associated with lymphatic vessel failure.
Max ERC Funding
2 368 439 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym LYVICAM
Project Lymphatic Vessels in Inflammation and Cancer Metastasis
Researcher (PI) Michael Johannes Detmar
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), LS4, ERC-2010-AdG_20100317
Summary Primary cancers can induce lymphatic vessel growth (lymphangiogenesis), enhancing metastasis to draining lymph nodes (LNs). We found that tumors also induce lymphangiogenesis in draining LNs, leading to increased cancer spread to distal LNs and beyond. Very recently, we found that lymphatic vessel activation in peripheral tissues and draining LNs also plays a previously unanticipated role in the control of chronic inflammatory diseases. This proposal aims at a comprehensive characterization of the function of lymphatic vessels in inflammation, using a variety of genetic mouse models for enhanced or reduced lymphatic function, novel quantitative techniques for the in vivo imaging of lymphatic function, and a miniaturized 3-dimensional in vitro platform for the high-throughput phenotypic screening of libraries of small, drug-like molecules for modulators of lymphatic function. A genome-wide analysis of the gene expression profile shall be made from lymphatic vessels isolated by high-speed cell sorting and by immuno-laser capture microdissection from tumors and their lymph node metastases, inflamed tissue and its draining lymph nodes, and normal tissues, followed by functional characterization of potential therapeutic targets and diagnostic markers. Finally, we will establish novel genetically fluorescent mouse models for the in vivo real-time imaging of lymphatic activation, and we will develop an innovative approach for the in vivo detection of early micrometastases, using antibody-based PET and near-infrared imaging of tumor-induced stromal changes. These studies will improve our understanding of lymphatic involvement in inflammation and cancer metastasis, and will provide the basis for completely novel approaches to treat and detect inflammation and cancer metastasis.
Summary
Primary cancers can induce lymphatic vessel growth (lymphangiogenesis), enhancing metastasis to draining lymph nodes (LNs). We found that tumors also induce lymphangiogenesis in draining LNs, leading to increased cancer spread to distal LNs and beyond. Very recently, we found that lymphatic vessel activation in peripheral tissues and draining LNs also plays a previously unanticipated role in the control of chronic inflammatory diseases. This proposal aims at a comprehensive characterization of the function of lymphatic vessels in inflammation, using a variety of genetic mouse models for enhanced or reduced lymphatic function, novel quantitative techniques for the in vivo imaging of lymphatic function, and a miniaturized 3-dimensional in vitro platform for the high-throughput phenotypic screening of libraries of small, drug-like molecules for modulators of lymphatic function. A genome-wide analysis of the gene expression profile shall be made from lymphatic vessels isolated by high-speed cell sorting and by immuno-laser capture microdissection from tumors and their lymph node metastases, inflamed tissue and its draining lymph nodes, and normal tissues, followed by functional characterization of potential therapeutic targets and diagnostic markers. Finally, we will establish novel genetically fluorescent mouse models for the in vivo real-time imaging of lymphatic activation, and we will develop an innovative approach for the in vivo detection of early micrometastases, using antibody-based PET and near-infrared imaging of tumor-induced stromal changes. These studies will improve our understanding of lymphatic involvement in inflammation and cancer metastasis, and will provide the basis for completely novel approaches to treat and detect inflammation and cancer metastasis.
Max ERC Funding
2 493 300 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym M2C
Project FOLLOWING THE MOST MASSIVE GALAXY CLUSTERS ACROSS COSMIC TIME
Researcher (PI) Monique Arnaud
Host Institution (HI) COMMISSARIAT A L ENERGIE ATOMIQUE ET AUX ENERGIES ALTERNATIVES
Call Details Advanced Grant (AdG), PE9, ERC-2013-ADG
Summary "Our project aims at testing the standard LCDM scenario for the formation of collapsed structures. Taking advantage of the advent of cluster detection via the SZ effect, we will use the most massive clusters of galaxies and their evolution as laboratory. We build on the Planck SZ survey, the first All Sky survey since the RASS X-ray survey. We will substantially extend the nominal Planck cluster catalogue by developing novel detection techniques based on a simultaneous search of objects in Planck and RASS maps, reaching lower masses and higher redshifts, while keeping a high catalogue purity.
For the first time, we will have the sample size, redshift leverage, and completeness, for a decisive test of the standard LCDM model of the dark matter gravitational collapse on cluster scale. The test will be provided by a full statistical analysis of the dark matter profiles and their evolution. The X-ray technique to derive mass profile will be extended to the full cluster population. This will be made possible by an integrated approach involving systematic confrontation of observations with tailor-made numerical simulations.
Using multi-wavelength data and simulations, we will also assess the dynamical behaviour of the baryons as they collect in dark matter potential. We will i) provide the first complete census of the dark matter, hot and cold baryons and its evolution ii) quantify the thermo- dynamical state up to z~1 as a probe of hierarchical structure formation and gravitational heating iii) probe the link between non-thermal and thermal components.
The project will either cement our current understanding of the dark matter collapse, a prerequisite to any assessment of the specific baryon physics, or points towards the need for revision of the current paradigm, with important cosmological implications. The new detection techniques will be applicable to future surveys. Lastly, we will provide a ‘gold sample’ of galaxy clusters, ideal for cosmological parameter estimate."
Summary
"Our project aims at testing the standard LCDM scenario for the formation of collapsed structures. Taking advantage of the advent of cluster detection via the SZ effect, we will use the most massive clusters of galaxies and their evolution as laboratory. We build on the Planck SZ survey, the first All Sky survey since the RASS X-ray survey. We will substantially extend the nominal Planck cluster catalogue by developing novel detection techniques based on a simultaneous search of objects in Planck and RASS maps, reaching lower masses and higher redshifts, while keeping a high catalogue purity.
For the first time, we will have the sample size, redshift leverage, and completeness, for a decisive test of the standard LCDM model of the dark matter gravitational collapse on cluster scale. The test will be provided by a full statistical analysis of the dark matter profiles and their evolution. The X-ray technique to derive mass profile will be extended to the full cluster population. This will be made possible by an integrated approach involving systematic confrontation of observations with tailor-made numerical simulations.
Using multi-wavelength data and simulations, we will also assess the dynamical behaviour of the baryons as they collect in dark matter potential. We will i) provide the first complete census of the dark matter, hot and cold baryons and its evolution ii) quantify the thermo- dynamical state up to z~1 as a probe of hierarchical structure formation and gravitational heating iii) probe the link between non-thermal and thermal components.
The project will either cement our current understanding of the dark matter collapse, a prerequisite to any assessment of the specific baryon physics, or points towards the need for revision of the current paradigm, with important cosmological implications. The new detection techniques will be applicable to future surveys. Lastly, we will provide a ‘gold sample’ of galaxy clusters, ideal for cosmological parameter estimate."
Max ERC Funding
2 456 035 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym MADCIN
Project Defining the Impact of Chromosome Instability in Tumor Initiation, Maintenance and Relapse
Researcher (PI) Rocio Sotillo Roman
Host Institution (HI) DEUTSCHES KREBSFORSCHUNGSZENTRUM HEIDELBERG
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary Chromosomal instability (CIN), the inability to correctly segregate sister chromatids during mitosis, is a hallmark of cancer cells. Overexpression of the mitotic checkpoint protein Mad2, commonly found in human tumors, leads to CIN and the development of aneuploid tumors in mouse models. However, recent observations from various laboratories suggest that aneuploidy can promote or suppress tumorigenesis. Therefore understanding the relationship between aneuploidy and tumor formation, identifying in what context aneuploidy acts oncogenically and those in which it acts as a tumor suppressor, is thus vital if we want to make progress in battling cancer. We propose to generate regulatable mouse models that recapitulate the aneuploidy state of human tumors, using state-of–the–art mouse genetic strategies, to investigate the role of CIN in promoting or suppressing tumorigenesis.
Moreover, CIN has been shown to facilitate escape from oncogene addiction (the dependence of tumor cells on their initiating lesion for survival) and may be responsible for tumor relapse after targeted therapies. Due to the clinical relevance of these findings, this proposal aims to investigate how CIN potentiates oncogene independence. It is possible that some CIN cells in the primary tumor are already independent of the initiating oncogene prior to treatment. Alternatively, CIN cells are more susceptible of acquiring additional mutations and evolve to become independent of the initiating lesion. We propose to develop a highly innovative three-dimensional in vitro culture system to isolate and characterize these surviving cells to further eliminate them.
It is necessary to understand the molecular mechanisms that lead to CIN and the consequences it has in tumor initiation, suppression and relapse, hoping that the genes or proteins identified could be targeted therapeutically. We believe that answers to these specific aims will have important consequences for the treatment of human tumors.
Summary
Chromosomal instability (CIN), the inability to correctly segregate sister chromatids during mitosis, is a hallmark of cancer cells. Overexpression of the mitotic checkpoint protein Mad2, commonly found in human tumors, leads to CIN and the development of aneuploid tumors in mouse models. However, recent observations from various laboratories suggest that aneuploidy can promote or suppress tumorigenesis. Therefore understanding the relationship between aneuploidy and tumor formation, identifying in what context aneuploidy acts oncogenically and those in which it acts as a tumor suppressor, is thus vital if we want to make progress in battling cancer. We propose to generate regulatable mouse models that recapitulate the aneuploidy state of human tumors, using state-of–the–art mouse genetic strategies, to investigate the role of CIN in promoting or suppressing tumorigenesis.
Moreover, CIN has been shown to facilitate escape from oncogene addiction (the dependence of tumor cells on their initiating lesion for survival) and may be responsible for tumor relapse after targeted therapies. Due to the clinical relevance of these findings, this proposal aims to investigate how CIN potentiates oncogene independence. It is possible that some CIN cells in the primary tumor are already independent of the initiating oncogene prior to treatment. Alternatively, CIN cells are more susceptible of acquiring additional mutations and evolve to become independent of the initiating lesion. We propose to develop a highly innovative three-dimensional in vitro culture system to isolate and characterize these surviving cells to further eliminate them.
It is necessary to understand the molecular mechanisms that lead to CIN and the consequences it has in tumor initiation, suppression and relapse, hoping that the genes or proteins identified could be targeted therapeutically. We believe that answers to these specific aims will have important consequences for the treatment of human tumors.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-02-01, End date: 2018-01-31
Project acronym MAGALOPS
Project The MAgnetic field in the GALaxy, using Optical Polarization of Stars
Researcher (PI) Marijke Haverkorn van Rijsewijk
Host Institution (HI) STICHTING KATHOLIEKE UNIVERSITEIT
Call Details Consolidator Grant (CoG), PE9, ERC-2017-COG
Summary What makes our Galaxy’s ecosystem so fascinating is the complex interactions between its components: stars, gas, dust, magnetic fields, and cosmic rays. Of these components, the Galactic magnetic field (GMF) may well be the most enigmatic. Only partially observable through indirect means, its study relies heavily on modeling, almost exclusively using line-of-sight integrated radio-polarimetric data. Although much has been learned, many questions are still unanswered especially about the turbulent, small-scale field component and out-of-plane field.
The crucial innovations proposed here are large independent data sets with 3D (distance) information – which can only be provided by stars polarized due to differential absorption by interstellar dust, with known distances – and more advanced Bayesian statistics which allows including prior knowledge and enables quantitative model comparison.
I propose to use 2 new polarization surveys in the V (visual) band, resulting in polarimetry of millions of stars across the southern sky. With distance information provided by the GAIA satellite, this improves the current data situation by 3 orders of magnitude. We will test GMF models against all available data, employing a Bayesian inference software package which we are developing. In the process, we will produce the first 3D all-sky (out to absorption limits) dust distribution consistent with both UV/optical/near IR absorption and optical polarization.
This research will result in a next-generation GMF model that includes all observational GMF tracers and can use informative priors. It will allow mapping out interstellar magnetized turbulence in the Galaxy, instead of providing averaged parameters only, and understanding the interplay between the local GMF, gas and dust. Its legacy is a 1000x increased stellar polarization catalog, an all-sky 3D dust model, a bayesian sampler for GMF models, and a superior GMF model for use in cosmic ray modeling or foreground subtraction.
Summary
What makes our Galaxy’s ecosystem so fascinating is the complex interactions between its components: stars, gas, dust, magnetic fields, and cosmic rays. Of these components, the Galactic magnetic field (GMF) may well be the most enigmatic. Only partially observable through indirect means, its study relies heavily on modeling, almost exclusively using line-of-sight integrated radio-polarimetric data. Although much has been learned, many questions are still unanswered especially about the turbulent, small-scale field component and out-of-plane field.
The crucial innovations proposed here are large independent data sets with 3D (distance) information – which can only be provided by stars polarized due to differential absorption by interstellar dust, with known distances – and more advanced Bayesian statistics which allows including prior knowledge and enables quantitative model comparison.
I propose to use 2 new polarization surveys in the V (visual) band, resulting in polarimetry of millions of stars across the southern sky. With distance information provided by the GAIA satellite, this improves the current data situation by 3 orders of magnitude. We will test GMF models against all available data, employing a Bayesian inference software package which we are developing. In the process, we will produce the first 3D all-sky (out to absorption limits) dust distribution consistent with both UV/optical/near IR absorption and optical polarization.
This research will result in a next-generation GMF model that includes all observational GMF tracers and can use informative priors. It will allow mapping out interstellar magnetized turbulence in the Galaxy, instead of providing averaged parameters only, and understanding the interplay between the local GMF, gas and dust. Its legacy is a 1000x increased stellar polarization catalog, an all-sky 3D dust model, a bayesian sampler for GMF models, and a superior GMF model for use in cosmic ray modeling or foreground subtraction.
Max ERC Funding
2 000 000 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym MagBURST
Project Exploding stars from first principles: MAGnetars as engines of hypernovae and gamma-ray BURSTs
Researcher (PI) Jérôme GUILET
Host Institution (HI) COMMISSARIAT A L ENERGIE ATOMIQUE ET AUX ENERGIES ALTERNATIVES
Call Details Starting Grant (StG), PE9, ERC-2016-STG
Summary The birth of a neutron star with an extremely strong magnetic field, called a magnetar, has emerged as a promising scenario to power a variety of outstanding explosive events. This includes gamma-ray bursts, among the most luminous events observed up to high redshift and therefore useful as cosmological probes, but also supernovae with extreme kinetic energies called hypernovae and other classes of super-luminous supernovae. Simple phenomenological models, where the magnetar rotation period and magnetic field are adjusted, can explain many of these observations but lack a sound theoretical basis. The goal of this proposal is to develop an ab initio description of magnetar powered explosions in order to delineate the role they play for the production of gamma-ray bursts and super-luminous supernovae. This is urgently needed to interpret the growing diversity of explosions observed with ongoing transient surveys (iPTF, CRTS, Pan-STARRS) and in the perspective of future programs of observations such as SVOM and LSST. By using state-of-the-art numerical simulations, the following outstanding questions will be addressed:
1) What is the origin of the gigantic magnetic field observed in magnetars? The physics of the magnetic field amplification in a fast-rotating nascent neutron star will be investigated thoroughly from first principles. By developing the first global protoneutron star simulations of this amplification process, the magnetic field strength and geometry will be determined for varying rotation rates.
2) What variety of explosion paths can be explained by the birth of fast-rotating magnetars? Numerical simulations of the launch of a hypernova explosion and a relativistic GRB jet will provide the first self-consistent description of both events from a millisecond magnetar. Furthermore, the new understanding of magnetic field amplification will be used to improve the realism of these simulations.
Summary
The birth of a neutron star with an extremely strong magnetic field, called a magnetar, has emerged as a promising scenario to power a variety of outstanding explosive events. This includes gamma-ray bursts, among the most luminous events observed up to high redshift and therefore useful as cosmological probes, but also supernovae with extreme kinetic energies called hypernovae and other classes of super-luminous supernovae. Simple phenomenological models, where the magnetar rotation period and magnetic field are adjusted, can explain many of these observations but lack a sound theoretical basis. The goal of this proposal is to develop an ab initio description of magnetar powered explosions in order to delineate the role they play for the production of gamma-ray bursts and super-luminous supernovae. This is urgently needed to interpret the growing diversity of explosions observed with ongoing transient surveys (iPTF, CRTS, Pan-STARRS) and in the perspective of future programs of observations such as SVOM and LSST. By using state-of-the-art numerical simulations, the following outstanding questions will be addressed:
1) What is the origin of the gigantic magnetic field observed in magnetars? The physics of the magnetic field amplification in a fast-rotating nascent neutron star will be investigated thoroughly from first principles. By developing the first global protoneutron star simulations of this amplification process, the magnetic field strength and geometry will be determined for varying rotation rates.
2) What variety of explosion paths can be explained by the birth of fast-rotating magnetars? Numerical simulations of the launch of a hypernova explosion and a relativistic GRB jet will provide the first self-consistent description of both events from a millisecond magnetar. Furthermore, the new understanding of magnetic field amplification will be used to improve the realism of these simulations.
Max ERC Funding
1 500 000 €
Duration
Start date: 2017-05-01, End date: 2022-04-30
Project acronym MAGCOW
Project The Magnetised Cosmic Web
Researcher (PI) Franco VAZZA
Host Institution (HI) ALMA MATER STUDIORUM - UNIVERSITA DI BOLOGNA
Call Details Starting Grant (StG), PE9, ERC-2016-STG
Summary On large scales cosmic matter is distributed in a web consistent of clusters, filaments, walls and voids.
While the dark-matter skeleton of the cosmic web is closely traced by galaxies and galaxy clusters, the gaseous distribution has never been directly imaged at any wavelength. This situation might change within the next decade, thanks to the new generation of radio instruments that will survey the sky: LOFAR, MWA, ASKAP and the Square Kilometer Array. Non-thermal components, relativistic particles and magnetic fields are thought to have a spatial distribution that is broader than that of thermal baryons. For this reason, the new generation of radio telescopes should might be able to detect the tip of the iceberg from the rarefied intergalactic medium, provided that magnetic fields are sufficiently amplified in these regions. The detectable signal is expected to be weak and complex because of the contribution from radio galaxies and to the presence of diffuse fore- and backgrounds.
The developments proposed in this ERC proposal are exactly designed to address this complexity, and turn future radio observations into a unique probe of the growth of magnetic fields and of the acceleration of particles. This will be possible through the theoretical exploration of plasmas in extreme conditions with sophisticated numerical simulations. With these simulations I will be able to predict the specific radio signature for the origin of extragalactic fields. This will enable the community to use radio surveys in a quantitative way and to determine the origin of extragalactic magnetism, a longstanding puzzle connected to many open questions of modern astrophysics.
The legacy of this project will be its quantitative representation of non-thermal processes on the largest scales, ultimately going to be fully exploited by the Square Kilometer Array.
Summary
On large scales cosmic matter is distributed in a web consistent of clusters, filaments, walls and voids.
While the dark-matter skeleton of the cosmic web is closely traced by galaxies and galaxy clusters, the gaseous distribution has never been directly imaged at any wavelength. This situation might change within the next decade, thanks to the new generation of radio instruments that will survey the sky: LOFAR, MWA, ASKAP and the Square Kilometer Array. Non-thermal components, relativistic particles and magnetic fields are thought to have a spatial distribution that is broader than that of thermal baryons. For this reason, the new generation of radio telescopes should might be able to detect the tip of the iceberg from the rarefied intergalactic medium, provided that magnetic fields are sufficiently amplified in these regions. The detectable signal is expected to be weak and complex because of the contribution from radio galaxies and to the presence of diffuse fore- and backgrounds.
The developments proposed in this ERC proposal are exactly designed to address this complexity, and turn future radio observations into a unique probe of the growth of magnetic fields and of the acceleration of particles. This will be possible through the theoretical exploration of plasmas in extreme conditions with sophisticated numerical simulations. With these simulations I will be able to predict the specific radio signature for the origin of extragalactic fields. This will enable the community to use radio surveys in a quantitative way and to determine the origin of extragalactic magnetism, a longstanding puzzle connected to many open questions of modern astrophysics.
The legacy of this project will be its quantitative representation of non-thermal processes on the largest scales, ultimately going to be fully exploited by the Square Kilometer Array.
Max ERC Funding
1 465 943 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym MAGMIST
Project From the magnetized diffuse interstellar medium to the stars
Researcher (PI) Patrick Hennebelle
Host Institution (HI) COMMISSARIAT A L ENERGIE ATOMIQUE ET AUX ENERGIES ALTERNATIVES
Call Details Starting Grant (StG), PE9, ERC-2012-StG_20111012
Summary "Understanding star formation remains one of the greatest challenges of modern astronomy. Indeed in this field the progresses have been limited due, first, to the huge dynamics of spatial and temporal relevant scales and, second, the great variety and non-linearity of the physical processes involved in the formation of stars. The present proposal will contribute to provide a complete and coherent picture of the star formation process by self-consistently following the evolution of the interstellar matter from the very diffuse gas up to the protostars. This will be achieved by performing a series of heavy MHD numerical simulations with an adaptive mesh refinement code while subdivising the problem in three major steps namely the formation of large scale molecular clouds, the formation of star forming cores and the collapse of protostellar cores. In particular, the impact of the magnetic field and the radiative processes will be self-consistently treated using appropriate schemes. At each step, comparisons with both analytical models and observations will be performed by using existing models or developing new ones and calculating synthetic observations. The simulation results will also be used to test and improve the methods and the algorithms used by observers to extract the physical information from their data. An existing database, where the simulation results are available, will be further developed. The present proposal pursues two aims: i) achieving a global understanding of the star formation process, in particular by elucidating the link between the physical properties of the large scale ISM and the characteristics of the protostars, such as their mass, magnetisation and angular momentum ii) provide a better insight of the structure, nature and role of the magnetic field and the turbulence from the diffuse to the dense parts of the ISM."
Summary
"Understanding star formation remains one of the greatest challenges of modern astronomy. Indeed in this field the progresses have been limited due, first, to the huge dynamics of spatial and temporal relevant scales and, second, the great variety and non-linearity of the physical processes involved in the formation of stars. The present proposal will contribute to provide a complete and coherent picture of the star formation process by self-consistently following the evolution of the interstellar matter from the very diffuse gas up to the protostars. This will be achieved by performing a series of heavy MHD numerical simulations with an adaptive mesh refinement code while subdivising the problem in three major steps namely the formation of large scale molecular clouds, the formation of star forming cores and the collapse of protostellar cores. In particular, the impact of the magnetic field and the radiative processes will be self-consistently treated using appropriate schemes. At each step, comparisons with both analytical models and observations will be performed by using existing models or developing new ones and calculating synthetic observations. The simulation results will also be used to test and improve the methods and the algorithms used by observers to extract the physical information from their data. An existing database, where the simulation results are available, will be further developed. The present proposal pursues two aims: i) achieving a global understanding of the star formation process, in particular by elucidating the link between the physical properties of the large scale ISM and the characteristics of the protostars, such as their mass, magnetisation and angular momentum ii) provide a better insight of the structure, nature and role of the magnetic field and the turbulence from the diffuse to the dense parts of the ISM."
Max ERC Funding
1 312 267 €
Duration
Start date: 2013-01-01, End date: 2017-12-31
Project acronym MAGNESIA
Project The impact of highly magnetic neutron stars in the explosive and transient Universe
Researcher (PI) Nanda Rea
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Consolidator Grant (CoG), PE9, ERC-2018-COG
Summary The gravitational wave window is now open. It is then imperative to build quantitative models of neutron stars that use all the available tracers to constrain fundamental physics at the highest densities and magnetic fields. The most magnetic neutron stars, the magnetars, have been recently suggested to be powering a large variety of explosive and transient events. The enormous rotational power at birth, and the magnetic energy they can release via large flares, put the magnetars in the (yet) hand-wavy interpretations of gamma-ray bursts, the early phases of double neutron star mergers, super-luminous supernovae, hypernovae, fast radio bursts, and ultra-luminous X-ray sources. However, despite knowing about 30 magnetars, we are lacking a census of how many we expect within the pulsar population, nor we have robust constraints on their flaring rates. The recent discovery of transient magnetars, of magnetar-like flares from sources with measured low dipolar magnetic fields and from typical radio pulsars, clearly showed that the magnetar census in our Galaxy is largely under-estimated. This hampers our understanding not only of the pulsar and magnetar populations, but also of them as possibly related to many of Universe’s explosive events. MAGNESIA will infer a sound Magnetar Census via an innovative approach that will build the first Pulsar Population Synthesis model able to cope with constraints/limits from multi-band observations, and taking into account 3D magnetic field evolution models and flaring rates for neutron stars. Combining expertise in multi-band observations, numerical modeling, nuclear physics, and computation, MAGNESIA will solve the physics, the observational systematic errors, and the computational challenges that inhibited previous works, to finally constrain the spin period and magnetic field distribution at birth of the neutron star population.
Summary
The gravitational wave window is now open. It is then imperative to build quantitative models of neutron stars that use all the available tracers to constrain fundamental physics at the highest densities and magnetic fields. The most magnetic neutron stars, the magnetars, have been recently suggested to be powering a large variety of explosive and transient events. The enormous rotational power at birth, and the magnetic energy they can release via large flares, put the magnetars in the (yet) hand-wavy interpretations of gamma-ray bursts, the early phases of double neutron star mergers, super-luminous supernovae, hypernovae, fast radio bursts, and ultra-luminous X-ray sources. However, despite knowing about 30 magnetars, we are lacking a census of how many we expect within the pulsar population, nor we have robust constraints on their flaring rates. The recent discovery of transient magnetars, of magnetar-like flares from sources with measured low dipolar magnetic fields and from typical radio pulsars, clearly showed that the magnetar census in our Galaxy is largely under-estimated. This hampers our understanding not only of the pulsar and magnetar populations, but also of them as possibly related to many of Universe’s explosive events. MAGNESIA will infer a sound Magnetar Census via an innovative approach that will build the first Pulsar Population Synthesis model able to cope with constraints/limits from multi-band observations, and taking into account 3D magnetic field evolution models and flaring rates for neutron stars. Combining expertise in multi-band observations, numerical modeling, nuclear physics, and computation, MAGNESIA will solve the physics, the observational systematic errors, and the computational challenges that inhibited previous works, to finally constrain the spin period and magnetic field distribution at birth of the neutron star population.
Max ERC Funding
2 263 148 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym MagneticYSOs
Project Interpreting Dust Polarization Maps to Characterize the Role of the Magnetic Field in Star Formation Processes
Researcher (PI) Anaëlle Julie Maury
Host Institution (HI) COMMISSARIAT A L ENERGIE ATOMIQUE ET AUX ENERGIES ALTERNATIVES
Call Details Starting Grant (StG), PE9, ERC-2015-STG
Summary "Rotation and angular momentum transport play a critical role in the formation and evolution of astrophysical objects, including the fundamental bricks of astrophysical structures: stars. Stars like our Sun form when rotating dense cores, in the interstellar medium, collapse until they eventually reach temperatures at which nuclear fusion begins; while planets, including the Earth, form in the rotationally supported disks around these same young stars. One of the major challenges of modern astrophysics is the “angular momentum problem"": observations show that a typical star-forming cloud needs to reduce its specific angular momentum by 5 to 10 orders of magnitude to form a typical star such as our Sun. It is also crucial to solve the angular momentum problem to understand the formation of protoplanetary disks, stellar binaries and the initial mass function of newly formed stars. Magnetic fields are one of the key ways of transporting angular momentum in astrophysical structures: understanding how angular momentum is transported to allow star formation requires characterizing the role of magnetic fields in shaping the dynamics of star-forming structures. The MagneticYSOs project aims at characterizing the role of magnetic field in the earliest stage of star formation, during the main accretion phase.
The simultaneous major improvements of instrumental and computational facilities provide us, for the first time, with the opportunity to confront observational information to magnetized models predictions. Polarization capabilities on the last generation of instrument in large facilities are producing sensitive observations of magnetic fields with a great level of detail, while numerical simulations of star formation are now including most of the physical ingredients for a detailed description of protostellar collapse at all the relevant scales, such as resistive MHD, radiative transfer and chemical networks. These new tools will undoubtedly lead to major discovery in the fields of planets and star formation in the coming years. It is necessary to conduct comprehensive projects able to combine theory and observations in a detailed fashion, which in turn require a collaboration with access to cutting edge observational datasets and numerical models. Through an ambitious multi-faceted program of dedicated observations probing magnetic fields (polarized dust emission and Zeeman effect maps), gas kinematics (molecular lines emission maps), ionization rates and dust properties in Class 0 protostars, and their comparison to synthetic observations of MHD simulations of protostellar collapse, we aim to transform our understanding of:
1) The long-standing problem of angular momentum in star formation
2) The origin of the stellar initial mass function
3) The formation of multiple stellar systems and circumstellar disks around young stellar objects (YSOs)
Not only this project will enable a major leap forward in our understanding of low-mass star formation, answering yet unexplored questions with innovative methods, but it will also allow to spread the expertise in interpreting high-angular resolution (sub-)mm polarization data. Although characterizing magnetic fields in astrophysical structures represents the next frontier in many fields (solar physics, evolved stars, compact objects, galactic nuclei are a few examples), only a handful of astronomers in the EU community are familiar with interferometric polarization data, mostly because of the absence of large european facilities providing such capabilities until the recent advent of ALMA. It is now crucial to strengthen the European position in this research field by training a new generation of physicists with a strong expertise on tailoring, analyzing and interpreting high angular resolution polarization data."
Summary
"Rotation and angular momentum transport play a critical role in the formation and evolution of astrophysical objects, including the fundamental bricks of astrophysical structures: stars. Stars like our Sun form when rotating dense cores, in the interstellar medium, collapse until they eventually reach temperatures at which nuclear fusion begins; while planets, including the Earth, form in the rotationally supported disks around these same young stars. One of the major challenges of modern astrophysics is the “angular momentum problem"": observations show that a typical star-forming cloud needs to reduce its specific angular momentum by 5 to 10 orders of magnitude to form a typical star such as our Sun. It is also crucial to solve the angular momentum problem to understand the formation of protoplanetary disks, stellar binaries and the initial mass function of newly formed stars. Magnetic fields are one of the key ways of transporting angular momentum in astrophysical structures: understanding how angular momentum is transported to allow star formation requires characterizing the role of magnetic fields in shaping the dynamics of star-forming structures. The MagneticYSOs project aims at characterizing the role of magnetic field in the earliest stage of star formation, during the main accretion phase.
The simultaneous major improvements of instrumental and computational facilities provide us, for the first time, with the opportunity to confront observational information to magnetized models predictions. Polarization capabilities on the last generation of instrument in large facilities are producing sensitive observations of magnetic fields with a great level of detail, while numerical simulations of star formation are now including most of the physical ingredients for a detailed description of protostellar collapse at all the relevant scales, such as resistive MHD, radiative transfer and chemical networks. These new tools will undoubtedly lead to major discovery in the fields of planets and star formation in the coming years. It is necessary to conduct comprehensive projects able to combine theory and observations in a detailed fashion, which in turn require a collaboration with access to cutting edge observational datasets and numerical models. Through an ambitious multi-faceted program of dedicated observations probing magnetic fields (polarized dust emission and Zeeman effect maps), gas kinematics (molecular lines emission maps), ionization rates and dust properties in Class 0 protostars, and their comparison to synthetic observations of MHD simulations of protostellar collapse, we aim to transform our understanding of:
1) The long-standing problem of angular momentum in star formation
2) The origin of the stellar initial mass function
3) The formation of multiple stellar systems and circumstellar disks around young stellar objects (YSOs)
Not only this project will enable a major leap forward in our understanding of low-mass star formation, answering yet unexplored questions with innovative methods, but it will also allow to spread the expertise in interpreting high-angular resolution (sub-)mm polarization data. Although characterizing magnetic fields in astrophysical structures represents the next frontier in many fields (solar physics, evolved stars, compact objects, galactic nuclei are a few examples), only a handful of astronomers in the EU community are familiar with interferometric polarization data, mostly because of the absence of large european facilities providing such capabilities until the recent advent of ALMA. It is now crucial to strengthen the European position in this research field by training a new generation of physicists with a strong expertise on tailoring, analyzing and interpreting high angular resolution polarization data."
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym MAMLE
Project Understanding the mechanisms of human acute myeloid leukaemia (AML) evolution
Researcher (PI) Liran SHLUSH
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS4, ERC-2016-STG
Summary Acute myeloid leukemia (AML) is one of the most deadly cancers. Currently, we do not fully understand how and why AML starts or why it tends to relapse after treatment. Recent discoveries by the applicant and others have identified preleukemic stem and progenitor cells (preL-HSPCs) as the root of AML evolution. Many healthy elderly individuals carry the preleukemic mutations in their preL-HSPCs and yet, do not develop AML. It is also becoming clearer that leukemia evolution is spanning over many years but most research is focused on the late stages of the disease.
Population genetics tools are specifically suited for the study of historical evolution however such tools are not well developed in the field of somatic evolution. This proposal will integrate population genetics, stem cell and leukemia biology in order to unravel human leukemia evolution from the very early preleukemic phase to relapse.
Novel single cell population genetics tools will be used to describe the naive clonal structure of the human hematopoietic system in both health and disease. A unique cohort of half a million Europeans, who have been followed for years, will be used to understand why only a small fraction of the individuals carrying preleukemic mutations develop AML. Novel genetic analysis will be developed to study the clonal structure of blood cells, years before AML was diagnosed. A large cohort (N=100) of AML patients were collected serially over a year and will be collected until relapse. Detailed molecular and population genetics of this cohort will aid in understanding the mechanism of AML relapse and in developing novel molecular methodologies, that will allow early relapse diagnosis. AML like many other malignancies is diagnosed late in its evolutionary path. In this proposal the evolution of AML before diagnosis and before it relapses will be studied by novel population genetic tools so that the vision of early diagnosis and treatment will become reality.
Summary
Acute myeloid leukemia (AML) is one of the most deadly cancers. Currently, we do not fully understand how and why AML starts or why it tends to relapse after treatment. Recent discoveries by the applicant and others have identified preleukemic stem and progenitor cells (preL-HSPCs) as the root of AML evolution. Many healthy elderly individuals carry the preleukemic mutations in their preL-HSPCs and yet, do not develop AML. It is also becoming clearer that leukemia evolution is spanning over many years but most research is focused on the late stages of the disease.
Population genetics tools are specifically suited for the study of historical evolution however such tools are not well developed in the field of somatic evolution. This proposal will integrate population genetics, stem cell and leukemia biology in order to unravel human leukemia evolution from the very early preleukemic phase to relapse.
Novel single cell population genetics tools will be used to describe the naive clonal structure of the human hematopoietic system in both health and disease. A unique cohort of half a million Europeans, who have been followed for years, will be used to understand why only a small fraction of the individuals carrying preleukemic mutations develop AML. Novel genetic analysis will be developed to study the clonal structure of blood cells, years before AML was diagnosed. A large cohort (N=100) of AML patients were collected serially over a year and will be collected until relapse. Detailed molecular and population genetics of this cohort will aid in understanding the mechanism of AML relapse and in developing novel molecular methodologies, that will allow early relapse diagnosis. AML like many other malignancies is diagnosed late in its evolutionary path. In this proposal the evolution of AML before diagnosis and before it relapses will be studied by novel population genetic tools so that the vision of early diagnosis and treatment will become reality.
Max ERC Funding
1 750 000 €
Duration
Start date: 2017-02-01, End date: 2022-01-31
Project acronym MAMSIE
Project Mixing and Angular Momentum tranSport of massIvE stars
Researcher (PI) Conny Aerts
Host Institution (HI) KATHOLIEKE UNIVERSITEIT LEUVEN
Call Details Advanced Grant (AdG), PE9, ERC-2014-ADG
Summary With the CoRoT & Kepler data analysed, the time is optimal to move from observational asteroseismology to innovative stellar modelling of the steal factories of the Universe. With MAMSIE, we follow the footsteps of helioseismologists some 30 years after them, but this time we shall be developing inversion methods for stellar structure based on gravity-mode oscillations that probe the deep stellar interior. MAMSIE will lead to new models for a variety of single and binary stars with masses between 3 and 30 M⊙ whose space photometry and high-resolution spectroscopy reveal sufficient seismic information on their gravity modes to invert the frequencies and compute the stars’ structure. In contrast to the conventional theoretical approach to stellar evolution, the data-driven approach of MAMSIE will allow us to include angular momentum transport due to internal gravity waves, as well as mixing prescriptions for turbulent entrainment, from coupling of the output of 3D hydrodynamical simulations of these phenomena to specialised seismic observables of relevance for massive stars. Our sample includes slow and fast rotators, with and without a magnetic field, with and without a stellar wind. The new models will be placed in an evolutionary context for optimal assessment of the evolution of internal rotation, angular momentum, and chemical mixing throughout stellar life of massive stars. The output of the stellar modelling will provide fundamentals for all topics in modern astrophysics that rely on massive star models. MAMSIE is overarching and will require a multidisciplinary team led by an expert in gravity-mode oscillations working in close collaboration with a 3D hydrodynamics expert; it will offer a highly competitive environment for PhD and postdoctoral research on the astrophysics of massive stars.
Summary
With the CoRoT & Kepler data analysed, the time is optimal to move from observational asteroseismology to innovative stellar modelling of the steal factories of the Universe. With MAMSIE, we follow the footsteps of helioseismologists some 30 years after them, but this time we shall be developing inversion methods for stellar structure based on gravity-mode oscillations that probe the deep stellar interior. MAMSIE will lead to new models for a variety of single and binary stars with masses between 3 and 30 M⊙ whose space photometry and high-resolution spectroscopy reveal sufficient seismic information on their gravity modes to invert the frequencies and compute the stars’ structure. In contrast to the conventional theoretical approach to stellar evolution, the data-driven approach of MAMSIE will allow us to include angular momentum transport due to internal gravity waves, as well as mixing prescriptions for turbulent entrainment, from coupling of the output of 3D hydrodynamical simulations of these phenomena to specialised seismic observables of relevance for massive stars. Our sample includes slow and fast rotators, with and without a magnetic field, with and without a stellar wind. The new models will be placed in an evolutionary context for optimal assessment of the evolution of internal rotation, angular momentum, and chemical mixing throughout stellar life of massive stars. The output of the stellar modelling will provide fundamentals for all topics in modern astrophysics that rely on massive star models. MAMSIE is overarching and will require a multidisciplinary team led by an expert in gravity-mode oscillations working in close collaboration with a 3D hydrodynamics expert; it will offer a highly competitive environment for PhD and postdoctoral research on the astrophysics of massive stars.
Max ERC Funding
2 498 941 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym MANNA
Project MacroAutophagy and Necrotic Neurodegeneration in Ageing
Researcher (PI) Nektarios TAVERNARAKIS
Host Institution (HI) IDRYMA TECHNOLOGIAS KAI EREVNAS
Call Details Advanced Grant (AdG), LS4, ERC-2015-AdG
Summary Necrosis contributes critically in devastating human pathologies such as stroke, ischemia, and age-associated neurodegenerative disorders. Ageing increases susceptibility to neurodegeneration, in diverse species ranging from the lowly nematode Caenorhabditis elegans to humans. The mechanisms that govern necrotic neurodegeneration and its modulation by ageing are poorly understood. Autophagy has been implicated in necrosis and neurodegeneration, both with pro-survival and a pro-death roles. Autophagic flux declines with age, while induction of autophagy enhances longevity under conditions such as low insulin/IGF1 signalling and dietary restriction, which extend lifespan across diverse taxa. Our recent findings indicate that organelle-specific autophagy, including mitophagy, pexophagy and nucleophagy, is an important, evolutionarily conserved, determinant of longevity. We propose to dissect the molecular underpinnings of neuron vulnerability to necrosis during ageing, focusing on cargo-specific macroautophagy. To this end, we will implement a multifaceted approach that combines the power and versatility of C. elegans genetics with advanced, in vivo neuronal imaging and microfluidics technology. Our objectives are fourfold. First, we will monitor autophagic flux of organellar cargo, during neurodegeneration, under conditions that alter lifespan and identify mediators of organelle-specific autophagy in neurons. Second, we will conduct genome-wide screens for modifiers of age-inflicted neurodegeneration. Third, we will interrogate nematode models of human neurodegenerative disorders for organelle-specific autophagy and susceptibility to necrosis, upon manipulations that alter lifespan. Fourth, we will investigate the functional conservation of key mechanisms in mammalian models of neuronal necrosis. Together, these studies will deepen our understanding of age-related neurodegeneration and provide critical insights with broad relevance to human health and quality of life.
Summary
Necrosis contributes critically in devastating human pathologies such as stroke, ischemia, and age-associated neurodegenerative disorders. Ageing increases susceptibility to neurodegeneration, in diverse species ranging from the lowly nematode Caenorhabditis elegans to humans. The mechanisms that govern necrotic neurodegeneration and its modulation by ageing are poorly understood. Autophagy has been implicated in necrosis and neurodegeneration, both with pro-survival and a pro-death roles. Autophagic flux declines with age, while induction of autophagy enhances longevity under conditions such as low insulin/IGF1 signalling and dietary restriction, which extend lifespan across diverse taxa. Our recent findings indicate that organelle-specific autophagy, including mitophagy, pexophagy and nucleophagy, is an important, evolutionarily conserved, determinant of longevity. We propose to dissect the molecular underpinnings of neuron vulnerability to necrosis during ageing, focusing on cargo-specific macroautophagy. To this end, we will implement a multifaceted approach that combines the power and versatility of C. elegans genetics with advanced, in vivo neuronal imaging and microfluidics technology. Our objectives are fourfold. First, we will monitor autophagic flux of organellar cargo, during neurodegeneration, under conditions that alter lifespan and identify mediators of organelle-specific autophagy in neurons. Second, we will conduct genome-wide screens for modifiers of age-inflicted neurodegeneration. Third, we will interrogate nematode models of human neurodegenerative disorders for organelle-specific autophagy and susceptibility to necrosis, upon manipulations that alter lifespan. Fourth, we will investigate the functional conservation of key mechanisms in mammalian models of neuronal necrosis. Together, these studies will deepen our understanding of age-related neurodegeneration and provide critical insights with broad relevance to human health and quality of life.
Max ERC Funding
2 254 109 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym Mars through time
Project Modeling the past climates of planet Mars to understand its geology, its habitability and its evolution
Researcher (PI) Francois FORGET
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), PE9, ERC-2018-ADG
Summary Over the past decades, the robotic exploration of the planet Mars has produced a wealth of geological observations. They show that Mars has not always been the desert planet of today. It has seen eras conducive to rivers and lakes, ice ages, and even periods with a collapsed atmosphere. These different epochs are the reason why Mars remains the objective of space agencies, as they evoke the possibility of past habitability and spectacular climate changes.
Yet, in spite of all the data, the climatic processes that have shaped Mars’ surface through time remain largely unknown. What happened on Mars? Was the Red Planet suitable for life? What explains its evolution?
The objective of this project is to develop numerical models to simulate the past environments of Mars.A completely new “Mars Evolution Model” will be created by asynchronously coupling hydrology, glacial flows and ground ice models with a new generation 3D Global Climate Model (GCM). This GCM will be derived from the one that we have previously designed to simulate present day Mars. We will radically update it using new technologies to represent the details of the surface as well as all the processes that affected Mars when its environment evolved because of the oscillations of its orbit and obliquity, during changes in the atmospheric composition, or through events like meteoritic impacts or volcanic eruptions. Notably, we will highlight the last ten millions years that have been recorded in the polar layered deposits, whose formation will be simulated for the first time realistically.
These new tools will address numerous enigmas found in Mars sciences. They will also offer a new platform to study specific processes such as the atmospheric escape through time or the chemical alteration of the soil. Furthermore, the project will test our capacity to model planetary environments and climate changes, as well as provide lessons on the evolution of terrestrial planets and the possibility of life elsewhere.
Summary
Over the past decades, the robotic exploration of the planet Mars has produced a wealth of geological observations. They show that Mars has not always been the desert planet of today. It has seen eras conducive to rivers and lakes, ice ages, and even periods with a collapsed atmosphere. These different epochs are the reason why Mars remains the objective of space agencies, as they evoke the possibility of past habitability and spectacular climate changes.
Yet, in spite of all the data, the climatic processes that have shaped Mars’ surface through time remain largely unknown. What happened on Mars? Was the Red Planet suitable for life? What explains its evolution?
The objective of this project is to develop numerical models to simulate the past environments of Mars.A completely new “Mars Evolution Model” will be created by asynchronously coupling hydrology, glacial flows and ground ice models with a new generation 3D Global Climate Model (GCM). This GCM will be derived from the one that we have previously designed to simulate present day Mars. We will radically update it using new technologies to represent the details of the surface as well as all the processes that affected Mars when its environment evolved because of the oscillations of its orbit and obliquity, during changes in the atmospheric composition, or through events like meteoritic impacts or volcanic eruptions. Notably, we will highlight the last ten millions years that have been recorded in the polar layered deposits, whose formation will be simulated for the first time realistically.
These new tools will address numerous enigmas found in Mars sciences. They will also offer a new platform to study specific processes such as the atmospheric escape through time or the chemical alteration of the soil. Furthermore, the project will test our capacity to model planetary environments and climate changes, as well as provide lessons on the evolution of terrestrial planets and the possibility of life elsewhere.
Max ERC Funding
2 493 836 €
Duration
Start date: 2019-10-01, End date: 2024-09-30
Project acronym MarsFirstWater
Project The physicochemical nature of water on early Mars
Researcher (PI) Alberto Gonzalez Fairen
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Consolidator Grant (CoG), PE9, ERC-2018-COG
Summary Concepts of large bodies of glacial ice and liquid standing water, a robust hydrological cycle, and a rich Martian history of climate change are part of the current consensus model for early Mars. However, questions still poorly constrained include: a precise understanding of the inventory of water during the first billion years of Mars history and its early evolution on both global and local scales; whether liquid or solid H2O dominated, for what duration of time and where the water resided; what were the host-rock weathering rates and patterns and the physicochemical parameters defining such interactions; what specific landforms and mineralogies were generated during those periods; and what implications all these processes had on the possible inception of life on Mars. These fundamental questions represent large uncertainties and knowledge gaps. Therefore, a quantitative understanding of the basic characteristics of water on early Mars is very much needed and is the focus of this proposal.
This application outlines a plan for my research in the next five years, and explains how I propose to fully characterize the aqueous environments of early Mars through a quantitative and truly interdisciplinary investigation. Spacecraft mission-derived datasets will be consistently used to test hypotheses through paleogeomorphological reconstructions, geochemical modeling, mineralogical studies, and astrobiological investigations. The derived results will produce hard constraints on the physical evolution, chemical alteration and habitability of surface and near-surface aqueous environments on early Mars. The planned investigations will benefit from the combination of working with first-hand data from ongoing Mars missions and with the state-of-the-art laboratory tools at the host institution. The final expected result will be a complete understanding of the physicochemical nature of water on early Mars, also opening new paths for the astrobiological exploration of the planet.
Summary
Concepts of large bodies of glacial ice and liquid standing water, a robust hydrological cycle, and a rich Martian history of climate change are part of the current consensus model for early Mars. However, questions still poorly constrained include: a precise understanding of the inventory of water during the first billion years of Mars history and its early evolution on both global and local scales; whether liquid or solid H2O dominated, for what duration of time and where the water resided; what were the host-rock weathering rates and patterns and the physicochemical parameters defining such interactions; what specific landforms and mineralogies were generated during those periods; and what implications all these processes had on the possible inception of life on Mars. These fundamental questions represent large uncertainties and knowledge gaps. Therefore, a quantitative understanding of the basic characteristics of water on early Mars is very much needed and is the focus of this proposal.
This application outlines a plan for my research in the next five years, and explains how I propose to fully characterize the aqueous environments of early Mars through a quantitative and truly interdisciplinary investigation. Spacecraft mission-derived datasets will be consistently used to test hypotheses through paleogeomorphological reconstructions, geochemical modeling, mineralogical studies, and astrobiological investigations. The derived results will produce hard constraints on the physical evolution, chemical alteration and habitability of surface and near-surface aqueous environments on early Mars. The planned investigations will benefit from the combination of working with first-hand data from ongoing Mars missions and with the state-of-the-art laboratory tools at the host institution. The final expected result will be a complete understanding of the physicochemical nature of water on early Mars, also opening new paths for the astrobiological exploration of the planet.
Max ERC Funding
1 998 368 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym MassiveCosmo
Project Massive Gravity and Cosmology
Researcher (PI) Claudia Anna DE RHAM
Host Institution (HI) IMPERIAL COLLEGE OF SCIENCE TECHNOLOGY AND MEDICINE
Call Details Consolidator Grant (CoG), PE9, ERC-2016-COG
Summary The ambition of this research program is to challenge the nature of gravity, provide an alternative to dark energy, and pave the way towards a potential resolution of one the most tantalizing problems of physics today: The Old Cosmological Constant Problem.
As General Relativity celebrates its centennial, its predictive successes and its status as our most elegant theory of gravity are incontrovertible. Nevertheless, while the recent discovery of the late-time acceleration of the Universe is in perfect agreement with observations, the 120 orders of magnitude discrepancy between expectations and observations is one of today's most challenging puzzles and may be the sign of new physics to uncover. This conundrum has driven the development of dark energy models as alternative sources for acceleration, but many of them suffer from a similar discrepancy and require an unnatural tuning of their parameters. Despite decades of attempts, the Old Cosmological Constant Problem remains yet unsolved.
This program proposes a distinct direction to address this problem and to explain the acceleration of the Universe where the graviton, the particle carrier of gravity, has a mass, or is effectively massive. Not only will this open a new panorama for cosmology, it will also answer the fundamental question of the nature of the graviton. Signatures and constraints will be derived through astrophysical and cosmological probes.
While striving to address these fundamental challenges, the program will also elucidate new aspects of massive gravity by establishing its theoretical viability and embedding as an effective field theory. These developments will feed into new breakthroughs that have recently emerged from massive gravity.
As major missions and experiments are underway to probe dark energy and to detect gravitational waves, there is no better time to question gravity at the fundamental level, to provide alternatives to dark energy and to determine their unique signatures.
Summary
The ambition of this research program is to challenge the nature of gravity, provide an alternative to dark energy, and pave the way towards a potential resolution of one the most tantalizing problems of physics today: The Old Cosmological Constant Problem.
As General Relativity celebrates its centennial, its predictive successes and its status as our most elegant theory of gravity are incontrovertible. Nevertheless, while the recent discovery of the late-time acceleration of the Universe is in perfect agreement with observations, the 120 orders of magnitude discrepancy between expectations and observations is one of today's most challenging puzzles and may be the sign of new physics to uncover. This conundrum has driven the development of dark energy models as alternative sources for acceleration, but many of them suffer from a similar discrepancy and require an unnatural tuning of their parameters. Despite decades of attempts, the Old Cosmological Constant Problem remains yet unsolved.
This program proposes a distinct direction to address this problem and to explain the acceleration of the Universe where the graviton, the particle carrier of gravity, has a mass, or is effectively massive. Not only will this open a new panorama for cosmology, it will also answer the fundamental question of the nature of the graviton. Signatures and constraints will be derived through astrophysical and cosmological probes.
While striving to address these fundamental challenges, the program will also elucidate new aspects of massive gravity by establishing its theoretical viability and embedding as an effective field theory. These developments will feed into new breakthroughs that have recently emerged from massive gravity.
As major missions and experiments are underway to probe dark energy and to detect gravitational waves, there is no better time to question gravity at the fundamental level, to provide alternatives to dark energy and to determine their unique signatures.
Max ERC Funding
1 975 829 €
Duration
Start date: 2017-05-01, End date: 2022-04-30
Project acronym MATRICAN
Project Matrix during cancer progression
Researcher (PI) Janine Terra Erler
Host Institution (HI) KOBENHAVNS UNIVERSITET
Call Details Consolidator Grant (CoG), LS4, ERC-2015-CoG
Summary The extracellular matrix (ECM) is known to play a critical role in driving cancer progression, and yet we lack knowledge of its composition and structure. The goal of my ERC project is to investigate how alterations in biochemical composition and structural properties of the ECM during cancer progression impact on cell behaviour to drive metastasis, which is responsible for over 90% of cancer patient deaths. In order to do this, my lab has developed a method to in situ decellularise organs leaving structurally intact ECM scaffolds for subsequent analysis or for repopulation to study cell-ECM interactions in situ. We have deployed our method to decellularise primary tumour and metastatic organs in mice bearing orthotopic breast cancer tumours for subsequent quantitative global mass spectrometry (MS) proteomics, spatio-structural mapping of ECM components in 3D, and live imaging of repopulated cells. We observed fundamental alterations in ECM composition and structure between normal and tumour, and primary and metastatic tissue. We have selected two ECM components specifically upregulated in metastatic organs for subsequent validation. We discovered a marked decrease in proteins associated with fibrillogenesis in metastatic organs and will investigate the impact of this on metastatic ECM stiffness. We will decellularise organs from transgenic mouse models of breast and pancreatic cancer, at specific stages during cancer progression to determine the evolution of global ECM composition and structure, and how this impacts on cell behaviour through functional perturbation. Finally, we shall validate relevance of findings to human disease through use of human cancer lines and analysis of human patient samples. The research proposed will provide ground-breaking insight into how the ECM regulates cellular behaviour during normal and pathological conditions, and will test new strategies to combat metastasis that could be translated into the clinic to benefit cancer patients.
Summary
The extracellular matrix (ECM) is known to play a critical role in driving cancer progression, and yet we lack knowledge of its composition and structure. The goal of my ERC project is to investigate how alterations in biochemical composition and structural properties of the ECM during cancer progression impact on cell behaviour to drive metastasis, which is responsible for over 90% of cancer patient deaths. In order to do this, my lab has developed a method to in situ decellularise organs leaving structurally intact ECM scaffolds for subsequent analysis or for repopulation to study cell-ECM interactions in situ. We have deployed our method to decellularise primary tumour and metastatic organs in mice bearing orthotopic breast cancer tumours for subsequent quantitative global mass spectrometry (MS) proteomics, spatio-structural mapping of ECM components in 3D, and live imaging of repopulated cells. We observed fundamental alterations in ECM composition and structure between normal and tumour, and primary and metastatic tissue. We have selected two ECM components specifically upregulated in metastatic organs for subsequent validation. We discovered a marked decrease in proteins associated with fibrillogenesis in metastatic organs and will investigate the impact of this on metastatic ECM stiffness. We will decellularise organs from transgenic mouse models of breast and pancreatic cancer, at specific stages during cancer progression to determine the evolution of global ECM composition and structure, and how this impacts on cell behaviour through functional perturbation. Finally, we shall validate relevance of findings to human disease through use of human cancer lines and analysis of human patient samples. The research proposed will provide ground-breaking insight into how the ECM regulates cellular behaviour during normal and pathological conditions, and will test new strategies to combat metastasis that could be translated into the clinic to benefit cancer patients.
Max ERC Funding
1 997 500 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym MCTRinIA
Project Resolution Pharmacology and Physiology of MCTR in Arthritis
Researcher (PI) Jesmond Dalli
Host Institution (HI) QUEEN MARY UNIVERSITY OF LONDON
Call Details Starting Grant (StG), LS4, ERC-2015-STG
Summary Chronic inflammation may result from failure of the host response to engage pro-resolving pathways. The current treatment armamentarium for chronic inflammatory conditions may lead to immune suppression. Thus, identification of novel therapeutics that control inflammation without immune suppression will provide an attractive alternative approach. This is especially important since incidence of these conditions increases with an ageing global population. In planaria, mice, human peripheral blood and milk I recently uncovered a new family of endogenous molecules, named Maresin Conjugates in Tissue Regeneration (MCTR). These potently regulate white blood cell responses, promote the resolution of acute inflammation and accelerate tissue regeneration. The aim of this Starting Grant is to identify pathways that lead to failed resolution in inflammatory arthritis, as a prototypical chronic inflammatory condition. The hypothesis is that MCTR biosynthesis is dysregulated in inflammatory arthritis, leading to an unbridled host response, chronic inflammation and tissue destruction. This proposal will employ a multipronged approach to test this hypothesis by 1) Determining MCTR regulation in self-resolving and delayed-resolving arthritis; 2) Investigating the host protective and tissue regenerative actions of MCTRs in inflammatory arthritis; 3) Establishing the MCTR biosynthetic pathway and 4) Determining the regulation if its components during self-limited and delayed-resolving arthritis. Anticipated results will uncover novel pathways that become dysregulated during failed resolution. Results from this Starting Grant will also identify targets and new therapeutic approaches that will engage pro-resolution programs as well as tissue regeneration in conditions characterised by persistent inflammation and hence failed resolution. This will lay the basis for informed structure-activity based studies and the design of therapeutics for treatment of chronic inflammatory conditions.
Summary
Chronic inflammation may result from failure of the host response to engage pro-resolving pathways. The current treatment armamentarium for chronic inflammatory conditions may lead to immune suppression. Thus, identification of novel therapeutics that control inflammation without immune suppression will provide an attractive alternative approach. This is especially important since incidence of these conditions increases with an ageing global population. In planaria, mice, human peripheral blood and milk I recently uncovered a new family of endogenous molecules, named Maresin Conjugates in Tissue Regeneration (MCTR). These potently regulate white blood cell responses, promote the resolution of acute inflammation and accelerate tissue regeneration. The aim of this Starting Grant is to identify pathways that lead to failed resolution in inflammatory arthritis, as a prototypical chronic inflammatory condition. The hypothesis is that MCTR biosynthesis is dysregulated in inflammatory arthritis, leading to an unbridled host response, chronic inflammation and tissue destruction. This proposal will employ a multipronged approach to test this hypothesis by 1) Determining MCTR regulation in self-resolving and delayed-resolving arthritis; 2) Investigating the host protective and tissue regenerative actions of MCTRs in inflammatory arthritis; 3) Establishing the MCTR biosynthetic pathway and 4) Determining the regulation if its components during self-limited and delayed-resolving arthritis. Anticipated results will uncover novel pathways that become dysregulated during failed resolution. Results from this Starting Grant will also identify targets and new therapeutic approaches that will engage pro-resolution programs as well as tissue regeneration in conditions characterised by persistent inflammation and hence failed resolution. This will lay the basis for informed structure-activity based studies and the design of therapeutics for treatment of chronic inflammatory conditions.
Max ERC Funding
1 964 303 €
Duration
Start date: 2016-03-01, End date: 2021-02-28
Project acronym MechanoFate
Project From mechanical stress to vascular fate
Researcher (PI) Christophe, Daniel Guilluy
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Starting Grant (StG), LS4, ERC-2014-STG
Summary In the vascular system, cell phenotype and fate are driven by the mechanical environment. Whereas physiological mechanical stress defines and stabilizes normal cell phenotype, aberrant mechanical signals trigger phenotypic alteration, leading to inflammation and vascular remodelling. Despite recent advances, how mechanical cues impact gene expression to specify cell phenotype remains poorly understood.
Our hypothesis is that mechanical stresses are transmitted to the nucleus where they activate signaling pathways, which in turn regulate gene expression, but what are these mechanotransduction mechanisms occurring within the nucleus? Besides, while most vascular cells respond to mechanical force, Resident Stem Cells (RSCs) are virtually insensitive and remain undifferentiated despite constant cyclic stretch. What are the molecular mechanisms which protect RSCs from stretch-induced differentiation?
To answer these questions, we designed an interdisciplinary proposal which gathers biophysical, biochemical and genetic assays, with the following objectives: I) To determine how nuclear mechanotransduction pathways regulate vascular cell phenotype in response to mechanical cues. By combining proteomic and biophysical assays, we will identify nuclear proteins that are post-translationally modified in response to mechanical stress, then we will determine their contribution to gene expression regulation and vascular cell differentiation. II) To identify the molecular mechanisms which protect RSCs from stretch-induced differentiation. We will identify differentially expressed force-bearing structural elements in RSCs compared to more differentiated vascular cells and we will evaluate their impact on gene expression, stress transmission, RSC differentiation and blood vessel formation. The proposed project will yield new insights in different areas of life science from cell biology to potential identification of new therapeutic targets in cardiovascular and regenerative medicine.
Summary
In the vascular system, cell phenotype and fate are driven by the mechanical environment. Whereas physiological mechanical stress defines and stabilizes normal cell phenotype, aberrant mechanical signals trigger phenotypic alteration, leading to inflammation and vascular remodelling. Despite recent advances, how mechanical cues impact gene expression to specify cell phenotype remains poorly understood.
Our hypothesis is that mechanical stresses are transmitted to the nucleus where they activate signaling pathways, which in turn regulate gene expression, but what are these mechanotransduction mechanisms occurring within the nucleus? Besides, while most vascular cells respond to mechanical force, Resident Stem Cells (RSCs) are virtually insensitive and remain undifferentiated despite constant cyclic stretch. What are the molecular mechanisms which protect RSCs from stretch-induced differentiation?
To answer these questions, we designed an interdisciplinary proposal which gathers biophysical, biochemical and genetic assays, with the following objectives: I) To determine how nuclear mechanotransduction pathways regulate vascular cell phenotype in response to mechanical cues. By combining proteomic and biophysical assays, we will identify nuclear proteins that are post-translationally modified in response to mechanical stress, then we will determine their contribution to gene expression regulation and vascular cell differentiation. II) To identify the molecular mechanisms which protect RSCs from stretch-induced differentiation. We will identify differentially expressed force-bearing structural elements in RSCs compared to more differentiated vascular cells and we will evaluate their impact on gene expression, stress transmission, RSC differentiation and blood vessel formation. The proposed project will yield new insights in different areas of life science from cell biology to potential identification of new therapeutic targets in cardiovascular and regenerative medicine.
Max ERC Funding
1 498 413 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym Medulloblastoma
Project Molecularly defined models of human childhood brain tumors
Researcher (PI) Fredrik Swartling
Host Institution (HI) UPPSALA UNIVERSITET
Call Details Starting Grant (StG), LS4, ERC-2014-STG
Summary MYC proteins like MYC and MYCN are transcription factors that are mis-regulated in more than half of all types of human cancer including medulloblastoma, the most common brain malignancy in children. The two main challenges that can guide research in the field of pediatric brain tumors is improving survival and reducing long-term detriments due to treatment toxicities, especially from craniospinal radiotherapy. Medulloblastoma is suggested to originate from specific cells in the small brain, cerebellum. These brain tumors have recently been classified into four distinct molecular subgroups and subgroup-specific driver genes have been suggested. However, the precise role of such drivers in tumor initiation and their importance in specifying particular subgroups has not been sufficiently evaluated in proper cells of medulloblastoma origin.
We have generated clinically relevant animal models that carefully resemble some of the defined subgroups of medulloblastoma. In this proposal we intend to use the models to identify the specific cell type these brain tumors originates from. We also aim to refine our medulloblastoma models and develop novel models to define and study cells involved in brain metastasis and tumor recurrence; the main cause of death in brain tumor patients.
We have managed to culture normal human cerebellar stem cells and we now plan to model human medulloblastoma development by overexpressing oncogenes or silencing suppressor genes that are defined as clinically relevant medulloblastoma drivers. We will use a forward genetics screen to identify novel drivers and specifiers of various subtypes of medulloblastoma. We hope these combined efforts will help us better model human medulloblastoma formation and we expect to generate tumors that correlate well, both pathologically and molecularly, with primary cell cultures derived from medulloblastoma patients.
Summary
MYC proteins like MYC and MYCN are transcription factors that are mis-regulated in more than half of all types of human cancer including medulloblastoma, the most common brain malignancy in children. The two main challenges that can guide research in the field of pediatric brain tumors is improving survival and reducing long-term detriments due to treatment toxicities, especially from craniospinal radiotherapy. Medulloblastoma is suggested to originate from specific cells in the small brain, cerebellum. These brain tumors have recently been classified into four distinct molecular subgroups and subgroup-specific driver genes have been suggested. However, the precise role of such drivers in tumor initiation and their importance in specifying particular subgroups has not been sufficiently evaluated in proper cells of medulloblastoma origin.
We have generated clinically relevant animal models that carefully resemble some of the defined subgroups of medulloblastoma. In this proposal we intend to use the models to identify the specific cell type these brain tumors originates from. We also aim to refine our medulloblastoma models and develop novel models to define and study cells involved in brain metastasis and tumor recurrence; the main cause of death in brain tumor patients.
We have managed to culture normal human cerebellar stem cells and we now plan to model human medulloblastoma development by overexpressing oncogenes or silencing suppressor genes that are defined as clinically relevant medulloblastoma drivers. We will use a forward genetics screen to identify novel drivers and specifiers of various subtypes of medulloblastoma. We hope these combined efforts will help us better model human medulloblastoma formation and we expect to generate tumors that correlate well, both pathologically and molecularly, with primary cell cultures derived from medulloblastoma patients.
Max ERC Funding
1 497 059 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym MeerTRAP
Project Discovering Fast Transients and Pulsars with MeerKAT for Cosmology and to Test the Laws of Gravity
Researcher (PI) Benjamin STAPPERS
Host Institution (HI) THE UNIVERSITY OF MANCHESTER
Call Details Advanced Grant (AdG), PE9, ERC-2015-AdG
Summary "Short duration bursts of cosmic radio emission provide us with a potent tool for studying the extremes of physics. In the form of periodic pulses from pulsars they can be used as precision instruments to test theories of gravity, understand the emission processes and the equation of state of ultra-dense matter. In the form of transient bursts we can use them to study the physics governing explosive events such as gamma-ray bursts, merging neutron stars, annihilating black holes or hitherto unknown phenomena. Observing how these bursts are affected by their passage through the distant Universe allows us to probe its physical state. To harness the combined power of periodic and bursting radio sources to explore physics beyond that possible in a terrestrial laboratory I propose MeerTRAP: Meer (more) TRAnsients and Pulsars, a project to continuously use the MeerKAT telescope to search the radio sky for pulsars and fast transients and to rapidly and accurately locate them. Utilising the excellent sensitivity and sky coverage of MeerTRAP my team will discover many rare and scientifically important pulsar types: relativistic binaries, intermittent emitters, transitioning systems. Current radio telescopes have only explored the tip of the transients ""iceberg"" and MeerTRAP will transform our knowledge of these manifestations of extreme physics. It will detect hundreds of new bursts, which will all be well localised, allowing us to identify hosts and distances, greatly enhancing their use as cosmological probes. Localisation also enables measurement of their true fluxes, polarisation and spectral indices which are all crucial to identify their origin. To achieve this we will design, implement and exploit state-of-the-art hardware and software. We will also use the MeerLICHT optical telescope, which will track MeerKAT, to give us a crucial glimpse of the optical sky immediately before and after any radio transient to further constrain their origin and the associated physics."
Summary
"Short duration bursts of cosmic radio emission provide us with a potent tool for studying the extremes of physics. In the form of periodic pulses from pulsars they can be used as precision instruments to test theories of gravity, understand the emission processes and the equation of state of ultra-dense matter. In the form of transient bursts we can use them to study the physics governing explosive events such as gamma-ray bursts, merging neutron stars, annihilating black holes or hitherto unknown phenomena. Observing how these bursts are affected by their passage through the distant Universe allows us to probe its physical state. To harness the combined power of periodic and bursting radio sources to explore physics beyond that possible in a terrestrial laboratory I propose MeerTRAP: Meer (more) TRAnsients and Pulsars, a project to continuously use the MeerKAT telescope to search the radio sky for pulsars and fast transients and to rapidly and accurately locate them. Utilising the excellent sensitivity and sky coverage of MeerTRAP my team will discover many rare and scientifically important pulsar types: relativistic binaries, intermittent emitters, transitioning systems. Current radio telescopes have only explored the tip of the transients ""iceberg"" and MeerTRAP will transform our knowledge of these manifestations of extreme physics. It will detect hundreds of new bursts, which will all be well localised, allowing us to identify hosts and distances, greatly enhancing their use as cosmological probes. Localisation also enables measurement of their true fluxes, polarisation and spectral indices which are all crucial to identify their origin. To achieve this we will design, implement and exploit state-of-the-art hardware and software. We will also use the MeerLICHT optical telescope, which will track MeerKAT, to give us a crucial glimpse of the optical sky immediately before and after any radio transient to further constrain their origin and the associated physics."
Max ERC Funding
3 488 956 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym MeLiLoN
Project Metabolic Networks that Link Longevity to Reproduction in Response to Nutrition
Researcher (PI) Hugo Georges Roger Aguilaniu
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS4, ERC-2014-CoG
Summary In most western countries, life expectancy is increasing by 3 months a year. As the average age of
the population increases, so too does the prevalence of age-related diseases such as cancer, cardiovascular
and metabolic diseases, and neurodegenerative disorders. An essential component of ageing research is to
understand the biological mechanisms that contribute to the ageing process at the cellular and molecular
levels. This has major implications not only for the treatment of age-associated diseases but also for the
promotion of healthy ageing.
Studies of experimental animals and observations in humans have identified an array of genes and
nutritional conditions that increase lifespan. However, these manipulations (genetic or nutritional) often
have detrimental effects on other biological processes; for example, reproduction, metabolism, immunity
or growth. This is an area of ageing research that has largely been ignored but that is critical to the
success of strategies intended to slow ageing and thus the onset of disease.
The primary goal of this research proposal is to understand how lifespan extension is linked to
reproduction. We will use the nematode Caenorhabditis elegans as a model organism to identify novel
conserved genes, molecules, and metabolic networks that link reproduction and longevity through
nutrition. The proposed study is based on a unique set of preliminary data that identifies the first
clear molecular links between these traits: a steroid hormone receptor and a reproduction-responsive
lipase, both of which modulate lifespan extension achieved through changes in nutrition.
Understanding the regulation and function of these genes and pathways will clarify at the molecular
level how reproduction is linked to longevity. This may ultimately lead to interventions that optimise
metabolic activity to promote healthy ageing.
Summary
In most western countries, life expectancy is increasing by 3 months a year. As the average age of
the population increases, so too does the prevalence of age-related diseases such as cancer, cardiovascular
and metabolic diseases, and neurodegenerative disorders. An essential component of ageing research is to
understand the biological mechanisms that contribute to the ageing process at the cellular and molecular
levels. This has major implications not only for the treatment of age-associated diseases but also for the
promotion of healthy ageing.
Studies of experimental animals and observations in humans have identified an array of genes and
nutritional conditions that increase lifespan. However, these manipulations (genetic or nutritional) often
have detrimental effects on other biological processes; for example, reproduction, metabolism, immunity
or growth. This is an area of ageing research that has largely been ignored but that is critical to the
success of strategies intended to slow ageing and thus the onset of disease.
The primary goal of this research proposal is to understand how lifespan extension is linked to
reproduction. We will use the nematode Caenorhabditis elegans as a model organism to identify novel
conserved genes, molecules, and metabolic networks that link reproduction and longevity through
nutrition. The proposed study is based on a unique set of preliminary data that identifies the first
clear molecular links between these traits: a steroid hormone receptor and a reproduction-responsive
lipase, both of which modulate lifespan extension achieved through changes in nutrition.
Understanding the regulation and function of these genes and pathways will clarify at the molecular
level how reproduction is linked to longevity. This may ultimately lead to interventions that optimise
metabolic activity to promote healthy ageing.
Max ERC Funding
1 941 499 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym METABASE
Project Metagenome and Bariatric Surgery - New Avenues to Treat Metabolic Disease
Researcher (PI) Gert Fredrik Bäckhed
Host Institution (HI) GOETEBORGS UNIVERSITET
Call Details Consolidator Grant (CoG), LS4, ERC-2013-CoG
Summary Obesity and associated metabolic diseases such as type 2 diabetes are increasing worldwide and are the result of complex gene-environment interactions. Recent studies indicate that socio-demographic and environmental factors are more important for disease development than genetics, and we and others have demonstrated that the gut microbiota can be considered an environmental factor that contributes to obesity. Effective treatment for obesity remains a challenge and bariatric surgery is the only available therapy that is proven to maintain weight loss. Intriguingly, bariatric surgery also improves glucose metabolism, but the underlying molecular mechanisms for this beneficial effect are unclear. An altered gut microbiota has been linked to metabolic diseases and pilot studies indicate that the gut microbiota is also altered upon bariatric surgery; these findings suggest that some of the improved metabolic features following bariatric surgery may be mediated by altered composition of the gut microbiota. The overall goal of this proposal is to integrate clinical research with mechanistic studies in mice to determine if and how the gut microbiota mediates the beneficial effects of bariatric surgery. We will define how bariatric surgery alters the gut metagenome in humans, both at a species and at a functional level. By transferring the fecal microbiota from these patients before and after surgery to germ-free mice, we will determine if an altered gut microbiota directly modulates host metabolism. Finally, we will establish surgical methods in germ-free mice to directly test whether the beneficial effects observed following surgery require a microbiota. Increased understanding of these mechanisms may provide the basis for non-surgical treatments based on supplementation of novel probiotics to treat metabolic diseases. Follow-up work in larger clinical cohorts may also indicate how patients can be stratified to determine who would benefit the most from bariatric surgery.
Summary
Obesity and associated metabolic diseases such as type 2 diabetes are increasing worldwide and are the result of complex gene-environment interactions. Recent studies indicate that socio-demographic and environmental factors are more important for disease development than genetics, and we and others have demonstrated that the gut microbiota can be considered an environmental factor that contributes to obesity. Effective treatment for obesity remains a challenge and bariatric surgery is the only available therapy that is proven to maintain weight loss. Intriguingly, bariatric surgery also improves glucose metabolism, but the underlying molecular mechanisms for this beneficial effect are unclear. An altered gut microbiota has been linked to metabolic diseases and pilot studies indicate that the gut microbiota is also altered upon bariatric surgery; these findings suggest that some of the improved metabolic features following bariatric surgery may be mediated by altered composition of the gut microbiota. The overall goal of this proposal is to integrate clinical research with mechanistic studies in mice to determine if and how the gut microbiota mediates the beneficial effects of bariatric surgery. We will define how bariatric surgery alters the gut metagenome in humans, both at a species and at a functional level. By transferring the fecal microbiota from these patients before and after surgery to germ-free mice, we will determine if an altered gut microbiota directly modulates host metabolism. Finally, we will establish surgical methods in germ-free mice to directly test whether the beneficial effects observed following surgery require a microbiota. Increased understanding of these mechanisms may provide the basis for non-surgical treatments based on supplementation of novel probiotics to treat metabolic diseases. Follow-up work in larger clinical cohorts may also indicate how patients can be stratified to determine who would benefit the most from bariatric surgery.
Max ERC Funding
2 000 000 €
Duration
Start date: 2014-11-01, End date: 2019-10-31
Project acronym MetabolicPolycombics
Project Polycomb/Trithorax: Functional EpiGenomics Integrators for Metabolic Disease
Researcher (PI) John Andrew Pospisilik
Host Institution (HI) MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN EV
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary The last decade has seen an explosion of research into the epigenetic regulation of cell biology. Indeed, great efforts have been made to elucidate epigenetic regulation in stem cell biology, development, and within the plastic states of cancer. Current estimates place the prevalence of obesity in the range of 300 million to beyond 1 billion by the year 2030. As a critical risk factor for heart disease, diabetes and stroke, obesity currently represents one of the world’s chief economic and health care challenges. While studies have established a genetic framework for our current understanding of obesity, the contribution of several critical regulatory layers, in particular epigenetic regulation, remains poorly understood.
We recently performed the first genome-wide RNAi screen for obesity regulators in the adult fly. Intriguingly, developmental regulators scored among the most enriched pathways (Pospisilik, Cell 2010). Systematic interrogation of the 500 candidate obesity genes by tissue-specific knockdown has now identified the Polycomb-Trithorax system (PcG-Trx) as the most enriched obesity-altering pathway. Here, we propose to translate these findings to the mammalian context through completion of three aims: i. generation and characterization of 8 PcG conditional knockout mouse lines, ii. dissection of the functional roles of PcG in adipose tissue differentiation, function and disease, and iii. building of a functionally interrogated unbiased epigenetic map of the PcG-system for murine and human obesity. To achieve these goals we will combine targeted mouse genetics, complex phenotyping and state-of-the-art integrative bioinformatics.
Using this unique functional-genetics-to-epigenomics approach we will provide an unprecedented functionally validated genomics resource for obesity research worldwide, unravel an entire regulatory niveau in obesity, and if we are lucky, highlight novel therapeutic strategies for metabolic disease.
Summary
The last decade has seen an explosion of research into the epigenetic regulation of cell biology. Indeed, great efforts have been made to elucidate epigenetic regulation in stem cell biology, development, and within the plastic states of cancer. Current estimates place the prevalence of obesity in the range of 300 million to beyond 1 billion by the year 2030. As a critical risk factor for heart disease, diabetes and stroke, obesity currently represents one of the world’s chief economic and health care challenges. While studies have established a genetic framework for our current understanding of obesity, the contribution of several critical regulatory layers, in particular epigenetic regulation, remains poorly understood.
We recently performed the first genome-wide RNAi screen for obesity regulators in the adult fly. Intriguingly, developmental regulators scored among the most enriched pathways (Pospisilik, Cell 2010). Systematic interrogation of the 500 candidate obesity genes by tissue-specific knockdown has now identified the Polycomb-Trithorax system (PcG-Trx) as the most enriched obesity-altering pathway. Here, we propose to translate these findings to the mammalian context through completion of three aims: i. generation and characterization of 8 PcG conditional knockout mouse lines, ii. dissection of the functional roles of PcG in adipose tissue differentiation, function and disease, and iii. building of a functionally interrogated unbiased epigenetic map of the PcG-system for murine and human obesity. To achieve these goals we will combine targeted mouse genetics, complex phenotyping and state-of-the-art integrative bioinformatics.
Using this unique functional-genetics-to-epigenomics approach we will provide an unprecedented functionally validated genomics resource for obesity research worldwide, unravel an entire regulatory niveau in obesity, and if we are lucky, highlight novel therapeutic strategies for metabolic disease.
Max ERC Funding
1 654 430 €
Duration
Start date: 2011-12-01, End date: 2016-11-30
Project acronym METABOLOMIRS
Project Elucidation of MicroRNAs as Regulators of Metabolism and Targets for Therapeutic Intervention
Researcher (PI) Markus Stoffel
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), LS4, ERC-2009-AdG
Summary Small RNA-mediated regulation of gene expression is a recent addition to fundamental gene regulatory mechanisms that directly or indirectly affect possibly every gene of a eukaryotic genome. The predominant sources of small RNA in somatic tissues are microRNA genes that encode short dsRNA hairpins of evolutionary conserved sequence. Disorders of metabolism, such as obesity and type 2 diabetes are poorly understood at a molecular level. In this application we propose to explore if miRNA regulatory networks play a role in these diseases. We will employ state of the art methods for identification of small RNAs and their regulated targets and use biochemical, cell and animal model systems to study the detailed molecular mechanisms of metabolic gene regulation by miRNAs. In addition, we will investigate the underlying principles of how RNAs are taken up by cells and develop methods that will improve delivery of miRNA mimetics or inhibitors through cell-specific uptake. The specific aims of this study are: Aim 1: To define the small regulatory miRNA content of liver, muscle and adipose tissue that are associated with abnormal glucose and lipid homeostasis and to dissect the underlying molecular pathways that govern their expression. Aim 2: To characterize the functions of miRNAs in insulin resistance, glucose uptake and production, fatty acid oxidation and lipogenesis. Aim 3: To identify factors and dissect the pathways that regulate RNA uptake by cells and to develop novel pharmacological treatment strategies to manipulate miRNA-expression. Together, this proposal will shed light on the function that miRNA regulatory networks play in metabolism and in the pathophysiology of obesity/type 2 diabetes. In addition, these studies will contribute to the development of new RNA delivery technologies that are urgently needed as experimental tools as well as for novel therapeutic strategies.
Summary
Small RNA-mediated regulation of gene expression is a recent addition to fundamental gene regulatory mechanisms that directly or indirectly affect possibly every gene of a eukaryotic genome. The predominant sources of small RNA in somatic tissues are microRNA genes that encode short dsRNA hairpins of evolutionary conserved sequence. Disorders of metabolism, such as obesity and type 2 diabetes are poorly understood at a molecular level. In this application we propose to explore if miRNA regulatory networks play a role in these diseases. We will employ state of the art methods for identification of small RNAs and their regulated targets and use biochemical, cell and animal model systems to study the detailed molecular mechanisms of metabolic gene regulation by miRNAs. In addition, we will investigate the underlying principles of how RNAs are taken up by cells and develop methods that will improve delivery of miRNA mimetics or inhibitors through cell-specific uptake. The specific aims of this study are: Aim 1: To define the small regulatory miRNA content of liver, muscle and adipose tissue that are associated with abnormal glucose and lipid homeostasis and to dissect the underlying molecular pathways that govern their expression. Aim 2: To characterize the functions of miRNAs in insulin resistance, glucose uptake and production, fatty acid oxidation and lipogenesis. Aim 3: To identify factors and dissect the pathways that regulate RNA uptake by cells and to develop novel pharmacological treatment strategies to manipulate miRNA-expression. Together, this proposal will shed light on the function that miRNA regulatory networks play in metabolism and in the pathophysiology of obesity/type 2 diabetes. In addition, these studies will contribute to the development of new RNA delivery technologies that are urgently needed as experimental tools as well as for novel therapeutic strategies.
Max ERC Funding
2 021 235 €
Duration
Start date: 2010-07-01, End date: 2015-06-30
Project acronym METABOMIT
Project Metabolic consequences of mitochondrial dysfunction
Researcher (PI) Anu Elina Wartiovaara
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Advanced Grant (AdG), LS4, ERC-2010-AdG_20100317
Summary This proposal aims to clarify mitochondrial contribution to obesity and thinness, using carefully characterized mitochondrial disease and obese patient materials, and genetically modified disease models. Manifestations of mitochondrial respiratory chain (RC) defects range from infantile multisystem disorders to adult-onset myopathies or neurodegeneration, and even aging-related wasting. Why defects in oxidative ATP production can lead to such variety of manifestations and tissue specificity is unknown. We have previously identified a number of gene defects that lead to RC disorders. In addition to neurological symptoms, these patients often show various metabolic manifestations: specific gene defects associate with short stature and thinness, whereas others with metabolic syndrome or obesity. This implies that specific mitochondrial defects can have opposing effects for fat storage or utilization. The involved pathways may contribute to mitochondrial disease progression, but are unknown.
We propose to a) undertake a major clinical study on genetically defined, obese or thin, mitochondrial patients, and examine their metabolic phenotype in finest detail. These data will be compared to those from normal obesity, to search for common mechanisms between mitochondrial and general obesity. b) generate a set of disease models for mitochondrial disorders associated with obesity, and knock-out models for specific signallers for crossing with the disease models. c) identify in detail the involved regulatory pathways, and utilize these for searching chemical compounds that could modulate the response, and have therapeutic potential. The project has potential for major breakthroughs in the fields of mitochondrial disease pathogenesis and treatment, neurodegeneration and obesity.
Summary
This proposal aims to clarify mitochondrial contribution to obesity and thinness, using carefully characterized mitochondrial disease and obese patient materials, and genetically modified disease models. Manifestations of mitochondrial respiratory chain (RC) defects range from infantile multisystem disorders to adult-onset myopathies or neurodegeneration, and even aging-related wasting. Why defects in oxidative ATP production can lead to such variety of manifestations and tissue specificity is unknown. We have previously identified a number of gene defects that lead to RC disorders. In addition to neurological symptoms, these patients often show various metabolic manifestations: specific gene defects associate with short stature and thinness, whereas others with metabolic syndrome or obesity. This implies that specific mitochondrial defects can have opposing effects for fat storage or utilization. The involved pathways may contribute to mitochondrial disease progression, but are unknown.
We propose to a) undertake a major clinical study on genetically defined, obese or thin, mitochondrial patients, and examine their metabolic phenotype in finest detail. These data will be compared to those from normal obesity, to search for common mechanisms between mitochondrial and general obesity. b) generate a set of disease models for mitochondrial disorders associated with obesity, and knock-out models for specific signallers for crossing with the disease models. c) identify in detail the involved regulatory pathways, and utilize these for searching chemical compounds that could modulate the response, and have therapeutic potential. The project has potential for major breakthroughs in the fields of mitochondrial disease pathogenesis and treatment, neurodegeneration and obesity.
Max ERC Funding
2 500 000 €
Duration
Start date: 2011-06-01, End date: 2016-05-31
Project acronym MetaFlex
Project Metabolic flexibility: breaking down food effectively to prolong life
Researcher (PI) Richardus Hendricus Leonardus Houtkooper
Host Institution (HI) ACADEMISCH MEDISCH CENTRUM BIJ DE UNIVERSITEIT VAN AMSTERDAM
Call Details Starting Grant (StG), LS4, ERC-2014-STG
Summary Aging has long been considered a passive process. More recently studies have defined an important, active role for metabolic pathways in aging and age-related diseases. I have previously demonstrated a marked dysregulation of fat metabolism in aged mice that contributes to their overweight and glucose intolerance. Here, I propose a model that links healthy aging to efficient processing of nutrients, a state termed metabolic flexibility: reducing protein or carbohydrate metabolism will strongly stimulate fat breakdown. I suggest that improved metabolic flexibility will thus prevent the accumulation of lipids and protect against its detrimental effects.
In this project, I aim to elucidate how nutrient breakdown is regulated and can be adapted to improve metabolic flexibility and promote healthy aging. I will use C. elegans, as well as mammalian models and human population studies. Specifically, I aim to (1) dissect the molecular actors of metabolic aging pathways; (2) identify genes that translate nutritional cues to lifespan variation; (3) find novel genetic regulators that prevent toxicity and accelerated aging caused by fat-rich diets; (4) identify associations between variants in genes involved in metabolic flexibility and aging phenotypes in humans.
This set of experiments should clarify the role of nutrient breakdown and metabolic flexibility in aging. Better understanding of these processes can lead to a prolonged healthy state of aged individuals.
Summary
Aging has long been considered a passive process. More recently studies have defined an important, active role for metabolic pathways in aging and age-related diseases. I have previously demonstrated a marked dysregulation of fat metabolism in aged mice that contributes to their overweight and glucose intolerance. Here, I propose a model that links healthy aging to efficient processing of nutrients, a state termed metabolic flexibility: reducing protein or carbohydrate metabolism will strongly stimulate fat breakdown. I suggest that improved metabolic flexibility will thus prevent the accumulation of lipids and protect against its detrimental effects.
In this project, I aim to elucidate how nutrient breakdown is regulated and can be adapted to improve metabolic flexibility and promote healthy aging. I will use C. elegans, as well as mammalian models and human population studies. Specifically, I aim to (1) dissect the molecular actors of metabolic aging pathways; (2) identify genes that translate nutritional cues to lifespan variation; (3) find novel genetic regulators that prevent toxicity and accelerated aging caused by fat-rich diets; (4) identify associations between variants in genes involved in metabolic flexibility and aging phenotypes in humans.
This set of experiments should clarify the role of nutrient breakdown and metabolic flexibility in aging. Better understanding of these processes can lead to a prolonged healthy state of aged individuals.
Max ERC Funding
1 499 446 €
Duration
Start date: 2015-04-01, End date: 2020-03-31
Project acronym MetAGEn
Project Metabolic and Genetic Regulation of Ageing
Researcher (PI) Martin Denzel
Host Institution (HI) MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN EV
Call Details Starting Grant (StG), LS4, ERC-2014-STG
Summary Ageing is a complex physiological process that affects almost all species, including humans. Despite its importance for all of us, the biology of ageing is insufficiently understood. To uncover the molecular underpinnings of ageing, I propose an interdisciplinary research program that will identify and investigate metabolic and genetic regulators of ageing.
Progressive loss of cellular homeostasis causes ageing and an age-associated decline in protein quality control has been implicated in numerous diseases, including neurodegeneration. Seeking for ways to improve protein quality, I have identified a novel longevity pathway in Caenorhabditis elegans. In a forward genetic screen, I found a link between metabolites in the hexosamine pathway and cellular protein quality control. Hexosamine pathway activation extends C. elegans lifespan, suggesting modulation of ageing by endogenous molecules.
In a first step, I will explore the mechanism by which hexosamine metabolites improve protein quality control in mammals, using cultured mammalian cells and a mouse model for neurodegeneration. Preliminary data show that hexosamine pathway metabolites enhance proteolytic capacity in cells and reduce protein aggregation, suggesting conservation. Second, I will investigate molecular mechanisms that activate the hexosamine pathway to promote protein homeostasis and counter ageing. Third, I will perform a direct forward genetic screen for modulators of ageing in C. elegans. For the first time, mutagenesis and next generation sequencing can be paired in forward genetic screens to interrogate the whole genome for lifespan-extending mutations in a truly unbiased manner. This innovative approach has the potential to reveal novel modulators of the ageing process.
Taken together, this work aims to understand molecular mechanisms that maintain cellular homeostasis to slow the ageing process, and to develop a new technology to identify yet unknown genetic modulators of ageing.
Summary
Ageing is a complex physiological process that affects almost all species, including humans. Despite its importance for all of us, the biology of ageing is insufficiently understood. To uncover the molecular underpinnings of ageing, I propose an interdisciplinary research program that will identify and investigate metabolic and genetic regulators of ageing.
Progressive loss of cellular homeostasis causes ageing and an age-associated decline in protein quality control has been implicated in numerous diseases, including neurodegeneration. Seeking for ways to improve protein quality, I have identified a novel longevity pathway in Caenorhabditis elegans. In a forward genetic screen, I found a link between metabolites in the hexosamine pathway and cellular protein quality control. Hexosamine pathway activation extends C. elegans lifespan, suggesting modulation of ageing by endogenous molecules.
In a first step, I will explore the mechanism by which hexosamine metabolites improve protein quality control in mammals, using cultured mammalian cells and a mouse model for neurodegeneration. Preliminary data show that hexosamine pathway metabolites enhance proteolytic capacity in cells and reduce protein aggregation, suggesting conservation. Second, I will investigate molecular mechanisms that activate the hexosamine pathway to promote protein homeostasis and counter ageing. Third, I will perform a direct forward genetic screen for modulators of ageing in C. elegans. For the first time, mutagenesis and next generation sequencing can be paired in forward genetic screens to interrogate the whole genome for lifespan-extending mutations in a truly unbiased manner. This innovative approach has the potential to reveal novel modulators of the ageing process.
Taken together, this work aims to understand molecular mechanisms that maintain cellular homeostasis to slow the ageing process, and to develop a new technology to identify yet unknown genetic modulators of ageing.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-08-01, End date: 2020-07-31
Project acronym MetaMeta
Project Metastability of proteins during tumor metastasis
Researcher (PI) Ramunas Martynas Vabulas
Host Institution (HI) JOHANN WOLFGANG GOETHE-UNIVERSITATFRANKFURT AM MAIN
Call Details Starting Grant (StG), LS4, ERC-2012-StG_20111109
Summary Mutational heterogeneity bestows tumors with the phenotypic plasticity and adaptability required for expansion. On the other hand, mutations destabilize proteins – lower stability (metastability) of the tumor proteome must be the inevitable consequence. We set out to systematically investigate this biochemical aspect of metastasis aiming to uncover and therapeutically exploit specific vulnerabilities resulting from protein destabilization. We will approach this goal by cataloging associations between metastasis-promoting proteins and molecular chaperones. Chaperones are obvious candidates to stabilize the proteome, therefore we will prepare a BAC-based mouse model of metastasis, where the contribution of 63 chaperones, comprising the entire murine HSP70 superfamily, to metastasis development will be individually investigated. The role of metastasis-relevant chaperones at the molecular level will be elucidated using mass spectrometry, complemented by next-generation sequencing of metastatic exome. In parallel, a novel proteomics-based method to evaluate aberrant complex formation in tumor cells will be established.
Because of the high heterogeneity of cancer, molecularly tailored and combined therapies are needed. To this end, we will capitalize on insights regarding the role of chaperones in metastasis by identifying proteasomal degradation activators able to support or replace the activity of individual chaperones from the HSP70 superfamily. Finally, we will validate the potential of combined, yet specific manipulation of the folding and degradation machineries to suppress metastasis development.
Summary
Mutational heterogeneity bestows tumors with the phenotypic plasticity and adaptability required for expansion. On the other hand, mutations destabilize proteins – lower stability (metastability) of the tumor proteome must be the inevitable consequence. We set out to systematically investigate this biochemical aspect of metastasis aiming to uncover and therapeutically exploit specific vulnerabilities resulting from protein destabilization. We will approach this goal by cataloging associations between metastasis-promoting proteins and molecular chaperones. Chaperones are obvious candidates to stabilize the proteome, therefore we will prepare a BAC-based mouse model of metastasis, where the contribution of 63 chaperones, comprising the entire murine HSP70 superfamily, to metastasis development will be individually investigated. The role of metastasis-relevant chaperones at the molecular level will be elucidated using mass spectrometry, complemented by next-generation sequencing of metastatic exome. In parallel, a novel proteomics-based method to evaluate aberrant complex formation in tumor cells will be established.
Because of the high heterogeneity of cancer, molecularly tailored and combined therapies are needed. To this end, we will capitalize on insights regarding the role of chaperones in metastasis by identifying proteasomal degradation activators able to support or replace the activity of individual chaperones from the HSP70 superfamily. Finally, we will validate the potential of combined, yet specific manipulation of the folding and degradation machineries to suppress metastasis development.
Max ERC Funding
1 366 800 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym METANICHE
Project Regulation of bone metastases by age-associated angiocrine signals
Researcher (PI) Anjali KUSUMBE
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Starting Grant (StG), LS4, ERC-2018-STG
Summary Blood vessels form a versatile transport network and provide inductive signals called angiocrine factors to regulate tissue-specific functions. Blood vessels in bone are heterogeneous with distinct capillary subtypes that exhibit remarkable alterations with age. Bone is the most prevalent site of metastasis, and ageing is linked to the reactivation of dormant tumor cells (dorTCs) and metastatic relapse. Bone remodeling processes are also associated with metastatic relapse. Here, I will define the role of distinct vascular niches in regulating the fate of DTCs in bone. Finally, I will unravel the age-related angiocrine factors and identify key angiocrine signals that drive the reactivation of dorTCs. I will employ a powerful combination of advanced 3D, intravital, and whole body imaging, cell specific-inducible mouse genetics, transcriptional profiling and bioinformatics in an unprecedented manner to achieve my goals. New cutting-edge techniques such as advanced 3D and 4D bone imaging are important aspects of my proposal. I will also define the role of highly promising novel candidate age-related angiocrine signals with sophisticated inducible endothelial-specific humanised mouse models. My work will break new ground by unraveling a repertoire of age-related angiocrine factors and will contribute to a wider scientific community in bone, blood, and age-related diseases. This interdisciplinary work at the frontiers of bone, cancer and vascular biology will provide the first conceptual link between vascular ageing and bone metastasis and will contribute towards the development of therapeutic strategies for targeting DTCs in bone.
Summary
Blood vessels form a versatile transport network and provide inductive signals called angiocrine factors to regulate tissue-specific functions. Blood vessels in bone are heterogeneous with distinct capillary subtypes that exhibit remarkable alterations with age. Bone is the most prevalent site of metastasis, and ageing is linked to the reactivation of dormant tumor cells (dorTCs) and metastatic relapse. Bone remodeling processes are also associated with metastatic relapse. Here, I will define the role of distinct vascular niches in regulating the fate of DTCs in bone. Finally, I will unravel the age-related angiocrine factors and identify key angiocrine signals that drive the reactivation of dorTCs. I will employ a powerful combination of advanced 3D, intravital, and whole body imaging, cell specific-inducible mouse genetics, transcriptional profiling and bioinformatics in an unprecedented manner to achieve my goals. New cutting-edge techniques such as advanced 3D and 4D bone imaging are important aspects of my proposal. I will also define the role of highly promising novel candidate age-related angiocrine signals with sophisticated inducible endothelial-specific humanised mouse models. My work will break new ground by unraveling a repertoire of age-related angiocrine factors and will contribute to a wider scientific community in bone, blood, and age-related diseases. This interdisciplinary work at the frontiers of bone, cancer and vascular biology will provide the first conceptual link between vascular ageing and bone metastasis and will contribute towards the development of therapeutic strategies for targeting DTCs in bone.
Max ERC Funding
1 496 613 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym MetaRegulation
Project Metabolic regulation of metastatic growth
Researcher (PI) Sarah-Maria FENDT
Host Institution (HI) VIB
Call Details Consolidator Grant (CoG), LS4, ERC-2017-COG
Summary Metastatic growth of cancer cells requires extracellular matrix (ECM) production. The current understanding is that transcription factors regulate ECM production and thus metastatic growth by increasing the expression of collagen prolyl 4-hydroxylase (CP4H). In contrast, we recently discovered that metabolism regulates CP4H activity independently of the known transcription factors. Specifically, we found that loss of pyruvate metabolism inhibits CP4H activity and consequently ECM–dependent breast cancer cell growth. Based on this discovery we propose the novel concept that metabolism regulates metastatic growth by increasing ECM production.
In this project we will investigate the following questions: 1) What is the mechanism by which pyruvate regulates CP4H activity in breast cancer cells? To address this question we will investigate pyruvate metabolism and ECM production in 3D cultures of various breast cancer cell lines using 13C tracer analysis, metabolomics, and two-photon microscopy based ECM visualization. 2) How can this novel metabolic regulation be exploited to inhibit breast cancer-derived lung metastases growth? To address this question we will inhibit pyruvate metabolism in metastatic breast cancer mouse models using genetically modified cells and small molecules in combination with immuno- and chemotherapy. 3) How can this novel regulation be translated to different metastatic sites and cancers of different origin? To address this question we will determine the in vivo metabolism of breast cancer-, lung cancer-, and melanoma-derived liver and lung metastases (using metabolomics and 13C tracer analysis), and link it to ECM production (using two-photon microscopy based ECM visualization).
With this project we will deliver a novel concept by which metabolism regulates metastatic growth. In a long-term perspective we expect that targeting this novel metabolic regulation will pave the way for an unexplored approach to treat cancer metastases.
Summary
Metastatic growth of cancer cells requires extracellular matrix (ECM) production. The current understanding is that transcription factors regulate ECM production and thus metastatic growth by increasing the expression of collagen prolyl 4-hydroxylase (CP4H). In contrast, we recently discovered that metabolism regulates CP4H activity independently of the known transcription factors. Specifically, we found that loss of pyruvate metabolism inhibits CP4H activity and consequently ECM–dependent breast cancer cell growth. Based on this discovery we propose the novel concept that metabolism regulates metastatic growth by increasing ECM production.
In this project we will investigate the following questions: 1) What is the mechanism by which pyruvate regulates CP4H activity in breast cancer cells? To address this question we will investigate pyruvate metabolism and ECM production in 3D cultures of various breast cancer cell lines using 13C tracer analysis, metabolomics, and two-photon microscopy based ECM visualization. 2) How can this novel metabolic regulation be exploited to inhibit breast cancer-derived lung metastases growth? To address this question we will inhibit pyruvate metabolism in metastatic breast cancer mouse models using genetically modified cells and small molecules in combination with immuno- and chemotherapy. 3) How can this novel regulation be translated to different metastatic sites and cancers of different origin? To address this question we will determine the in vivo metabolism of breast cancer-, lung cancer-, and melanoma-derived liver and lung metastases (using metabolomics and 13C tracer analysis), and link it to ECM production (using two-photon microscopy based ECM visualization).
With this project we will deliver a novel concept by which metabolism regulates metastatic growth. In a long-term perspective we expect that targeting this novel metabolic regulation will pave the way for an unexplored approach to treat cancer metastases.
Max ERC Funding
2 000 000 €
Duration
Start date: 2018-06-01, End date: 2023-05-31