Project acronym 3DBrainStrom
Project Brain metastases: Deciphering tumor-stroma interactions in three dimensions for the rational design of nanomedicines
Researcher (PI) Ronit Satchi Fainaro
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Advanced Grant (AdG), LS7, ERC-2018-ADG
Summary Brain metastases represent a major therapeutic challenge. Despite significant breakthroughs in targeted therapies, survival rates of patients with brain metastases remain poor. Nowadays, discovery, development and evaluation of new therapies are performed on human cancer cells grown in 2D on rigid plastic plates followed by in vivo testing in immunodeficient mice. These experimental settings are lacking and constitute a fundamental hurdle for the translation of preclinical discoveries into clinical practice. We propose to establish 3D-printed models of brain metastases (Aim 1), which include brain extracellular matrix, stroma and serum containing immune cells flowing in functional tumor vessels. Our unique models better capture the clinical physio-mechanical tissue properties, signaling pathways, hemodynamics and drug responsiveness. Using our 3D-printed models, we aim to develop two new fronts for identifying novel clinically-relevant molecular drivers (Aim 2) followed by the development of precision nanomedicines (Aim 3). We will exploit our vast experience in anticancer nanomedicines to design three therapeutic approaches that target various cellular compartments involved in brain metastases: 1) Prevention of brain metastatic colonization using targeted nano-vaccines, which elicit antitumor immune response; 2) Intervention of tumor-brain stroma cells crosstalk when brain micrometastases establish; 3) Regression of macrometastatic disease by selectively targeting tumor cells. These approaches will materialize using our libraries of polymeric nanocarriers that selectively accumulate in tumors.
This project will result in a paradigm shift by generating new preclinical cancer models that will bridge the translational gap in cancer therapeutics. The insights and tumor-stroma-targeted nanomedicines developed here will pave the way for prediction of patient outcome, revolutionizing our perception of tumor modelling and consequently the way we prevent and treat cancer.
Summary
Brain metastases represent a major therapeutic challenge. Despite significant breakthroughs in targeted therapies, survival rates of patients with brain metastases remain poor. Nowadays, discovery, development and evaluation of new therapies are performed on human cancer cells grown in 2D on rigid plastic plates followed by in vivo testing in immunodeficient mice. These experimental settings are lacking and constitute a fundamental hurdle for the translation of preclinical discoveries into clinical practice. We propose to establish 3D-printed models of brain metastases (Aim 1), which include brain extracellular matrix, stroma and serum containing immune cells flowing in functional tumor vessels. Our unique models better capture the clinical physio-mechanical tissue properties, signaling pathways, hemodynamics and drug responsiveness. Using our 3D-printed models, we aim to develop two new fronts for identifying novel clinically-relevant molecular drivers (Aim 2) followed by the development of precision nanomedicines (Aim 3). We will exploit our vast experience in anticancer nanomedicines to design three therapeutic approaches that target various cellular compartments involved in brain metastases: 1) Prevention of brain metastatic colonization using targeted nano-vaccines, which elicit antitumor immune response; 2) Intervention of tumor-brain stroma cells crosstalk when brain micrometastases establish; 3) Regression of macrometastatic disease by selectively targeting tumor cells. These approaches will materialize using our libraries of polymeric nanocarriers that selectively accumulate in tumors.
This project will result in a paradigm shift by generating new preclinical cancer models that will bridge the translational gap in cancer therapeutics. The insights and tumor-stroma-targeted nanomedicines developed here will pave the way for prediction of patient outcome, revolutionizing our perception of tumor modelling and consequently the way we prevent and treat cancer.
Max ERC Funding
2 353 125 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym 4D-PET
Project Innovative PET scanner for dynamic imaging
Researcher (PI) José María BENLLOCH BAVIERA
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Advanced Grant (AdG), LS7, ERC-2015-AdG
Summary The main objective of 4D-PET is to develop an innovative whole-body PET scanner based in a new detector concept that stores 3D position and time of every single gamma interaction with unprecedented resolution. The combination of scanner geometrical design and high timing resolution will enable developing a full sequence of all gamma-ray interactions inside the scanner, including Compton interactions, like in a 3D movie. 4D-PET fully exploits Time Of Flight (TOF) information to obtain a better image quality and to increase scanner sensitivity, through the inclusion in the image formation of all Compton events occurring inside the detector, which are always rejected in state-of-the-art PET scanners. The new PET design will radically improve state-of-the-art PET performance features, overcoming limitations of current PET technology and opening up new diagnostic venues and very valuable physiological information
Summary
The main objective of 4D-PET is to develop an innovative whole-body PET scanner based in a new detector concept that stores 3D position and time of every single gamma interaction with unprecedented resolution. The combination of scanner geometrical design and high timing resolution will enable developing a full sequence of all gamma-ray interactions inside the scanner, including Compton interactions, like in a 3D movie. 4D-PET fully exploits Time Of Flight (TOF) information to obtain a better image quality and to increase scanner sensitivity, through the inclusion in the image formation of all Compton events occurring inside the detector, which are always rejected in state-of-the-art PET scanners. The new PET design will radically improve state-of-the-art PET performance features, overcoming limitations of current PET technology and opening up new diagnostic venues and very valuable physiological information
Max ERC Funding
2 048 386 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym 5D Heart Patch
Project A Functional, Mature In vivo Human Ventricular Muscle Patch for Cardiomyopathy
Researcher (PI) Kenneth Randall Chien
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Developing new therapeutic strategies for heart regeneration is a major goal for cardiac biology and medicine. While cardiomyocytes can be generated from human pluripotent stem (hPSC) cells in vitro, it has proven difficult to use these cells to generate a large scale, mature human heart ventricular muscle graft on the injured heart in vivo. The central objective of this proposal is to optimize the generation of a large-scale pure, fully functional human ventricular muscle patch in vivo through the self-assembly of purified human ventricular progenitors and the localized expression of defined paracrine factors that drive their expansion, differentiation, vascularization, matrix formation, and maturation. Recently, we have found that purified hPSC-derived ventricular progenitors (HVPs) can self-assemble in vivo on the epicardial surface into a 3D vascularized, and functional ventricular patch with its own extracellular matrix via a cell autonomous pathway. A two-step protocol and FACS purification of HVP receptors can generate billions of pure HVPs- The current proposal will lead to the identification of defined paracrine pathways to enhance the survival, grafting/implantation, expansion, differentiation, matrix formation, vascularization and maturation of the graft in vivo. We will captalize on our unique HVP system and our novel modRNA technology to deliver therapeutic strategies by using the in vivo human ventricular muscle to model in vivo arrhythmogenic cardiomyopathy, and optimize the ability of the graft to compensate for the massive loss of functional muscle during ischemic cardiomyopathy and post-myocardial infarction. The studies will lead to new in vivo chimeric models of human cardiac disease and an experimental paradigm to optimize organ-on-organ cardiac tissue engineers of an in vivo, functional mature ventricular patch for cardiomyopathy
Summary
Developing new therapeutic strategies for heart regeneration is a major goal for cardiac biology and medicine. While cardiomyocytes can be generated from human pluripotent stem (hPSC) cells in vitro, it has proven difficult to use these cells to generate a large scale, mature human heart ventricular muscle graft on the injured heart in vivo. The central objective of this proposal is to optimize the generation of a large-scale pure, fully functional human ventricular muscle patch in vivo through the self-assembly of purified human ventricular progenitors and the localized expression of defined paracrine factors that drive their expansion, differentiation, vascularization, matrix formation, and maturation. Recently, we have found that purified hPSC-derived ventricular progenitors (HVPs) can self-assemble in vivo on the epicardial surface into a 3D vascularized, and functional ventricular patch with its own extracellular matrix via a cell autonomous pathway. A two-step protocol and FACS purification of HVP receptors can generate billions of pure HVPs- The current proposal will lead to the identification of defined paracrine pathways to enhance the survival, grafting/implantation, expansion, differentiation, matrix formation, vascularization and maturation of the graft in vivo. We will captalize on our unique HVP system and our novel modRNA technology to deliver therapeutic strategies by using the in vivo human ventricular muscle to model in vivo arrhythmogenic cardiomyopathy, and optimize the ability of the graft to compensate for the massive loss of functional muscle during ischemic cardiomyopathy and post-myocardial infarction. The studies will lead to new in vivo chimeric models of human cardiac disease and an experimental paradigm to optimize organ-on-organ cardiac tissue engineers of an in vivo, functional mature ventricular patch for cardiomyopathy
Max ERC Funding
2 149 228 €
Duration
Start date: 2017-12-01, End date: 2022-11-30
Project acronym AAA
Project Adaptive Actin Architectures
Researcher (PI) Laurent Blanchoin
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS3, ERC-2016-ADG
Summary Although we have extensive knowledge of many important processes in cell biology, including information on many of the molecules involved and the physical interactions among them, we still do not understand most of the dynamical features that are the essence of living systems. This is particularly true for the actin cytoskeleton, a major component of the internal architecture of eukaryotic cells. In living cells, actin networks constantly assemble and disassemble filaments while maintaining an apparent stable structure, suggesting a perfect balance between the two processes. Such behaviors are called “dynamic steady states”. They confer upon actin networks a high degree of plasticity allowing them to adapt in response to external changes and enable cells to adjust to their environments. Despite their fundamental importance in the regulation of cell physiology, the basic mechanisms that control the coordinated dynamics of co-existing actin networks are poorly understood. In the AAA project, first, we will characterize the parameters that allow the coupling among co-existing actin networks at steady state. In vitro reconstituted systems will be used to control the actin nucleation patterns, the closed volume of the reaction chamber and the physical interaction of the networks. We hope to unravel the mechanism allowing the global coherence of a dynamic actin cytoskeleton. Second, we will use our unique capacity to perform dynamic micropatterning, to add or remove actin nucleation sites in real time, in order to investigate the ability of dynamic networks to adapt to changes and the role of coupled network dynamics in this emergent property. In this part, in vitro experiments will be complemented by the analysis of actin network remodeling in living cells. In the end, our project will provide a comprehensive understanding of how the adaptive response of the cytoskeleton derives from the complex interplay between its biochemical, structural and mechanical properties.
Summary
Although we have extensive knowledge of many important processes in cell biology, including information on many of the molecules involved and the physical interactions among them, we still do not understand most of the dynamical features that are the essence of living systems. This is particularly true for the actin cytoskeleton, a major component of the internal architecture of eukaryotic cells. In living cells, actin networks constantly assemble and disassemble filaments while maintaining an apparent stable structure, suggesting a perfect balance between the two processes. Such behaviors are called “dynamic steady states”. They confer upon actin networks a high degree of plasticity allowing them to adapt in response to external changes and enable cells to adjust to their environments. Despite their fundamental importance in the regulation of cell physiology, the basic mechanisms that control the coordinated dynamics of co-existing actin networks are poorly understood. In the AAA project, first, we will characterize the parameters that allow the coupling among co-existing actin networks at steady state. In vitro reconstituted systems will be used to control the actin nucleation patterns, the closed volume of the reaction chamber and the physical interaction of the networks. We hope to unravel the mechanism allowing the global coherence of a dynamic actin cytoskeleton. Second, we will use our unique capacity to perform dynamic micropatterning, to add or remove actin nucleation sites in real time, in order to investigate the ability of dynamic networks to adapt to changes and the role of coupled network dynamics in this emergent property. In this part, in vitro experiments will be complemented by the analysis of actin network remodeling in living cells. In the end, our project will provide a comprehensive understanding of how the adaptive response of the cytoskeleton derives from the complex interplay between its biochemical, structural and mechanical properties.
Max ERC Funding
2 349 898 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym ACTOMYOSIN RING
Project Understanding Cytokinetic Actomyosin Ring Assembly Through Genetic Code Expansion, Click Chemistry, DNA origami, and in vitro Reconstitution
Researcher (PI) Mohan Balasubramanian
Host Institution (HI) THE UNIVERSITY OF WARWICK
Call Details Advanced Grant (AdG), LS3, ERC-2014-ADG
Summary The mechanism of cell division is conserved in many eukaryotes, from yeast to man. A contractile ring of filamentous actin and myosin II motors generates the force to bisect a mother cell into two daughters. The actomyosin ring is among the most complex cellular machines, comprising over 150 proteins. Understanding how these proteins organize themselves into a functional ring with appropriate contractile properties remains one of the great challenges in cell biology. Efforts to generate a comprehensive understanding of the mechanism of actomyosin ring assembly have been hampered by the lack of structural information on the arrangement of actin, myosin II, and actin modulators in the ring in its native state. Fundamental questions such as how actin filaments are assembled and organized into a ring remain actively debated. This project will investigate key issues pertaining to cytokinesis in the fission yeast Schizosaccharomyces pombe, which divides employing an actomyosin based contractile ring, using the methods of genetics, biochemistry, cellular imaging, DNA origami, genetic code expansion, and click chemistry. Specifically, we will (1) attempt to visualize actin filament assembly in live cells expressing fluorescent actin generated through synthetic biological approaches, including genetic code expansion and click chemistry (2) decipher actin filament polarity in the actomyosin ring using total internal reflection fluorescence microscopy of labelled dimeric and multimeric myosins V and VI generated through DNA origami approaches (3) address when, where, and how actin filaments for cytokinesis are assembled and organized into a ring and (4) reconstitute actin filament and functional actomyosin ring assembly in permeabilized spheroplasts and in supported bilayers. Success in the project will provide major insight into the mechanism of actomyosin ring assembly and illuminate principles behind cytoskeletal self-organization.
Summary
The mechanism of cell division is conserved in many eukaryotes, from yeast to man. A contractile ring of filamentous actin and myosin II motors generates the force to bisect a mother cell into two daughters. The actomyosin ring is among the most complex cellular machines, comprising over 150 proteins. Understanding how these proteins organize themselves into a functional ring with appropriate contractile properties remains one of the great challenges in cell biology. Efforts to generate a comprehensive understanding of the mechanism of actomyosin ring assembly have been hampered by the lack of structural information on the arrangement of actin, myosin II, and actin modulators in the ring in its native state. Fundamental questions such as how actin filaments are assembled and organized into a ring remain actively debated. This project will investigate key issues pertaining to cytokinesis in the fission yeast Schizosaccharomyces pombe, which divides employing an actomyosin based contractile ring, using the methods of genetics, biochemistry, cellular imaging, DNA origami, genetic code expansion, and click chemistry. Specifically, we will (1) attempt to visualize actin filament assembly in live cells expressing fluorescent actin generated through synthetic biological approaches, including genetic code expansion and click chemistry (2) decipher actin filament polarity in the actomyosin ring using total internal reflection fluorescence microscopy of labelled dimeric and multimeric myosins V and VI generated through DNA origami approaches (3) address when, where, and how actin filaments for cytokinesis are assembled and organized into a ring and (4) reconstitute actin filament and functional actomyosin ring assembly in permeabilized spheroplasts and in supported bilayers. Success in the project will provide major insight into the mechanism of actomyosin ring assembly and illuminate principles behind cytoskeletal self-organization.
Max ERC Funding
2 863 705 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym AIR-NB
Project Pre-natal exposure to urban AIR pollution and pre- and post-Natal Brain development
Researcher (PI) Jordi Sunyer
Host Institution (HI) FUNDACION PRIVADA INSTITUTO DE SALUD GLOBAL BARCELONA
Call Details Advanced Grant (AdG), LS7, ERC-2017-ADG
Summary Air pollution is the main urban-related environmental hazard. It appears to affect brain development, although current evidence is inadequate given the lack of studies during the most vulnerable stages of brain development and the lack of brain anatomical structure and regional connectivity data underlying these effects. Of particular interest is the prenatal period, when brain structures are forming and growing, and when the effect of in utero exposure to environmental factors may cause permanent brain injury. I and others have conducted studies focused on effects during school age which could be less profound. I postulate that: pre-natal exposure to urban air pollution during pregnancy impairs foetal and postnatal brain development, mainly by affecting myelination; these effects are at least partially mediated by translocation of airborne particulate matter to the placenta and by placental dysfunction; and prenatal exposure to air pollution impairs post-natal brain development independently of urban context and post-natal exposure to air pollution. I aim to evaluate the effect of pre-natal exposure to urban air pollution on pre- and post-natal brain structure and function by following 900 pregnant women and their neonates with contrasting levels of pre-natal exposure to air pollutants by: i) establishing a new pregnancy cohort and evaluating brain imaging (pre-natal and neo-natal brain structure, connectivity and function), and post-natal motor and cognitive development; ii) measuring total personal exposure and inhaled dose of air pollutants during specific time-windows of gestation, noise, paternal stress and other stressors, using personal samplers and sensors; iii) detecting nanoparticles in placenta and its vascular function; iv) modelling mathematical causality and mediation, including a replication study in an external cohort. The expected results will create an impulse to implement policy interventions that genuinely protect the health of urban citizens.
Summary
Air pollution is the main urban-related environmental hazard. It appears to affect brain development, although current evidence is inadequate given the lack of studies during the most vulnerable stages of brain development and the lack of brain anatomical structure and regional connectivity data underlying these effects. Of particular interest is the prenatal period, when brain structures are forming and growing, and when the effect of in utero exposure to environmental factors may cause permanent brain injury. I and others have conducted studies focused on effects during school age which could be less profound. I postulate that: pre-natal exposure to urban air pollution during pregnancy impairs foetal and postnatal brain development, mainly by affecting myelination; these effects are at least partially mediated by translocation of airborne particulate matter to the placenta and by placental dysfunction; and prenatal exposure to air pollution impairs post-natal brain development independently of urban context and post-natal exposure to air pollution. I aim to evaluate the effect of pre-natal exposure to urban air pollution on pre- and post-natal brain structure and function by following 900 pregnant women and their neonates with contrasting levels of pre-natal exposure to air pollutants by: i) establishing a new pregnancy cohort and evaluating brain imaging (pre-natal and neo-natal brain structure, connectivity and function), and post-natal motor and cognitive development; ii) measuring total personal exposure and inhaled dose of air pollutants during specific time-windows of gestation, noise, paternal stress and other stressors, using personal samplers and sensors; iii) detecting nanoparticles in placenta and its vascular function; iv) modelling mathematical causality and mediation, including a replication study in an external cohort. The expected results will create an impulse to implement policy interventions that genuinely protect the health of urban citizens.
Max ERC Funding
2 499 992 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym AngioBone
Project Angiogenic growth, specialization, ageing and regeneration
of bone vessels
Researcher (PI) Ralf Heinrich Adams
Host Institution (HI) WESTFAELISCHE WILHELMS-UNIVERSITAET MUENSTER
Call Details Advanced Grant (AdG), LS3, ERC-2013-ADG
Summary The skeleton and the sinusoidal vasculature form a functional unit with great relevance in health, regeneration, and disease. Currently, fundamental aspects of sinusoidal vessel growth, specialization, arteriovenous organization and the consequences for tissue perfusion, or the changes occurring during ageing remain unknown. Our preliminary data indicate that key principles of bone vascularization and the role of molecular regulators are highly distinct from other organs. I therefore propose to use powerful combination of mouse genetics, fate mapping, transcriptional profiling, computational biology, confocal and two-photon microscopy, micro-CT and PET imaging, biochemistry and cell biology to characterize the growth, differentiation, dynamics, and ageing of the bone vasculature. In addition to established angiogenic pathways, the role of highly promising novel candidate regulators will be investigated in endothelial cells and perivascular osteoprogenitors with sophisticated inducible and cell type-specific genetic methods in the mouse. Complementing these powerful in vivo approaches, 3D co-cultures generated by cell printing technologies will provide insight into the communication between different cell types. The dynamics of sinusoidal vessel growth and regeneration will be monitored by two-photon imaging in the skull. Finally, I will explore the architectural, cellular and molecular changes and the role of capillary endothelial subpopulations in the sinusoidal vasculature of ageing and osteoporotic mice.
Technological advancements, such as new transgenic strains, mutant models or cell printing approaches, are important aspects of this proposal. AngioBone will provide a first conceptual framework for normal and deregulated function of the bone sinusoidal vasculature. It will also break new ground by analyzing the role of blood vessels in ageing and identifying novel strategies for tissue engineering and, potentially, the prevention/treatment of osteoporosis.
Summary
The skeleton and the sinusoidal vasculature form a functional unit with great relevance in health, regeneration, and disease. Currently, fundamental aspects of sinusoidal vessel growth, specialization, arteriovenous organization and the consequences for tissue perfusion, or the changes occurring during ageing remain unknown. Our preliminary data indicate that key principles of bone vascularization and the role of molecular regulators are highly distinct from other organs. I therefore propose to use powerful combination of mouse genetics, fate mapping, transcriptional profiling, computational biology, confocal and two-photon microscopy, micro-CT and PET imaging, biochemistry and cell biology to characterize the growth, differentiation, dynamics, and ageing of the bone vasculature. In addition to established angiogenic pathways, the role of highly promising novel candidate regulators will be investigated in endothelial cells and perivascular osteoprogenitors with sophisticated inducible and cell type-specific genetic methods in the mouse. Complementing these powerful in vivo approaches, 3D co-cultures generated by cell printing technologies will provide insight into the communication between different cell types. The dynamics of sinusoidal vessel growth and regeneration will be monitored by two-photon imaging in the skull. Finally, I will explore the architectural, cellular and molecular changes and the role of capillary endothelial subpopulations in the sinusoidal vasculature of ageing and osteoporotic mice.
Technological advancements, such as new transgenic strains, mutant models or cell printing approaches, are important aspects of this proposal. AngioBone will provide a first conceptual framework for normal and deregulated function of the bone sinusoidal vasculature. It will also break new ground by analyzing the role of blood vessels in ageing and identifying novel strategies for tissue engineering and, potentially, the prevention/treatment of osteoporosis.
Max ERC Funding
2 478 750 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym Antivessel-T-Cells
Project Development of Vascular-Disrupting Lymphocyte Therapy for Tumours
Researcher (PI) Georgios Coukos
Host Institution (HI) CENTRE HOSPITALIER UNIVERSITAIRE VAUDOIS
Call Details Advanced Grant (AdG), LS7, ERC-2012-ADG_20120314
Summary T cell engineering with chimeric antigen receptors has opened the door to effective immunotherapy. CARs are fusion genes encoding receptors whose extracellular domain comprises a single chain variable fragment (scFv) antibody that binds to a tumour surface epitope, while the intracellular domain comprises the signalling module of CD3ζ along with powerful costimulatory domains (e.g. CD28 and/or 4-1BB). CARs are a major breakthrough, since they allow bypassing HLA restrictions or loss, and they can incorporate potent costimulatory signals tailored to optimize T cell function. However, solid tumours present challenges, since they are often genetically unstable, and the tumour microenvironment impedes T cell function. The tumour vasculature is a much more stable and accessible target, and its disruption has catastrophic consequences for tumours. Nevertheless, the lack of affinity reagents has impeded progress in this area. The objectives of this proposal are to develop the first potent and safe tumour vascular-disrupting tumour immunotherapy using scFv’s and CARs uniquely available in my laboratory.
I propose to use these innovative CARs to understand for the first time the molecular mechanisms underlying the interactions between anti-vascular CAR-T cells and tumour endothelium, and exploit them to maximize tumour vascular destruction. I also intend to employ innovative engineering approaches to minimize the chance of reactivity against normal vasculature. Lastly, I propose to manipulate the tumour damage mechanisms ensuing anti-vascular therapy, to maximize tumour rejection through immunomodulation. We are poised to elucidate critical interactions between tumour endothelium and anti-vascular T cells, and bring to bear cancer therapy of unparalleled power. The impact of this work could be transforming, given the applicability of tumour-vascular disruption across most common tumour types.
Summary
T cell engineering with chimeric antigen receptors has opened the door to effective immunotherapy. CARs are fusion genes encoding receptors whose extracellular domain comprises a single chain variable fragment (scFv) antibody that binds to a tumour surface epitope, while the intracellular domain comprises the signalling module of CD3ζ along with powerful costimulatory domains (e.g. CD28 and/or 4-1BB). CARs are a major breakthrough, since they allow bypassing HLA restrictions or loss, and they can incorporate potent costimulatory signals tailored to optimize T cell function. However, solid tumours present challenges, since they are often genetically unstable, and the tumour microenvironment impedes T cell function. The tumour vasculature is a much more stable and accessible target, and its disruption has catastrophic consequences for tumours. Nevertheless, the lack of affinity reagents has impeded progress in this area. The objectives of this proposal are to develop the first potent and safe tumour vascular-disrupting tumour immunotherapy using scFv’s and CARs uniquely available in my laboratory.
I propose to use these innovative CARs to understand for the first time the molecular mechanisms underlying the interactions between anti-vascular CAR-T cells and tumour endothelium, and exploit them to maximize tumour vascular destruction. I also intend to employ innovative engineering approaches to minimize the chance of reactivity against normal vasculature. Lastly, I propose to manipulate the tumour damage mechanisms ensuing anti-vascular therapy, to maximize tumour rejection through immunomodulation. We are poised to elucidate critical interactions between tumour endothelium and anti-vascular T cells, and bring to bear cancer therapy of unparalleled power. The impact of this work could be transforming, given the applicability of tumour-vascular disruption across most common tumour types.
Max ERC Funding
2 500 000 €
Duration
Start date: 2013-08-01, End date: 2018-07-31
Project acronym ARFMEMBRANESENSORS
Project Membrane sensors in the Arf orbit
Researcher (PI) Bruno Antonny
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS3, ERC-2010-AdG_20100317
Summary Cellular organelles are continuously remodelled by numerous cytosolic proteins that associate transiently with their lipid membrane. Some distort the bilayer, others change its composition, extract lipids or bridge membranes at distance. Previous works from my laboratory have underlined the importance of membrane sensors, i.e. elements within proteins that help to organize membrane-remodelling events by sensing the physical and chemical state of the underlying membrane. A membrane sensor is not necessarily of well-folded domain that interacts with a specific lipid polar head: some intrinsically unfolded motifs harboring deceptively simple sequences can display remarkable membrane adhesive properties. Among these are some amphipathic helices: the ALPS motif with a polar face made mostly by small uncharged polar residues, the Spo20 helix with several histidines in its polar face and, like a mirror image of the ALPS motif, the alpha-synuclein helix with very small hydrophobic residues. Using biochemistry and molecular dynamics, we will compare the membrane binding properties of these sequences (effect of curvature, charge, lipid unsaturation); using bioinformatics we will look for new motifs, using cell biology we will assess the adaptation of these motifs to the physical and chemical features of organelle membranes. Concurrently, we will use reconstitution approaches on artificial membranes to dissect how membrane sensors contribute to the organization of vesicle tethering by golgins and sterol transport by ORP proteins. We surmise that the combination of a molecular ¿switch¿, a small G protein of the Arf family, and of membrane sensors permit to organize these complex reactions in time and in space.
Summary
Cellular organelles are continuously remodelled by numerous cytosolic proteins that associate transiently with their lipid membrane. Some distort the bilayer, others change its composition, extract lipids or bridge membranes at distance. Previous works from my laboratory have underlined the importance of membrane sensors, i.e. elements within proteins that help to organize membrane-remodelling events by sensing the physical and chemical state of the underlying membrane. A membrane sensor is not necessarily of well-folded domain that interacts with a specific lipid polar head: some intrinsically unfolded motifs harboring deceptively simple sequences can display remarkable membrane adhesive properties. Among these are some amphipathic helices: the ALPS motif with a polar face made mostly by small uncharged polar residues, the Spo20 helix with several histidines in its polar face and, like a mirror image of the ALPS motif, the alpha-synuclein helix with very small hydrophobic residues. Using biochemistry and molecular dynamics, we will compare the membrane binding properties of these sequences (effect of curvature, charge, lipid unsaturation); using bioinformatics we will look for new motifs, using cell biology we will assess the adaptation of these motifs to the physical and chemical features of organelle membranes. Concurrently, we will use reconstitution approaches on artificial membranes to dissect how membrane sensors contribute to the organization of vesicle tethering by golgins and sterol transport by ORP proteins. We surmise that the combination of a molecular ¿switch¿, a small G protein of the Arf family, and of membrane sensors permit to organize these complex reactions in time and in space.
Max ERC Funding
1 997 321 €
Duration
Start date: 2011-05-01, End date: 2015-04-30
Project acronym ArtifiCell
Project Synthetic Cell Biology: Designing organelle transport mechanisms
Researcher (PI) James Edward Rothman
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Advanced Grant (AdG), LS3, ERC-2014-ADG
Summary Imagine being able to design into living cells and organisms de novo vesicle transport mechanisms that do not naturally exist? At one level this is a wild-eyed notion of synthetic biology.
But we contend that this vision can be approached even today, focusing first on the process of exocytosis, a fundamental process that impacts almost every area of physiology. Enough has now been learned about the natural core machinery (as recognized by the award of the 2013 Nobel Prize in Physiology or Medicine to the PI and others) to take highly innovative physics/engineering- and DNA-based approaches to design synthetic versions of the secretory apparatus that could someday open new avenues in genetic medicine.
The central idea is to introduce DNA-based functional equivalents of the core protein machinery that naturally form (coats), target (tethers), and fuse (SNAREs) vesicles. We have already taken first steps by using DNA origami-based templates to produce synthetic phospholipid vesicles and complementary DNA-based tethers to specifically capture these DNA-templated vesicles on targeted bilayers. Others have linked DNA oligonucleotides to trigger vesicle fusion.
The next and much more challenging step is to introduce such processes into living cells. We hope to break this barrier, and in the process start a new field of research into “synthetic exocytosis”, by introducing Peptide-Nucleic Acids (PNAs) of tethers and SNAREs to re-direct naturally-produced secretory vesicles to artificially-programmed targets and provide artificially-programmed regulation. PNAs are chosen mainly because they lack the negatively charged phosphate backbones of DNA, and therefore are more readily delivered into the cell across the plasma membrane. Future steps, would include producing the transport vesicles synthetically within the cell by externally supplied origami-based PNA or similar cages, and - much more speculatively - ultimately using encoded DNA and RNAs to provide these functions.
Summary
Imagine being able to design into living cells and organisms de novo vesicle transport mechanisms that do not naturally exist? At one level this is a wild-eyed notion of synthetic biology.
But we contend that this vision can be approached even today, focusing first on the process of exocytosis, a fundamental process that impacts almost every area of physiology. Enough has now been learned about the natural core machinery (as recognized by the award of the 2013 Nobel Prize in Physiology or Medicine to the PI and others) to take highly innovative physics/engineering- and DNA-based approaches to design synthetic versions of the secretory apparatus that could someday open new avenues in genetic medicine.
The central idea is to introduce DNA-based functional equivalents of the core protein machinery that naturally form (coats), target (tethers), and fuse (SNAREs) vesicles. We have already taken first steps by using DNA origami-based templates to produce synthetic phospholipid vesicles and complementary DNA-based tethers to specifically capture these DNA-templated vesicles on targeted bilayers. Others have linked DNA oligonucleotides to trigger vesicle fusion.
The next and much more challenging step is to introduce such processes into living cells. We hope to break this barrier, and in the process start a new field of research into “synthetic exocytosis”, by introducing Peptide-Nucleic Acids (PNAs) of tethers and SNAREs to re-direct naturally-produced secretory vesicles to artificially-programmed targets and provide artificially-programmed regulation. PNAs are chosen mainly because they lack the negatively charged phosphate backbones of DNA, and therefore are more readily delivered into the cell across the plasma membrane. Future steps, would include producing the transport vesicles synthetically within the cell by externally supplied origami-based PNA or similar cages, and - much more speculatively - ultimately using encoded DNA and RNAs to provide these functions.
Max ERC Funding
3 000 000 €
Duration
Start date: 2015-09-01, End date: 2021-08-31
Project acronym ARTimmune
Project Programmable ARTificial immune systems to fight cancer
Researcher (PI) Carl FIGDOR
Host Institution (HI) STICHTING KATHOLIEKE UNIVERSITEIT
Call Details Advanced Grant (AdG), LS7, ERC-2018-ADG
Summary Immunotherapy has entered centre stage as a novel treatment modality for cancer. Notwithstanding this major step forward, toxicity and immunosuppression remain major obstacles, and illustrate the pressing need for more powerful and specific immunotherapies against cancer. To overcome these roadblocks, in ARTimmune, I propose to follow a radically different approach by developing local rather than systemic immunotherapies. Taking advantage of the architecture of a lymph node (LN), I aim to design fully synthetic immune niches to locally instruct immune cell function. I hypothesize that programmable synthetic immune niches, when injected next to a tumour, will act as local powerhouses to generate bursts of cytotoxic T cells for tumour destruction, without toxic side effects. Single cell transcriptomics on LN, obtained from patients that are vaccinated against cancer, will provide unique insight in communication within immune cell clusters and provide a blueprint for the intelligent design of synthetic immune niches. Chemical tools will be used to build branched polymeric structures decorated with immunomodulating molecules to mimic LN architecture. These will be injected, mixed with sponge-like scaffolds to provide porosity needed for immune cell infiltration. Programming of immune cell function will be accomplished by in vivo targeting- and proteolytic activation- of immunomodulators for fine-tuning, and to extend the life span of these local powerhouses. The innovative character of ARTimmune comes from: 1) novel fundamental immunological insight in complex communication within LN cell clusters, 2) a revolutionary new approach in immunotherapy, by the development of 3) injectable- and 4) programmable- synthetic immune niches by state-of-the-art chemical technology. When successful, it will revolutionize cancer immunotherapy, moving from maximal tolerable dose systemic treatment with significant toxicity to local low dose treatment in the direct vicinity of a tumour
Summary
Immunotherapy has entered centre stage as a novel treatment modality for cancer. Notwithstanding this major step forward, toxicity and immunosuppression remain major obstacles, and illustrate the pressing need for more powerful and specific immunotherapies against cancer. To overcome these roadblocks, in ARTimmune, I propose to follow a radically different approach by developing local rather than systemic immunotherapies. Taking advantage of the architecture of a lymph node (LN), I aim to design fully synthetic immune niches to locally instruct immune cell function. I hypothesize that programmable synthetic immune niches, when injected next to a tumour, will act as local powerhouses to generate bursts of cytotoxic T cells for tumour destruction, without toxic side effects. Single cell transcriptomics on LN, obtained from patients that are vaccinated against cancer, will provide unique insight in communication within immune cell clusters and provide a blueprint for the intelligent design of synthetic immune niches. Chemical tools will be used to build branched polymeric structures decorated with immunomodulating molecules to mimic LN architecture. These will be injected, mixed with sponge-like scaffolds to provide porosity needed for immune cell infiltration. Programming of immune cell function will be accomplished by in vivo targeting- and proteolytic activation- of immunomodulators for fine-tuning, and to extend the life span of these local powerhouses. The innovative character of ARTimmune comes from: 1) novel fundamental immunological insight in complex communication within LN cell clusters, 2) a revolutionary new approach in immunotherapy, by the development of 3) injectable- and 4) programmable- synthetic immune niches by state-of-the-art chemical technology. When successful, it will revolutionize cancer immunotherapy, moving from maximal tolerable dose systemic treatment with significant toxicity to local low dose treatment in the direct vicinity of a tumour
Max ERC Funding
2 500 000 €
Duration
Start date: 2019-11-01, End date: 2024-10-31
Project acronym AsthmaPhenotypes
Project Understanding asthma phenotypes: going beyond the atopic/non-atopic paradigm
Researcher (PI) Neil Pearce
Host Institution (HI) LONDON SCHOOL OF HYGIENE AND TROPICAL MEDICINE ROYAL CHARTER
Call Details Advanced Grant (AdG), LS7, ERC-2014-ADG
Summary Fifteen years ago it was widely believed that asthma was an allergic/atopic disease caused by allergen exposure in infancy; this produced atopic sensitization and continued exposure resulted in eosinophilic airways inflammation, bronchial hyper-responsiveness and reversible airflow obstruction. It is now clear that this model is at best incomplete. Less than one-half of asthma cases involve allergic (atopic) mechanisms, and most asthma in low-and-middle income countries is non-atopic. Westernization may be contributing to the global increases in asthma prevalence, but this process appears to involve changes in asthma susceptibility rather than increased exposure to “established” asthma risk factors. Understanding why these changes are occurring is essential in order to halt the growing global asthma epidemic.This will require a combination of epidemiological, clinical and basic science studies in a variety of environments.
A key task is to reclassify asthma phenotypes. These are important to: (i) better understand the aetiological mechanisms of asthma; (ii) identify new causes; and (iii) identify new therapeutic measures. There are major opportunities to address these issues using new techniques for sample collection from the airways (sputum induction, nasal lavage), new methods of analysis (microbiome, epigenetics), and new bioinformatics methods for integrating data from multiple sources and levels. There is an unprecedented potential to go beyond the old atopic/non-atopic categorization of phenotypes.
I will therefore conduct analyses to re-examine and reclassify asthma phenotypes. The key features are the inclusion of: (i) both high and low prevalence centres from both high income countries and low-and-middle income countries; (ii) much more detailed biomarker information than has been used for previous studies of asthma phenotypes; and (iii) new bioinformatics methods for integrating data from multiple sources and levels.
Summary
Fifteen years ago it was widely believed that asthma was an allergic/atopic disease caused by allergen exposure in infancy; this produced atopic sensitization and continued exposure resulted in eosinophilic airways inflammation, bronchial hyper-responsiveness and reversible airflow obstruction. It is now clear that this model is at best incomplete. Less than one-half of asthma cases involve allergic (atopic) mechanisms, and most asthma in low-and-middle income countries is non-atopic. Westernization may be contributing to the global increases in asthma prevalence, but this process appears to involve changes in asthma susceptibility rather than increased exposure to “established” asthma risk factors. Understanding why these changes are occurring is essential in order to halt the growing global asthma epidemic.This will require a combination of epidemiological, clinical and basic science studies in a variety of environments.
A key task is to reclassify asthma phenotypes. These are important to: (i) better understand the aetiological mechanisms of asthma; (ii) identify new causes; and (iii) identify new therapeutic measures. There are major opportunities to address these issues using new techniques for sample collection from the airways (sputum induction, nasal lavage), new methods of analysis (microbiome, epigenetics), and new bioinformatics methods for integrating data from multiple sources and levels. There is an unprecedented potential to go beyond the old atopic/non-atopic categorization of phenotypes.
I will therefore conduct analyses to re-examine and reclassify asthma phenotypes. The key features are the inclusion of: (i) both high and low prevalence centres from both high income countries and low-and-middle income countries; (ii) much more detailed biomarker information than has been used for previous studies of asthma phenotypes; and (iii) new bioinformatics methods for integrating data from multiple sources and levels.
Max ERC Funding
2 348 803 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym AVATAR
Project Integrating Genomics and Avatar Mouse Models to Personalize Pancreatic Cancer Treatment
Researcher (PI) Manuel HIDALGO MEDINA
Host Institution (HI) HOSPITAL UNIVERSITARIO DE FUENLABRADA
Call Details Advanced Grant (AdG), LS7, ERC-2014-ADG
Summary The prognosis of patients with metastatic pancreatic cancer (PDAC) is very poor. Recent studies have started to elucidate the genetic landscape of this disease to show that PDAC is a genetically complex, unstable, and heterogeneous cancer. However, in-depth analysis of individual patient genomes couple with personalize Avatar mouse models is providing highly effective therapeutic opportunities for the individual patient. Thus, metastatic PDAC appears a candidate disease to implement a genomics-base, personalized treatment approach. In this project, we will conduct an open label, multicenter, randomized phase III study in patients with standard of care resistant metastatic pancreatic cancer aiming to test the hypothesis that an integrated personalized treatment approach improves survival compare to a conventional treatment. Patients randomized to the personalize treatment arm will undergo a biopsy of a metastatic lesion to perform a targeted genome analysis using next generation sequencing. In addition, we will generate a personalize Avatar mouse model from the same patient. We will employ sophisticated bioinformatic analysis as well as mining of drug response-genetic databases to select, for each individual patient, candidate therapeutic targets that will be experimentally tested in the patient´s Avatar model to select the most effective regimen that will ultimately applied to the patient. In addition, based on the genomic data, we will design an individualized monitoring plan for each patient using BEAMing technology to monitor circulating levels of mutated genes. Furthermore, with a discovery goal, we will perform in depth genomic analysis of metastatic PDAC lesions in this cohort of clinically well-annotated patients with Avatar mouse models for therapeutic validation. Overall we expect this work will contribute to our understanding of PDAC and will favourably impact the treatment of this dismal cancer.
Summary
The prognosis of patients with metastatic pancreatic cancer (PDAC) is very poor. Recent studies have started to elucidate the genetic landscape of this disease to show that PDAC is a genetically complex, unstable, and heterogeneous cancer. However, in-depth analysis of individual patient genomes couple with personalize Avatar mouse models is providing highly effective therapeutic opportunities for the individual patient. Thus, metastatic PDAC appears a candidate disease to implement a genomics-base, personalized treatment approach. In this project, we will conduct an open label, multicenter, randomized phase III study in patients with standard of care resistant metastatic pancreatic cancer aiming to test the hypothesis that an integrated personalized treatment approach improves survival compare to a conventional treatment. Patients randomized to the personalize treatment arm will undergo a biopsy of a metastatic lesion to perform a targeted genome analysis using next generation sequencing. In addition, we will generate a personalize Avatar mouse model from the same patient. We will employ sophisticated bioinformatic analysis as well as mining of drug response-genetic databases to select, for each individual patient, candidate therapeutic targets that will be experimentally tested in the patient´s Avatar model to select the most effective regimen that will ultimately applied to the patient. In addition, based on the genomic data, we will design an individualized monitoring plan for each patient using BEAMing technology to monitor circulating levels of mutated genes. Furthermore, with a discovery goal, we will perform in depth genomic analysis of metastatic PDAC lesions in this cohort of clinically well-annotated patients with Avatar mouse models for therapeutic validation. Overall we expect this work will contribute to our understanding of PDAC and will favourably impact the treatment of this dismal cancer.
Max ERC Funding
2 498 688 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym BARCODE
Project The use of genetic profiling to guide prostate cancer targeted screening and cancer care
Researcher (PI) Rosalind Anne Eeles
Host Institution (HI) THE INSTITUTE OF CANCER RESEARCH: ROYAL CANCER HOSPITAL
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary "Prostate cancer is the commonest solid cancer in men in the European Community. There is evidence for genetic predisposition to the development of prostate cancer and our group has found the largest number of such genetic variants described to date worldwide. The next challenge is to harness these discoveries to advance the clinical care of populations and prostate cancer patients to improve screening and target treatments. This proposal, BARCODE, aims to be ground-breaking in this area. BARCODE has two components (1) to profile a population in England using the current 77 genetic variant profile and compare screening outcomes with those from population based screening studies to determine if genetics can target screening more effectively in this disease by identifying prostate cancer that more often needs treatment and (2) genetically profiling men with prostate cancer in the uro-oncology clinic for a panel of genes which predict for worse outcome so that these men can be offered more intensive staging and treatment within clinical trials. This will use next generation sequencing technology using a barcoding system which we have developed to speed up throughput and reduce costs. The PI will spend 35% of her time on this project and she will not charge for her time spent on this grant as she is funded by The University of London UK. The research team at The Institute Of Cancer Research, London, UK is a multidisciplinary team which leads the field of genetic predisposition to prostate cancer and its clinical application and so is well placed to deliver on this research. This application will have a dramatic impact on other researchers as it is ground –breaking and state of the art in its application of genetic findings to public health and cancer care. It will therefore influence the work being undertaken in both these areas to integrate genetic profiling and gene panel analysis into population screening and cancer care respectively."
Summary
"Prostate cancer is the commonest solid cancer in men in the European Community. There is evidence for genetic predisposition to the development of prostate cancer and our group has found the largest number of such genetic variants described to date worldwide. The next challenge is to harness these discoveries to advance the clinical care of populations and prostate cancer patients to improve screening and target treatments. This proposal, BARCODE, aims to be ground-breaking in this area. BARCODE has two components (1) to profile a population in England using the current 77 genetic variant profile and compare screening outcomes with those from population based screening studies to determine if genetics can target screening more effectively in this disease by identifying prostate cancer that more often needs treatment and (2) genetically profiling men with prostate cancer in the uro-oncology clinic for a panel of genes which predict for worse outcome so that these men can be offered more intensive staging and treatment within clinical trials. This will use next generation sequencing technology using a barcoding system which we have developed to speed up throughput and reduce costs. The PI will spend 35% of her time on this project and she will not charge for her time spent on this grant as she is funded by The University of London UK. The research team at The Institute Of Cancer Research, London, UK is a multidisciplinary team which leads the field of genetic predisposition to prostate cancer and its clinical application and so is well placed to deliver on this research. This application will have a dramatic impact on other researchers as it is ground –breaking and state of the art in its application of genetic findings to public health and cancer care. It will therefore influence the work being undertaken in both these areas to integrate genetic profiling and gene panel analysis into population screening and cancer care respectively."
Max ERC Funding
2 499 123 €
Duration
Start date: 2014-10-01, End date: 2019-09-30
Project acronym BARRAGE
Project Cell compartmentalization, individuation and diversity
Researcher (PI) Yves Barral
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Advanced Grant (AdG), LS3, ERC-2009-AdG
Summary Asymmetric cell division is a key mechanism for the generation of cell diversity in eukaryotes. During this process, a polarized mother cell divides into non-equivalent daughters. These may differentially inherit fate determinants, irreparable damages or age determinants. Our aim is to decipher the mechanisms governing the individualization of daughters from each other. In the past ten years, our studies identified several lateral diffusion barriers located in the plasma membrane and the endoplasmic reticulum of budding yeast. These barriers all restrict molecular exchanges between the mother cell and its bud, and thereby compartmentalize the cell already long before its division. They play key roles in the asymmetric segregation of various factors. On one side, they help maintain polarized factors into the bud. Thereby, they reinforce cell polarity and sequester daughter-specific fate determinants into the bud. On the other side they prevent aging factors of the mother from entering the bud. Hence, they play key roles in the rejuvenation of the bud, in the aging of the mother, and in the differentiation of mother and daughter from each other. Recently, we accumulated evidence that some of these barriers are subject to regulation, such as to help modulate the longevity of the mother cell in response to environmental signals. Our data also suggest that barriers help the mother cell keep traces of its life history, thereby contributing to its individuation and adaption to the environment. In this project, we will address the following questions: 1 How are these barriers assembled, functioning, and regulated? 2 What type of differentiation processes are they involved in? 3 Are they conserved in other eukaryotes, and what are their functions outside of budding yeast? These studies will shed light into the principles underlying and linking aging, rejuvenation and differentiation.
Summary
Asymmetric cell division is a key mechanism for the generation of cell diversity in eukaryotes. During this process, a polarized mother cell divides into non-equivalent daughters. These may differentially inherit fate determinants, irreparable damages or age determinants. Our aim is to decipher the mechanisms governing the individualization of daughters from each other. In the past ten years, our studies identified several lateral diffusion barriers located in the plasma membrane and the endoplasmic reticulum of budding yeast. These barriers all restrict molecular exchanges between the mother cell and its bud, and thereby compartmentalize the cell already long before its division. They play key roles in the asymmetric segregation of various factors. On one side, they help maintain polarized factors into the bud. Thereby, they reinforce cell polarity and sequester daughter-specific fate determinants into the bud. On the other side they prevent aging factors of the mother from entering the bud. Hence, they play key roles in the rejuvenation of the bud, in the aging of the mother, and in the differentiation of mother and daughter from each other. Recently, we accumulated evidence that some of these barriers are subject to regulation, such as to help modulate the longevity of the mother cell in response to environmental signals. Our data also suggest that barriers help the mother cell keep traces of its life history, thereby contributing to its individuation and adaption to the environment. In this project, we will address the following questions: 1 How are these barriers assembled, functioning, and regulated? 2 What type of differentiation processes are they involved in? 3 Are they conserved in other eukaryotes, and what are their functions outside of budding yeast? These studies will shed light into the principles underlying and linking aging, rejuvenation and differentiation.
Max ERC Funding
2 200 000 €
Duration
Start date: 2010-05-01, End date: 2015-04-30
Project acronym Bio-Phononics
Project Advanced Microfluidics & Diagnostics using Acoustic Holograms – Bio-Phononics
Researcher (PI) Jonathan Cooper
Host Institution (HI) UNIVERSITY OF GLASGOW
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary This proposal seeks to develop a novel technique for fluid and particle manipulations, based upon exploiting the mechanical interactions between acoustic waves and phononic. The new platform involves generating surface acoustic waves (SAWs) on piezoelectric chips, but, unlike previous work, the ultrasonic waves are first coupled into a phononic lattice, which is placed in the path of the ultrasonic wave. The phononic lattice comprises a miniaturised array of mechanical elements which modulates the sound in a manner analogous to how light is “patterned” using a hologram. However, whilst in an optical hologram, the pattern is created by exploiting the differences in refractive indices of the elements of the structure, here the ultrasonic field is modulated both by the elastic contrast between the elements in the array, as well as by the dimensions of the array and its surrounding matrix (including the size and pitch of the features within the array). The result of passing the acoustic wave through a phononic crystal is the formation of new and complex ultrasonic landscapes.
As part of the proposed work we aim to understand the physics of this technology and to exploit its development in a range of medical devices. We will show that by using phononic crystals it is possible to create highly controllable patterns of acoustic field intensities, which propagate into the fluid, creating pressure differences that result in unique flow patterns to enable a new platform for including biological sample processing, medical diagnostics, drug delivery and blood clotting devices – all on low cost disposable devices. Different frequencies of ultrasound will interact with different phononic structures to give different functions, providing a toolbox of different functions. Just as in electronics, where discrete components are combined to create circuits, so we propose to combine different phononic lattices to create fluidic microcircuits with important new applications.
Summary
This proposal seeks to develop a novel technique for fluid and particle manipulations, based upon exploiting the mechanical interactions between acoustic waves and phononic. The new platform involves generating surface acoustic waves (SAWs) on piezoelectric chips, but, unlike previous work, the ultrasonic waves are first coupled into a phononic lattice, which is placed in the path of the ultrasonic wave. The phononic lattice comprises a miniaturised array of mechanical elements which modulates the sound in a manner analogous to how light is “patterned” using a hologram. However, whilst in an optical hologram, the pattern is created by exploiting the differences in refractive indices of the elements of the structure, here the ultrasonic field is modulated both by the elastic contrast between the elements in the array, as well as by the dimensions of the array and its surrounding matrix (including the size and pitch of the features within the array). The result of passing the acoustic wave through a phononic crystal is the formation of new and complex ultrasonic landscapes.
As part of the proposed work we aim to understand the physics of this technology and to exploit its development in a range of medical devices. We will show that by using phononic crystals it is possible to create highly controllable patterns of acoustic field intensities, which propagate into the fluid, creating pressure differences that result in unique flow patterns to enable a new platform for including biological sample processing, medical diagnostics, drug delivery and blood clotting devices – all on low cost disposable devices. Different frequencies of ultrasound will interact with different phononic structures to give different functions, providing a toolbox of different functions. Just as in electronics, where discrete components are combined to create circuits, so we propose to combine different phononic lattices to create fluidic microcircuits with important new applications.
Max ERC Funding
2 208 594 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym BIOMECAMORPH
Project The Biomechanics of Epithelial Cell and Tissue Morphogenesis
Researcher (PI) Thomas Marie Michel Lecuit
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS3, ERC-2012-ADG_20120314
Summary Tissue morphogenesis is a complex process that emerges from spatially controlled patterns of cell shape changes. Dedicated genetic programmes regulate cell behaviours, exemplified in animals by the specification of apical constriction in invaginating epithelial tissues, or the orientation of cell intercalation during tissue extension. This genetic control is constrained by physical properties of cells that dictate how they can modify their shape. A major challenge is to understand how biochemical pathways control subcellular mechanics in epithelia, such as how forces are produced by interactions between actin filaments and myosin motors, and how these forces are transmitted at cell junctions. The major objective of our project is to investigate the fundamental principles of epithelial mechanics and to understand how intercellular signals and mechanical coupling between cells coordinate individual behaviours at the tissue level.
We will study early Drosophila embryogenesis and combine quantitative cell biological studies of cell dynamics, biophysical characterization of cell mechanics and genetic control of cell signalling to answer the following questions: i) how are forces generated, in particular what underlies deformation and stabilization of cell shape by actomyosin networks, and pulsatile contractility; ii) how are forces transmitted at junctions, what are the feedback interactions between tension generation and transmission; iii) how are individual cell mechanics orchestrated at the tissue level to yield collective tissue morphogenesis?
We expect to encapsulate the information-based, cell biological and physical descriptions of morphogenesis in a single, coherent framework. The project should impact more broadly on morphogenesis in other organisms and shed light on the mechanisms underlying robustness and plasticity in epithelia.
Summary
Tissue morphogenesis is a complex process that emerges from spatially controlled patterns of cell shape changes. Dedicated genetic programmes regulate cell behaviours, exemplified in animals by the specification of apical constriction in invaginating epithelial tissues, or the orientation of cell intercalation during tissue extension. This genetic control is constrained by physical properties of cells that dictate how they can modify their shape. A major challenge is to understand how biochemical pathways control subcellular mechanics in epithelia, such as how forces are produced by interactions between actin filaments and myosin motors, and how these forces are transmitted at cell junctions. The major objective of our project is to investigate the fundamental principles of epithelial mechanics and to understand how intercellular signals and mechanical coupling between cells coordinate individual behaviours at the tissue level.
We will study early Drosophila embryogenesis and combine quantitative cell biological studies of cell dynamics, biophysical characterization of cell mechanics and genetic control of cell signalling to answer the following questions: i) how are forces generated, in particular what underlies deformation and stabilization of cell shape by actomyosin networks, and pulsatile contractility; ii) how are forces transmitted at junctions, what are the feedback interactions between tension generation and transmission; iii) how are individual cell mechanics orchestrated at the tissue level to yield collective tissue morphogenesis?
We expect to encapsulate the information-based, cell biological and physical descriptions of morphogenesis in a single, coherent framework. The project should impact more broadly on morphogenesis in other organisms and shed light on the mechanisms underlying robustness and plasticity in epithelia.
Max ERC Funding
2 473 313 €
Duration
Start date: 2013-05-01, End date: 2018-04-30
Project acronym BIOMECHTOOLS
Project Biomechanical diagnostic, pre-planning and outcome tools to improve musculoskeletal surgery
Researcher (PI) Nicolaas Verdonschot
Host Institution (HI) STICHTING KATHOLIEKE UNIVERSITEIT
Call Details Advanced Grant (AdG), LS7, ERC-2012-ADG_20120314
Summary The aetiology of many musculoskeletal (MS) diseases is related to biomechanical factors. However, the tools to assess the biomechanical condition of patients used by clinicians and researchers are often crude and subjective leading to non-optimal patient analyses and care. In this project innovations related to imaging, sensor technology and biomechanical modelling are utilized to generate versatile, accurate and objective methods to quantify the (pathological) MS condition of the lower extremity of patients in a unique manner. The project will produce advanced diagnostic, pre-planning and outcome tools which allow clinicians and researchers for detailed biomechanical analysis about abnormal tissue deformations, pathological loading of the joints, abnormal stresses in the hard and soft tissues, and aberrant joint kinematics.
The key objectives of this proposal are:
1) Develop and validate image-based 3-D volumetric elastographic diagnostic methods that can quantify normal and pathological conditions under dynamic loading and which can be linked to biomechanical modelling tools.
2) Create an ultrasound (US)-based system to assess internal joint kinematics which can be used as a diagnostic tool for clinicians and researchers and is a validation tool for biomechanical modelling.
3) Generate and validate an ambulant functional (force and kinematic) diagnostic system which is easy to use and which can be used to provide input data for biomechanical models.
4) Create and validate a new modelling approach that integrates muscle-models with finite element models at a highly personalized level.
5) Generate biomechanical models which have personalized mechanical properties of the hard and soft tissues.
6) Demonstrate the applicability of the personalized diagnostic and pre-planning platform by application to healthy individuals and patient subjects.
Support from the ERC will open new research fields related to biomechanical patient assessment and modeling of MS pathologies.
Summary
The aetiology of many musculoskeletal (MS) diseases is related to biomechanical factors. However, the tools to assess the biomechanical condition of patients used by clinicians and researchers are often crude and subjective leading to non-optimal patient analyses and care. In this project innovations related to imaging, sensor technology and biomechanical modelling are utilized to generate versatile, accurate and objective methods to quantify the (pathological) MS condition of the lower extremity of patients in a unique manner. The project will produce advanced diagnostic, pre-planning and outcome tools which allow clinicians and researchers for detailed biomechanical analysis about abnormal tissue deformations, pathological loading of the joints, abnormal stresses in the hard and soft tissues, and aberrant joint kinematics.
The key objectives of this proposal are:
1) Develop and validate image-based 3-D volumetric elastographic diagnostic methods that can quantify normal and pathological conditions under dynamic loading and which can be linked to biomechanical modelling tools.
2) Create an ultrasound (US)-based system to assess internal joint kinematics which can be used as a diagnostic tool for clinicians and researchers and is a validation tool for biomechanical modelling.
3) Generate and validate an ambulant functional (force and kinematic) diagnostic system which is easy to use and which can be used to provide input data for biomechanical models.
4) Create and validate a new modelling approach that integrates muscle-models with finite element models at a highly personalized level.
5) Generate biomechanical models which have personalized mechanical properties of the hard and soft tissues.
6) Demonstrate the applicability of the personalized diagnostic and pre-planning platform by application to healthy individuals and patient subjects.
Support from the ERC will open new research fields related to biomechanical patient assessment and modeling of MS pathologies.
Max ERC Funding
2 456 400 €
Duration
Start date: 2013-05-01, End date: 2018-04-30
Project acronym BODYBUILT
Project Building The Vertebrate Body
Researcher (PI) Olivier Pourquie
Host Institution (HI) CENTRE EUROPEEN DE RECHERCHE EN BIOLOGIE ET MEDECINE
Call Details Advanced Grant (AdG), LS3, ERC-2009-AdG
Summary My lab is interested in the development of the tissue that gives rise to vertebrae and skeletal muscles called the paraxial mesoderm. A striking feature of this tissue is its segmental organization and we have made major contributions to the understanding of the molecular control of the segmentation process. We identified a molecular oscillator associated to the rhythmic production of somites and proposed a model for vertebrate segmentation based on the integration of a rhythmic signaling pulse gated spatially by a system of traveling FGF and Wnt signaling gradients. We are also studying the differentiation of paraxial mesoderm precursors into the muscle, cartilage and dermis lineages. Our work identified the Wnt, FGF and Notch pathways as playing a prominent role in the patterning and differentiation of paraxial mesoderm. In this application, we largely focus on the molecular control of paraxial mesoderm development. Using microarray and high throughput sequencing-based approaches and bioinformatics, we will characterize the transcriptional network acting downstream of Wnt, FGF and Notch in the presomitic mesoderm (PSM). We will also use genetic and pharmacological approaches utilizing real-time imaging reporters to characterize the pacemaker of the segmentation clock in vivo, and also in vitro using differentiated embryonic stem cells. We further propose to characterize in detail a novel RA-dependent pathway that we identified and which controls the somite left-right symmetry. Our work is expected to have a strong impact in the field of congenital spine anomalies, currently an understudied biomedical problem, and will be of utility in elucidating the etiology and eventual prevention of these disorders. This work is also expected to further our understanding of the Notch, Wnt, FGF and RA signalling pathways which are involved in segmentation and in the establishment of the vertebrate body plan, and which play important roles in a wide array of human diseases.
Summary
My lab is interested in the development of the tissue that gives rise to vertebrae and skeletal muscles called the paraxial mesoderm. A striking feature of this tissue is its segmental organization and we have made major contributions to the understanding of the molecular control of the segmentation process. We identified a molecular oscillator associated to the rhythmic production of somites and proposed a model for vertebrate segmentation based on the integration of a rhythmic signaling pulse gated spatially by a system of traveling FGF and Wnt signaling gradients. We are also studying the differentiation of paraxial mesoderm precursors into the muscle, cartilage and dermis lineages. Our work identified the Wnt, FGF and Notch pathways as playing a prominent role in the patterning and differentiation of paraxial mesoderm. In this application, we largely focus on the molecular control of paraxial mesoderm development. Using microarray and high throughput sequencing-based approaches and bioinformatics, we will characterize the transcriptional network acting downstream of Wnt, FGF and Notch in the presomitic mesoderm (PSM). We will also use genetic and pharmacological approaches utilizing real-time imaging reporters to characterize the pacemaker of the segmentation clock in vivo, and also in vitro using differentiated embryonic stem cells. We further propose to characterize in detail a novel RA-dependent pathway that we identified and which controls the somite left-right symmetry. Our work is expected to have a strong impact in the field of congenital spine anomalies, currently an understudied biomedical problem, and will be of utility in elucidating the etiology and eventual prevention of these disorders. This work is also expected to further our understanding of the Notch, Wnt, FGF and RA signalling pathways which are involved in segmentation and in the establishment of the vertebrate body plan, and which play important roles in a wide array of human diseases.
Max ERC Funding
2 500 000 €
Duration
Start date: 2010-04-01, End date: 2015-03-31
Project acronym BrainDrain
Project Translational implications of the discovery of brain-draining lymphatics
Researcher (PI) Kari ALITALO
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary In 2010, 800 billion Euros was spent on brain diseases in Europe and the cost is expected to increase due to the aging population. – Here I propose to exploit our new discovery for research to alleviate this disease burden. In work selected by Nature Medicine among the top 10 ”Notable Advances” and by Science as one of the 10 ”Breakthroughs of the year” 2015, we discovered a meningeal lymphatic vascular system that serves brain homeostasis. We want to reassess current concepts about cerebrovascular dynamics, fluid drainage and cellular trafficking in physiological conditions, in Alzheimer’s disease mouse models and in human postmortem tissues. First, we will study the development and properties of meningeal lymphatics and how they are sustained during aging. We then want to analyse the clearance of macromolecules and protein aggregates in Alzheimer’s disease in mice that lack the newly discovered meningeal lymphatic drainage system. We will study if growth factor-mediated expansion of lymphatic vessels alleviates the parenchymal accumulation of neurotoxic amyloid beta and pathogenesis of Alzheimer’s disease and brain damage after traumatic brain injury. We will further analyse the role of lymphangiogenic growth factors and lymphatic vessels in brain solute clearance, immune cell trafficking and in a mouse model of multiple sclerosis. The meningeal lymphatics could be involved in a number of neurodegenerative and neuroinflammatory diseases of considerable human and socioeconomic burden. Several of our previous concepts have already been translated to clinical development and we aim to develop proof-of-principle therapeutic concepts in this project. I feel that we are just now in a unique position to advance frontline European translational biomedical research in this suddenly emerging field, which has received great attention worldwide.
Summary
In 2010, 800 billion Euros was spent on brain diseases in Europe and the cost is expected to increase due to the aging population. – Here I propose to exploit our new discovery for research to alleviate this disease burden. In work selected by Nature Medicine among the top 10 ”Notable Advances” and by Science as one of the 10 ”Breakthroughs of the year” 2015, we discovered a meningeal lymphatic vascular system that serves brain homeostasis. We want to reassess current concepts about cerebrovascular dynamics, fluid drainage and cellular trafficking in physiological conditions, in Alzheimer’s disease mouse models and in human postmortem tissues. First, we will study the development and properties of meningeal lymphatics and how they are sustained during aging. We then want to analyse the clearance of macromolecules and protein aggregates in Alzheimer’s disease in mice that lack the newly discovered meningeal lymphatic drainage system. We will study if growth factor-mediated expansion of lymphatic vessels alleviates the parenchymal accumulation of neurotoxic amyloid beta and pathogenesis of Alzheimer’s disease and brain damage after traumatic brain injury. We will further analyse the role of lymphangiogenic growth factors and lymphatic vessels in brain solute clearance, immune cell trafficking and in a mouse model of multiple sclerosis. The meningeal lymphatics could be involved in a number of neurodegenerative and neuroinflammatory diseases of considerable human and socioeconomic burden. Several of our previous concepts have already been translated to clinical development and we aim to develop proof-of-principle therapeutic concepts in this project. I feel that we are just now in a unique position to advance frontline European translational biomedical research in this suddenly emerging field, which has received great attention worldwide.
Max ERC Funding
2 420 429 €
Duration
Start date: 2017-08-01, End date: 2022-07-31
Project acronym BRAINEVODEVO
Project A Neuron Type Atlas of the Annelid Brain: Development and Evolution of Chemosensory-Motor Circuits
Researcher (PI) Detlev Arendt
Host Institution (HI) EUROPEAN MOLECULAR BIOLOGY LABORATORY
Call Details Advanced Grant (AdG), LS3, ERC-2011-ADG_20110310
Summary Neural circuits, composed of interconnected neurons, represent the basic unit of the nervous system. One way to understand the highly complex arrangement of cross-talking, serial and parallel circuits is to resolve its developmental and evolutionary emergence. The rationale of the research proposal presented here is to elucidate the complex circuitry of the vertebrate and insect forebrain by comparison to the much simpler and evolutionary ancient “connectome” of the marine annelid Platynereis dumerilii. We will build a unique resource, the Platynereis Neuron Type Atlas, combining, for the first time, neuronal morphologies, axonal projections, cellular expression profiling and developmental lineage for an entire bilaterian brain. We will focus on five days old larvae when most adult neuron types are already present in small number and large part of the axonal scaffold in place.
Building on the Neuron Type Atlas, the second part of the proposal envisages the functional dissection of the Platynereis chemosensory-motor forebrain circuits. A newly developed microfluidics behavioural assay system, together with a cell-based GPCR screening will identify partaking neurons. Zinc finger nuclease-mediated knockout of circuit-specific transcription factors as identified from the Atlas will reveal circuit-specific gene regulatory networks, downstream effector genes and functional characteristics. Laser ablation of GFP-labeled single neurons and axonal connections will yield further insight into the function of circuit components and subcircuits. Given the ancient nature of the Platynereis brain, this research is expected to reveal a simple, developmental and evolutionary “blueprint” for the olfactory circuits in mice and flies and to shed new light on the evolution of information processing in glomeruli and higher-level integration in sensory-associative brain centres.
Summary
Neural circuits, composed of interconnected neurons, represent the basic unit of the nervous system. One way to understand the highly complex arrangement of cross-talking, serial and parallel circuits is to resolve its developmental and evolutionary emergence. The rationale of the research proposal presented here is to elucidate the complex circuitry of the vertebrate and insect forebrain by comparison to the much simpler and evolutionary ancient “connectome” of the marine annelid Platynereis dumerilii. We will build a unique resource, the Platynereis Neuron Type Atlas, combining, for the first time, neuronal morphologies, axonal projections, cellular expression profiling and developmental lineage for an entire bilaterian brain. We will focus on five days old larvae when most adult neuron types are already present in small number and large part of the axonal scaffold in place.
Building on the Neuron Type Atlas, the second part of the proposal envisages the functional dissection of the Platynereis chemosensory-motor forebrain circuits. A newly developed microfluidics behavioural assay system, together with a cell-based GPCR screening will identify partaking neurons. Zinc finger nuclease-mediated knockout of circuit-specific transcription factors as identified from the Atlas will reveal circuit-specific gene regulatory networks, downstream effector genes and functional characteristics. Laser ablation of GFP-labeled single neurons and axonal connections will yield further insight into the function of circuit components and subcircuits. Given the ancient nature of the Platynereis brain, this research is expected to reveal a simple, developmental and evolutionary “blueprint” for the olfactory circuits in mice and flies and to shed new light on the evolution of information processing in glomeruli and higher-level integration in sensory-associative brain centres.
Max ERC Funding
2 489 048 €
Duration
Start date: 2012-03-01, End date: 2017-02-28
Project acronym BRCA-ERC
Project Understanding cancer development in BRCA 1/2 mutation carriers for improved Early detection and Risk Control
Researcher (PI) Martin WIDSCHWENDTER
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Recent evidence demonstrates that cancer is overtaking cardiovascular disease as the number one cause of mortality in Europe. This is largely due to the lack of preventative measures for common (e.g. breast) or highly fatal (e.g. ovarian) human cancers. Most cancers are multifactorial in origin. The core hypothesis of this research programme is that the extremely high risk of BRCA1/2 germline mutation carriers to develop breast and ovarian cancer is a net consequence of cell-autonomous (direct effect of BRCA mutation in cells at risk) and cell non-autonomous (produced in distant organs and affecting organs at risk) factors which both trigger epigenetic, cancer-initiating effects.
The project’s aims are centered around the principles of systems medicine and built on a large cohort of BRCA mutation carriers and controls who will be offered newly established cancer screening programmes. We will uncover how ‘cell non-autonomous’ factors work, provide detail on the epigenetic changes in at-risk tissues and investigate whether these changes are mechanistically linked to cancer, study whether we can neutralise this process and measure success in the organs at risk, and ideally in easy to access samples such as blood, buccal and cervical cells.
In my Department for Women’s Cancer we have assembled a powerful interdisciplinary team including computational biologists, functionalists, immunologists and clinician scientists linked to leading patient advocacy groups which is extremely well placed to lead this pioneering project to develop the fundamental understanding of cancer development in women with BRCA mutations. To reset the epigenome, re-establishing normal cell identity and consequently reducing cancer risk without the need for surgery and being able to monitor the efficacy using multicellular epigenetic outcome predictors will be a major scientific and medical breakthrough and possibly applicable to other chronic diseases.
Summary
Recent evidence demonstrates that cancer is overtaking cardiovascular disease as the number one cause of mortality in Europe. This is largely due to the lack of preventative measures for common (e.g. breast) or highly fatal (e.g. ovarian) human cancers. Most cancers are multifactorial in origin. The core hypothesis of this research programme is that the extremely high risk of BRCA1/2 germline mutation carriers to develop breast and ovarian cancer is a net consequence of cell-autonomous (direct effect of BRCA mutation in cells at risk) and cell non-autonomous (produced in distant organs and affecting organs at risk) factors which both trigger epigenetic, cancer-initiating effects.
The project’s aims are centered around the principles of systems medicine and built on a large cohort of BRCA mutation carriers and controls who will be offered newly established cancer screening programmes. We will uncover how ‘cell non-autonomous’ factors work, provide detail on the epigenetic changes in at-risk tissues and investigate whether these changes are mechanistically linked to cancer, study whether we can neutralise this process and measure success in the organs at risk, and ideally in easy to access samples such as blood, buccal and cervical cells.
In my Department for Women’s Cancer we have assembled a powerful interdisciplinary team including computational biologists, functionalists, immunologists and clinician scientists linked to leading patient advocacy groups which is extremely well placed to lead this pioneering project to develop the fundamental understanding of cancer development in women with BRCA mutations. To reset the epigenome, re-establishing normal cell identity and consequently reducing cancer risk without the need for surgery and being able to monitor the efficacy using multicellular epigenetic outcome predictors will be a major scientific and medical breakthrough and possibly applicable to other chronic diseases.
Max ERC Funding
2 497 841 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym BREATHE
Project BRain dEvelopment and Air polluTion ultrafine particles in scHool childrEn
Researcher (PI) Jordi Sunyer Deu
Host Institution (HI) FUNDACION PRIVADA INSTITUTO DE SALUD GLOBAL BARCELONA
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary Traffic-related air pollution is an important environmental problem that may affect neurodevelopment. Ultrafine particles (UFP) translocate to the brains of experimental animals resulting in local proinflammatory overexpression. As the basic elements for thinking are acquired by developing brains during infancy and childhood, susceptibility may be elevated in early life.
We postulate that traffic-related air pollution (particularly UFPs and metals/hydrocarbons content) impairs neurodevelopment in part via effects on frontal lobe maturation, likely increasing attention-deficit/hyperactivity disorder (ADHD). BREATHE objectives are to develop valid methods to measure children's personal UFP exposure and to develop valid neuroimaging methods to assess correlations between neurobehavior, neurostructural alterations and particle deposition in order to reveal how traffic pollution affects children¿s exposure to key contaminants and brain development, and identify susceptible subgroups.
We have conducted general population birth cohort studies providing preliminary evidence of residential air pollution effects on prenatal growth and mental development.
We aim to demonstrate short and long-term effects on neurodevelopment using innovative epidemiological methods interfaced with environmental chemistry and neuroimaging following 4000 children from 40 schools with contrasting high/low traffic exposure in six linked components involving: repeated psychometric tests, UFP exposure assessment using personal, school and home measurements, gene-environment interactions on inflammation, detoxification pathways and ADHD genome-wide-associated genes, neuroimaging (magnetic resonance imaging/spectroscopy) in ADHD/non-ADHD children, integrative causal modeling using mathematics, and replication in 2900 children with neurodevelopment followed from pregnancy.
We believe the expected results will have worldwide global planning and policy implications.
Summary
Traffic-related air pollution is an important environmental problem that may affect neurodevelopment. Ultrafine particles (UFP) translocate to the brains of experimental animals resulting in local proinflammatory overexpression. As the basic elements for thinking are acquired by developing brains during infancy and childhood, susceptibility may be elevated in early life.
We postulate that traffic-related air pollution (particularly UFPs and metals/hydrocarbons content) impairs neurodevelopment in part via effects on frontal lobe maturation, likely increasing attention-deficit/hyperactivity disorder (ADHD). BREATHE objectives are to develop valid methods to measure children's personal UFP exposure and to develop valid neuroimaging methods to assess correlations between neurobehavior, neurostructural alterations and particle deposition in order to reveal how traffic pollution affects children¿s exposure to key contaminants and brain development, and identify susceptible subgroups.
We have conducted general population birth cohort studies providing preliminary evidence of residential air pollution effects on prenatal growth and mental development.
We aim to demonstrate short and long-term effects on neurodevelopment using innovative epidemiological methods interfaced with environmental chemistry and neuroimaging following 4000 children from 40 schools with contrasting high/low traffic exposure in six linked components involving: repeated psychometric tests, UFP exposure assessment using personal, school and home measurements, gene-environment interactions on inflammation, detoxification pathways and ADHD genome-wide-associated genes, neuroimaging (magnetic resonance imaging/spectroscopy) in ADHD/non-ADHD children, integrative causal modeling using mathematics, and replication in 2900 children with neurodevelopment followed from pregnancy.
We believe the expected results will have worldwide global planning and policy implications.
Max ERC Funding
2 499 230 €
Duration
Start date: 2011-08-01, End date: 2016-07-31
Project acronym CAN-IT-BARRIERS
Project Disruption of systemic and microenvironmental barriers to immunotherapy of antigenic tumors
Researcher (PI) Douglas HANAHAN
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), LS7, ERC-2018-ADG
Summary The frontier in cancer therapy of orchestrating the immune system to attack tumors is producing unprecedented survival benefit in some patients. The corollary is lack of efficacy both in ostensibly responsive tumor types as well as others that are mostly non-responsive. The basis lies in pre-existing and adaptive resistance mechanisms that circumvent induction of tumor-reactive cytotoxic T cells (CTLs) capable of infiltrating solid tumors and eliminating cancer cells. A priori, cancers induced by expression of human papillomavirus oncogenes should be responsive to immunotherapy: these cancers encode immunogenic neo-antigens – the oncoproteins E6/7 – necessary for their manifestation. Rather, such tumors are poorly responsive to immunotherapies. Results from my lab and others using mouse models of HPV-induced cancer have established an actionable hypothesis: during tumorigenesis, such tumors erect multiple barriers to the induction, infiltration, and killing of cancer cells by tumor antigen-reactive CTLs. These include overarching systemic antigen-nonspecific immunosuppression mediated by expanded populations of myeloid cells in spleen and lymph nodes, complemented by immune response-impairing barriers operative in the tumor microenvironment. A spectrum of models will probe these barriers, genetically and pharmacologically, establishing their functional importance, alone and in concert. A major focus will be on how oncogene-expressing keratinocytes elicit a marked expansion of immunosuppressive myeloid cells in spleen and lymph nodes, and how these myeloid cells in turn inhibit development and activation of CD8 T cells and antigen-presenting dendritic cells. Then we’ll assess the therapeutic potential of barrier-breaking strategies combined with immuno-stimulatory modalities. This project will deliver new knowledge about multi-faceted barriers to immunotherapy in these refractory cancers, helping lay the groundwork for efficacious immunotherapy.
Summary
The frontier in cancer therapy of orchestrating the immune system to attack tumors is producing unprecedented survival benefit in some patients. The corollary is lack of efficacy both in ostensibly responsive tumor types as well as others that are mostly non-responsive. The basis lies in pre-existing and adaptive resistance mechanisms that circumvent induction of tumor-reactive cytotoxic T cells (CTLs) capable of infiltrating solid tumors and eliminating cancer cells. A priori, cancers induced by expression of human papillomavirus oncogenes should be responsive to immunotherapy: these cancers encode immunogenic neo-antigens – the oncoproteins E6/7 – necessary for their manifestation. Rather, such tumors are poorly responsive to immunotherapies. Results from my lab and others using mouse models of HPV-induced cancer have established an actionable hypothesis: during tumorigenesis, such tumors erect multiple barriers to the induction, infiltration, and killing of cancer cells by tumor antigen-reactive CTLs. These include overarching systemic antigen-nonspecific immunosuppression mediated by expanded populations of myeloid cells in spleen and lymph nodes, complemented by immune response-impairing barriers operative in the tumor microenvironment. A spectrum of models will probe these barriers, genetically and pharmacologically, establishing their functional importance, alone and in concert. A major focus will be on how oncogene-expressing keratinocytes elicit a marked expansion of immunosuppressive myeloid cells in spleen and lymph nodes, and how these myeloid cells in turn inhibit development and activation of CD8 T cells and antigen-presenting dendritic cells. Then we’ll assess the therapeutic potential of barrier-breaking strategies combined with immuno-stimulatory modalities. This project will deliver new knowledge about multi-faceted barriers to immunotherapy in these refractory cancers, helping lay the groundwork for efficacious immunotherapy.
Max ERC Funding
2 500 000 €
Duration
Start date: 2020-01-01, End date: 2024-12-31
Project acronym CANCERINNOVATION
Project Using novel methodologies to target and image cancer invasion and therapeutic resistance
Researcher (PI) Margaret Frame
Host Institution (HI) THE UNIVERSITY OF EDINBURGH
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary We aim to develop and apply a suite of new technologies in a novel cancer discovery platform that will link high-definition cancer biology, via state-of-the-art disease imaging and pathway modelling, with development of novel interrogative and therapeutic interventions to test in models of cancer that closely resemble human disease. The work will lead to a new understanding of cancer invasion, how to treat advanced disease in the metastatic niche, how to monitor therapeutic responses and the compensatory mechanisms that cause acquired resistance. Platform development will be based on combined, cross-informing technologies that will enable us to predict optimal ‘maintenance therapies’ for metastatic disease by targeting cancer evolution and spread through combination therapy. A key strand of the platform is the development of quantitative multi-modal imaging in vivo by use of optical window technology to inform detailed understanding of disease and drug mechanisms and predictive capability of pathway biomarkers. Innovative methodologies are urgently needed to address declining approval rates of novel medicines and the unmet clinical needs of treating cancer patients in the advanced disease setting, where tumour spread and survival generally continues unchecked by current therapies. This work will be largely pre-clinical, but will always be mindful of the clinical problem in managing late stage human disease through rationale design of combination therapies with companion diagnostic tests. The cancer survival statistics will be changed if we can curb continuing spread of aggressive, metastatic disease and resistance to therapy by taking smarter combined approaches that make best use of emerging technologies in an innovative way, particularly where they are more predictive of clinical efficacy.
Summary
We aim to develop and apply a suite of new technologies in a novel cancer discovery platform that will link high-definition cancer biology, via state-of-the-art disease imaging and pathway modelling, with development of novel interrogative and therapeutic interventions to test in models of cancer that closely resemble human disease. The work will lead to a new understanding of cancer invasion, how to treat advanced disease in the metastatic niche, how to monitor therapeutic responses and the compensatory mechanisms that cause acquired resistance. Platform development will be based on combined, cross-informing technologies that will enable us to predict optimal ‘maintenance therapies’ for metastatic disease by targeting cancer evolution and spread through combination therapy. A key strand of the platform is the development of quantitative multi-modal imaging in vivo by use of optical window technology to inform detailed understanding of disease and drug mechanisms and predictive capability of pathway biomarkers. Innovative methodologies are urgently needed to address declining approval rates of novel medicines and the unmet clinical needs of treating cancer patients in the advanced disease setting, where tumour spread and survival generally continues unchecked by current therapies. This work will be largely pre-clinical, but will always be mindful of the clinical problem in managing late stage human disease through rationale design of combination therapies with companion diagnostic tests. The cancer survival statistics will be changed if we can curb continuing spread of aggressive, metastatic disease and resistance to therapy by taking smarter combined approaches that make best use of emerging technologies in an innovative way, particularly where they are more predictive of clinical efficacy.
Max ERC Funding
2 499 000 €
Duration
Start date: 2012-08-01, End date: 2017-07-31
Project acronym CAPER/BREAST CANCE
Project CAPER in Invasive Breast Cancer
Researcher (PI) Michael Lisanti
Host Institution (HI) THE UNIVERSITY OF MANCHESTER
Call Details Advanced Grant (AdG), LS7, ERC-2008-AdG
Summary Breast cancer is a major cause of death in the United States and the Western World. Advanced medical technologies and therapeutic strategies are necessary for the successful detection, diagnosis, and treatment of breast cancer. Here, we propose to use novel technologies (tissue microarrays (TMA) and automated quantivative bioimaging (AQUA)) to identify new therapeutic and prognostic markers for human breast cancer. More specifically, we will study the activation status of a new signaling pathway which we have implicated in breast cancer pathogenesis, using both mouse animal models and cells in culture. For this purpose, we will study the association of CAPER expression with pre-malignant lesions and progression from pre-malignancy to full-blown breast cancer. We expect that this new molecular marker will allow us to improve diagnostic accuracy for individual patients, enhancing both the prognostic predictions as well as the prediction of drug responsiveness for a given patient.
Summary
Breast cancer is a major cause of death in the United States and the Western World. Advanced medical technologies and therapeutic strategies are necessary for the successful detection, diagnosis, and treatment of breast cancer. Here, we propose to use novel technologies (tissue microarrays (TMA) and automated quantivative bioimaging (AQUA)) to identify new therapeutic and prognostic markers for human breast cancer. More specifically, we will study the activation status of a new signaling pathway which we have implicated in breast cancer pathogenesis, using both mouse animal models and cells in culture. For this purpose, we will study the association of CAPER expression with pre-malignant lesions and progression from pre-malignancy to full-blown breast cancer. We expect that this new molecular marker will allow us to improve diagnostic accuracy for individual patients, enhancing both the prognostic predictions as well as the prediction of drug responsiveness for a given patient.
Max ERC Funding
1 500 000 €
Duration
Start date: 2010-01-01, End date: 2014-12-31
Project acronym CarnoMorph
Project The Evolution and Development of Complex Morphologies
Researcher (PI) Enrico Coen
Host Institution (HI) JOHN INNES CENTRE
Call Details Advanced Grant (AdG), LS3, ERC-2012-ADG_20120314
Summary Plant and animal organs display a remarkable diversity of shapes. A major challenge in developmental and evolutionary biology is to understand how this diversity of forms is generated. Recent advances in imaging, computational modelling and genomics now make it possible to address this challenge effectively for the first time. Leaf development is a particularly tractable system because of its accessibility to imaging and preservation of connectivity during growth. Leaves also display remarkable diversity in shape and form, with perhaps the most complex form being the pitcher-shaped (epiascidiate) leaves of carnivorous plants. This form has evolved four times independently, raising the question of whether its seeming complexity may have arisen through simple modulations in underlying morphogenetic mechanisms. To test this hypothesis, I aim to develop a model system for carnivorous plants based on Utricularia gibba (humped bladderwort), which has the advantage of having one of the smallest genomes known in plants (~2/3 the size of the Arabidopsis genome) and small transparent pitcher-shaped leaves amenable to imaging. I will use this system to define the morphogenetic events underlying the formation of pitcher-shaped leaves and their molecular genetic control. I will also develop and apply computational modelling to explore hypotheses that may account for the development of U. gibba bladders and further test these hypotheses experimentally. In addition, I will investigate the relationship between U. gibba bladder development and species with simpler leaf shapes, such as Arabidopsis, or species where the epiascidiate form has evolved independently. Taken together, these studies should show how developmental rules elucidated in current model systems might be extended and built upon to account for the diversity and complexity of tissue forms, integrating evo-devo approaches with a mechanistic understanding of morphogenesis.
Summary
Plant and animal organs display a remarkable diversity of shapes. A major challenge in developmental and evolutionary biology is to understand how this diversity of forms is generated. Recent advances in imaging, computational modelling and genomics now make it possible to address this challenge effectively for the first time. Leaf development is a particularly tractable system because of its accessibility to imaging and preservation of connectivity during growth. Leaves also display remarkable diversity in shape and form, with perhaps the most complex form being the pitcher-shaped (epiascidiate) leaves of carnivorous plants. This form has evolved four times independently, raising the question of whether its seeming complexity may have arisen through simple modulations in underlying morphogenetic mechanisms. To test this hypothesis, I aim to develop a model system for carnivorous plants based on Utricularia gibba (humped bladderwort), which has the advantage of having one of the smallest genomes known in plants (~2/3 the size of the Arabidopsis genome) and small transparent pitcher-shaped leaves amenable to imaging. I will use this system to define the morphogenetic events underlying the formation of pitcher-shaped leaves and their molecular genetic control. I will also develop and apply computational modelling to explore hypotheses that may account for the development of U. gibba bladders and further test these hypotheses experimentally. In addition, I will investigate the relationship between U. gibba bladder development and species with simpler leaf shapes, such as Arabidopsis, or species where the epiascidiate form has evolved independently. Taken together, these studies should show how developmental rules elucidated in current model systems might be extended and built upon to account for the diversity and complexity of tissue forms, integrating evo-devo approaches with a mechanistic understanding of morphogenesis.
Max ERC Funding
2 499 997 €
Duration
Start date: 2013-06-01, End date: 2018-05-31
Project acronym CASINO
Project Carbohydrate signals controlling nodulation
Researcher (PI) Jens Stougaard Jensen
Host Institution (HI) AARHUS UNIVERSITET
Call Details Advanced Grant (AdG), LS3, ERC-2010-AdG_20100317
Summary Mechanisms governing interaction between multicellular organisms and microbes are central for understanding pathogenesis, symbiosis and the function of ecosystems. We propose to address these mechanisms by pioneering an interdisciplinary approach for understanding cellular signalling, response processes and organ development. The challenge is to determine factors synchronising three processes, organogenesis, infection thread formation and bacterial infection, running in parallel to build a root nodule hosting symbiotic bacteria. We aim to exploit the unique possibilities for analysing endocytosis of bacteria in model legumes and to develop genomic, genetic and biological chemistry tools to break new ground in our understanding of carbohydrates in plant development and plant-microbe interaction. Surface exposed rhizobial polysaccharides play a crucial but poorly understood role in infection thread formation and rhizobial invasion resulting in endocytosis. We will undertake an integrated functional characterisation of receptor-ligand mechanisms mediating recognition of secreted polysaccharides and subsequent signal amplification. So far progress in this field has been limited by the complex nature of carbohydrate polymers, lack of a suitable experimental model system where both partners in an interaction could be manipulated and lack of corresponding methods for carbohydrate synthesis, analysis and interaction studies. In this context our legume model system and the discovery that the legume Nod-factor receptors recognise bacterial lipochitin-oligosaccharide signals at their LysM domains provides a new opportunity. Combined with advanced bioorganic chemistry and nanobioscience approaches this proposal will engage the above mentioned limitations.
Summary
Mechanisms governing interaction between multicellular organisms and microbes are central for understanding pathogenesis, symbiosis and the function of ecosystems. We propose to address these mechanisms by pioneering an interdisciplinary approach for understanding cellular signalling, response processes and organ development. The challenge is to determine factors synchronising three processes, organogenesis, infection thread formation and bacterial infection, running in parallel to build a root nodule hosting symbiotic bacteria. We aim to exploit the unique possibilities for analysing endocytosis of bacteria in model legumes and to develop genomic, genetic and biological chemistry tools to break new ground in our understanding of carbohydrates in plant development and plant-microbe interaction. Surface exposed rhizobial polysaccharides play a crucial but poorly understood role in infection thread formation and rhizobial invasion resulting in endocytosis. We will undertake an integrated functional characterisation of receptor-ligand mechanisms mediating recognition of secreted polysaccharides and subsequent signal amplification. So far progress in this field has been limited by the complex nature of carbohydrate polymers, lack of a suitable experimental model system where both partners in an interaction could be manipulated and lack of corresponding methods for carbohydrate synthesis, analysis and interaction studies. In this context our legume model system and the discovery that the legume Nod-factor receptors recognise bacterial lipochitin-oligosaccharide signals at their LysM domains provides a new opportunity. Combined with advanced bioorganic chemistry and nanobioscience approaches this proposal will engage the above mentioned limitations.
Max ERC Funding
2 399 127 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym CDK6-DrugOpp
Project CDK6 in transcription - turning a foe in a friend
Researcher (PI) Veronika SEXL
Host Institution (HI) VETERINAERMEDIZINISCHE UNIVERSITAET WIEN
Call Details Advanced Grant (AdG), LS7, ERC-2015-AdG
Summary "Translational research aims at applying mechanistic understanding in the development of "precision medicine", which depends on precise diagnostic tools and therapeutic approaches. Cancer therapy is experiencing a switch from non-specific, cytotoxic agents towards molecularly targeted and rationally designed compounds with the promise of greater efficacy and fewer side effects.
The two cell-cycle kinases CDK4 and CDK6 normally facilitate cell-cycle progression but are abnormally activated in certain cancers. CDK6 is up-regulated in hematopoietic malignancies, where it is the predominant cell-cycle kinase. The importance of CDK4/6 for tumor development is underscored by the fact that the US FDA selected inhibitors of the kinase activity of CDK4/6 as "breakthrough of the year 2013". Our recent findings suggest that the effects of the inhibitors may be limited as CDK6 is not only involved in cell-cycle progression: ground-breaking research in my group and others has shown that CDK6 is involved in regulation of transcription in a kinase-independent manner thereby driving the proliferation of leukemic stem cells and tumor formation. We have now identified mutations in CDK6 that convert it from a tumor promoter into a tumor suppressor. This unexpected outcome is accompanied by a distinct transcriptional profile. Separating the tumor-promoting from the tumor suppressive functions may open a novel therapeutic avenue for drug development. We aim at understanding which domains and residues of CDK6 are involved in rewiring the transcriptional landscape to pave the way for sophisticated inhibitors. The idea of turning a cancer cell's own most potent weapon against itself is novel and would represent a new paradigm for drug design. Finally, the understanding of CDK6 functions in tumor promotion and maintenance will also result in better patient stratification and improved treatment decisions for a broad spectrum of cancer types."
Summary
"Translational research aims at applying mechanistic understanding in the development of "precision medicine", which depends on precise diagnostic tools and therapeutic approaches. Cancer therapy is experiencing a switch from non-specific, cytotoxic agents towards molecularly targeted and rationally designed compounds with the promise of greater efficacy and fewer side effects.
The two cell-cycle kinases CDK4 and CDK6 normally facilitate cell-cycle progression but are abnormally activated in certain cancers. CDK6 is up-regulated in hematopoietic malignancies, where it is the predominant cell-cycle kinase. The importance of CDK4/6 for tumor development is underscored by the fact that the US FDA selected inhibitors of the kinase activity of CDK4/6 as "breakthrough of the year 2013". Our recent findings suggest that the effects of the inhibitors may be limited as CDK6 is not only involved in cell-cycle progression: ground-breaking research in my group and others has shown that CDK6 is involved in regulation of transcription in a kinase-independent manner thereby driving the proliferation of leukemic stem cells and tumor formation. We have now identified mutations in CDK6 that convert it from a tumor promoter into a tumor suppressor. This unexpected outcome is accompanied by a distinct transcriptional profile. Separating the tumor-promoting from the tumor suppressive functions may open a novel therapeutic avenue for drug development. We aim at understanding which domains and residues of CDK6 are involved in rewiring the transcriptional landscape to pave the way for sophisticated inhibitors. The idea of turning a cancer cell's own most potent weapon against itself is novel and would represent a new paradigm for drug design. Finally, the understanding of CDK6 functions in tumor promotion and maintenance will also result in better patient stratification and improved treatment decisions for a broad spectrum of cancer types."
Max ERC Funding
2 497 520 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym CELLMIG
Project Molecular and Cellular Mechanisms Promoting Single-Cell Migration in vivo
Researcher (PI) Erez Raz
Host Institution (HI) WESTFAELISCHE WILHELMS-UNIVERSITAET MUENSTER
Call Details Advanced Grant (AdG), LS3, ERC-2010-AdG_20100317
Summary The regulation of cell migration is central in pattern formation, homeostasis and disease. The proposed research is aimed at investigating the molecular basis for cell motility and the associated polarization of the cell. In view of the dynamic nature of these processes, we have chosen to utilize the migration of Primoridal Germ Cells (PGCs) in zebrafish - a model that offers unique experimental advantages for imaging and experimental manipulations. The fact that molecules facilitating the motility of zebrafish PGCs are evolutionary conserved and the finding that the cells are directed by chemokines, molecules that control a wide range of cell trafficking events in vertebrates, make this in vivo study of particular importance.
The proposed work involves both the functional analysis of previously identified candidates and the identification of molecules, which have a presently unknown effect on the migration process. For both objectives, we will employ novel experimental schemes. We will examine the role of proteins in achieving functional cell polarity compatible with efficient motility and response to directional cues, using unique techniques and analysis tools in the context of the living organism. The precise function of the identified proteins will be determined by combining mathematical tools aimed at quantitatively gauging the role of the molecules in conferring proper cell shape, biophysical methods aimed at measuring forces, rigidity and cytoplasm flow and determination of the effect on the organization of relevant structures using cryo electron tomography.
Together, this approach would provide a non-conventional understanding of cell migration by correlating structural, morphological and dynamic cellular properties with the ability of cells to effectively migrate towards their target.
Summary
The regulation of cell migration is central in pattern formation, homeostasis and disease. The proposed research is aimed at investigating the molecular basis for cell motility and the associated polarization of the cell. In view of the dynamic nature of these processes, we have chosen to utilize the migration of Primoridal Germ Cells (PGCs) in zebrafish - a model that offers unique experimental advantages for imaging and experimental manipulations. The fact that molecules facilitating the motility of zebrafish PGCs are evolutionary conserved and the finding that the cells are directed by chemokines, molecules that control a wide range of cell trafficking events in vertebrates, make this in vivo study of particular importance.
The proposed work involves both the functional analysis of previously identified candidates and the identification of molecules, which have a presently unknown effect on the migration process. For both objectives, we will employ novel experimental schemes. We will examine the role of proteins in achieving functional cell polarity compatible with efficient motility and response to directional cues, using unique techniques and analysis tools in the context of the living organism. The precise function of the identified proteins will be determined by combining mathematical tools aimed at quantitatively gauging the role of the molecules in conferring proper cell shape, biophysical methods aimed at measuring forces, rigidity and cytoplasm flow and determination of the effect on the organization of relevant structures using cryo electron tomography.
Together, this approach would provide a non-conventional understanding of cell migration by correlating structural, morphological and dynamic cellular properties with the ability of cells to effectively migrate towards their target.
Max ERC Funding
1 960 600 €
Duration
Start date: 2011-06-01, End date: 2017-05-31
Project acronym CENDUP
Project Decoding the mechanisms of centrosome duplication
Researcher (PI) Pierre Gönczy
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), LS3, ERC-2008-AdG
Summary Centrosome duplication entails the formation of a single procentriole next to each centriole once per cell cycle. The mechanisms governing procentriole formation are poorly understood and constitute a fundamental open question in cell biology. We will launch an innovative multidisciplinary research program to gain significant insight into these mechanisms using C. elegans and human cells. This research program is also expected to have a significant impact by contributing important novel assays to the field. Six specific aims will be pursued: 1) SAS-6 as a ZYG-1 substrate: mechanisms of procentriole formation in C. elegans. We will test in vivo the consequence of SAS-6 phosphorylation by ZYG-1. 2) Biochemical and structural analysis of SAS-6-containing macromolecular complexes (SAMACs). We will isolate and characterize SAMACs from C. elegans embryos and human cells, and analyze their structure using single-particle electron microscopy. 3) Novel cell-free assay for procentriole formation in human cells. We will develop such an assay and use it to test whether SAMACs can direct procentriole formation and whether candidate proteins are needed at centrioles or in the cytoplasm. 4) Mapping interactions between centriolar proteins in live human cells. We will use chemical methods developed by our collaborators to probe interactions between HsSAS-6 and centriolar proteins in a time- and space-resolved manner. 5) Functional genomic and chemical genetic screens in human cells. We will conduct high-throughput fluorescence-based screens in human cells to identify novel genes required for procentriole formation and small molecule inhibitors of this process. 6) Mechanisms underlying differential centriolar maintenance in the germline. In C. elegans, we will characterize how the sas-1 locus is required for centriole maintenance during spermatogenesis, as well as analyze centriole elimination during oogenesis and identify components needed for this process
Summary
Centrosome duplication entails the formation of a single procentriole next to each centriole once per cell cycle. The mechanisms governing procentriole formation are poorly understood and constitute a fundamental open question in cell biology. We will launch an innovative multidisciplinary research program to gain significant insight into these mechanisms using C. elegans and human cells. This research program is also expected to have a significant impact by contributing important novel assays to the field. Six specific aims will be pursued: 1) SAS-6 as a ZYG-1 substrate: mechanisms of procentriole formation in C. elegans. We will test in vivo the consequence of SAS-6 phosphorylation by ZYG-1. 2) Biochemical and structural analysis of SAS-6-containing macromolecular complexes (SAMACs). We will isolate and characterize SAMACs from C. elegans embryos and human cells, and analyze their structure using single-particle electron microscopy. 3) Novel cell-free assay for procentriole formation in human cells. We will develop such an assay and use it to test whether SAMACs can direct procentriole formation and whether candidate proteins are needed at centrioles or in the cytoplasm. 4) Mapping interactions between centriolar proteins in live human cells. We will use chemical methods developed by our collaborators to probe interactions between HsSAS-6 and centriolar proteins in a time- and space-resolved manner. 5) Functional genomic and chemical genetic screens in human cells. We will conduct high-throughput fluorescence-based screens in human cells to identify novel genes required for procentriole formation and small molecule inhibitors of this process. 6) Mechanisms underlying differential centriolar maintenance in the germline. In C. elegans, we will characterize how the sas-1 locus is required for centriole maintenance during spermatogenesis, as well as analyze centriole elimination during oogenesis and identify components needed for this process
Max ERC Funding
2 004 155 €
Duration
Start date: 2009-04-01, End date: 2014-03-31
Project acronym CENFOR
Project Dissecting the mechanisms governing centriole formation
Researcher (PI) Pierre Gönczy
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), LS3, ERC-2013-ADG
Summary "Centrioles are critical for the formation of cilia, flagella and centrosomes, as well as for human health. The mechanisms governing centriole formation constitute a long-standing question in cell biology. We will pursue an innovative multidisciplinary research program to gain further insight into these mechanisms, using human cells, C. elegans and Trichonympha as model systems. This program is expected to also have a major impact by contributing a novel cell free assay to the field, thus paving the way towards making synthetic centrioles. Six specific aims will be pursued:
1) Deciphering HsSAS-6/STIL distribution and dynamics. We will use super-resolution microscopy, molecular counting, photoconversion and FCS to further characterize these two key components required for centriole formation in human cells.
2) The SAS-6 ring model as a tool to redirect centriole organization. Utilizing predictions from the SAS-6 ring model, we will assay the consequences for centrioles and cilia of altering the diameter and symmetry of the structure.
3) Determining the architecture of C. elegans centrioles. We will conduct molecular counting and cryo-ET of purified C. elegans centrioles to determine if they contain a spiral or a cartwheel, as well as identify SAS-6-interacting components.
4) Comprehensive 3D map and proteomics of Trichonympha centriole. We will obtain a ~35 Å 3D map of the complete T. agilis centriole, perform proteomic analysis to identify its constituents and test their function using RNAi.
5) Regulation of cartwheel height and centriole length. We will explore whether cartwheel height is set by SAS-6 proteins and perform screens in human cells to identify novel components regulating cartwheel height and centriole length.
6) Novel cell free assay for cartwheel assembly and centriole formation. Using SAS-6 proteins on a lipid monolayer as starting point, we will develop and utilize a cell-free assay to reconstitute cartwheel assembly and centriole format"
Summary
"Centrioles are critical for the formation of cilia, flagella and centrosomes, as well as for human health. The mechanisms governing centriole formation constitute a long-standing question in cell biology. We will pursue an innovative multidisciplinary research program to gain further insight into these mechanisms, using human cells, C. elegans and Trichonympha as model systems. This program is expected to also have a major impact by contributing a novel cell free assay to the field, thus paving the way towards making synthetic centrioles. Six specific aims will be pursued:
1) Deciphering HsSAS-6/STIL distribution and dynamics. We will use super-resolution microscopy, molecular counting, photoconversion and FCS to further characterize these two key components required for centriole formation in human cells.
2) The SAS-6 ring model as a tool to redirect centriole organization. Utilizing predictions from the SAS-6 ring model, we will assay the consequences for centrioles and cilia of altering the diameter and symmetry of the structure.
3) Determining the architecture of C. elegans centrioles. We will conduct molecular counting and cryo-ET of purified C. elegans centrioles to determine if they contain a spiral or a cartwheel, as well as identify SAS-6-interacting components.
4) Comprehensive 3D map and proteomics of Trichonympha centriole. We will obtain a ~35 Å 3D map of the complete T. agilis centriole, perform proteomic analysis to identify its constituents and test their function using RNAi.
5) Regulation of cartwheel height and centriole length. We will explore whether cartwheel height is set by SAS-6 proteins and perform screens in human cells to identify novel components regulating cartwheel height and centriole length.
6) Novel cell free assay for cartwheel assembly and centriole formation. Using SAS-6 proteins on a lipid monolayer as starting point, we will develop and utilize a cell-free assay to reconstitute cartwheel assembly and centriole format"
Max ERC Funding
2 499 270 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym CENGIN
Project Deciphering and engineering centriole assembly
Researcher (PI) Pierre Jörg GÖNCZY
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), LS3, ERC-2018-ADG
Summary Deciphering and engineering the assembly of cellular organelles is a key pursuit in biology. The centriole is an evolutionarily conserved organelle well suited for this goal, and which is crucial for cell signaling, motility and division. The centriole exhibits a striking 9-fold radial symmetry of microtubules around a likewise symmetrical cartwheel containing stacked ring-bearing structures. Components essential for generating this remarkable architecture from alga to man have been identified. A next critical step is to engineer assays to probe the dynamics of centriole assembly with molecular precision to fully understand how these components together build a functional organelle. Our ambitious research proposal aims at taking groundbreaking steps in this direction through four specific aims:
1) Reconstituting cartwheel ring assembly dynamics. We will use high-speed AFM (HS-AFM) to dissect the biophysics of SAS-6 ring polymer dynamics at the root of cartwheel assembly. We will also use HS-AFM to analyze monobodies against SAS-6, as well as engineer surfaces and DNA origamis to further dissect ring assembly.
2) Deciphering ring stacking mechanisms. We will use cryo-ET to identify SAS-6 features that direct stacking of ring structures and set cartwheel height. Moreover, we will develop an HS-AFM stacking assay and a reconstituted stacking assay from human cells.
3) Understanding peripheral element contributions to centriole biogenesis. We will dissect the function of the peripheral centriole pinhead protein Cep135/Bld10p, as well as identify and likewise dissect peripheral A-C linker proteins. Furthermore, we will further engineer the HS-AFM assay to include such peripheral components.
4) Dissecting de novo centriole assembly mechanisms. We will dissect de novo centriole formation in human cells and water fern. We will also explore whether de novo formation involves a phase separation mechanism and repurpose the HS-AFM assay to probe de novo organelle biogenes
Summary
Deciphering and engineering the assembly of cellular organelles is a key pursuit in biology. The centriole is an evolutionarily conserved organelle well suited for this goal, and which is crucial for cell signaling, motility and division. The centriole exhibits a striking 9-fold radial symmetry of microtubules around a likewise symmetrical cartwheel containing stacked ring-bearing structures. Components essential for generating this remarkable architecture from alga to man have been identified. A next critical step is to engineer assays to probe the dynamics of centriole assembly with molecular precision to fully understand how these components together build a functional organelle. Our ambitious research proposal aims at taking groundbreaking steps in this direction through four specific aims:
1) Reconstituting cartwheel ring assembly dynamics. We will use high-speed AFM (HS-AFM) to dissect the biophysics of SAS-6 ring polymer dynamics at the root of cartwheel assembly. We will also use HS-AFM to analyze monobodies against SAS-6, as well as engineer surfaces and DNA origamis to further dissect ring assembly.
2) Deciphering ring stacking mechanisms. We will use cryo-ET to identify SAS-6 features that direct stacking of ring structures and set cartwheel height. Moreover, we will develop an HS-AFM stacking assay and a reconstituted stacking assay from human cells.
3) Understanding peripheral element contributions to centriole biogenesis. We will dissect the function of the peripheral centriole pinhead protein Cep135/Bld10p, as well as identify and likewise dissect peripheral A-C linker proteins. Furthermore, we will further engineer the HS-AFM assay to include such peripheral components.
4) Dissecting de novo centriole assembly mechanisms. We will dissect de novo centriole formation in human cells and water fern. We will also explore whether de novo formation involves a phase separation mechanism and repurpose the HS-AFM assay to probe de novo organelle biogenes
Max ERC Funding
2 500 000 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym ChAMPioN
Project Game-changing Precision Medicine for Curing All Myeloproliferative Neoplasms
Researcher (PI) Tessa Holyoake
Host Institution (HI) UNIVERSITY OF GLASGOW
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Despite decades of research, developing ways to overcome drug resistance in cancer is the most challenging bottleneck for curative therapies. This is because, in some forms of cancer, the cancer stem cells from which the diseases arise are constantly evolving, particularly under the selective pressures of drug therapies, in order to survive. The events leading to drug resistance can occur within one or more individual cancer stem cell(s) – and the features of each of these cells need to be studied in detail in order to develop drugs or drug combinations that can eradicate all of them. The BCR-ABL+ and BCR-ABL- myeloproliferative neoplasms (MPN) are a group of proliferative blood diseases that can be considered both exemplars of precision medicine and of the drug resistance bottleneck. While significant advances in the management of MPN have been made using life-long and expensive tyrosine kinase inhibitors (TKI), patients are rarely cured of their disease. This is because TKI fail to eradicate the leukaemia stem cells (LSC) from which MPN arise and which persist in patients on treatment, often leading to pervasive drug resistance, loss of response to therapy and progression to fatal forms of acute leukaemia. My goal is to change the way we study the LSC that persist in MPN patients as a means of delivering more effective precision medicine in MPN that is a “game-changer” leading to therapy-free remission (TFR) and cure. Here, I will apply an innovative strategy, ChAMPioN, to study the response of the MPN LSC to TKI in innovative pre-clinical laboratory models and directly in patients with MPN - up to the resolution of individual LSC. This work will reveal, for the first time, the molecular and clonal evolution of LSC during TKI therapies, thus enabling the development of more accurate predictions of TKI efficacy and resistance and rational approaches for curative drug therapies.
Summary
Despite decades of research, developing ways to overcome drug resistance in cancer is the most challenging bottleneck for curative therapies. This is because, in some forms of cancer, the cancer stem cells from which the diseases arise are constantly evolving, particularly under the selective pressures of drug therapies, in order to survive. The events leading to drug resistance can occur within one or more individual cancer stem cell(s) – and the features of each of these cells need to be studied in detail in order to develop drugs or drug combinations that can eradicate all of them. The BCR-ABL+ and BCR-ABL- myeloproliferative neoplasms (MPN) are a group of proliferative blood diseases that can be considered both exemplars of precision medicine and of the drug resistance bottleneck. While significant advances in the management of MPN have been made using life-long and expensive tyrosine kinase inhibitors (TKI), patients are rarely cured of their disease. This is because TKI fail to eradicate the leukaemia stem cells (LSC) from which MPN arise and which persist in patients on treatment, often leading to pervasive drug resistance, loss of response to therapy and progression to fatal forms of acute leukaemia. My goal is to change the way we study the LSC that persist in MPN patients as a means of delivering more effective precision medicine in MPN that is a “game-changer” leading to therapy-free remission (TFR) and cure. Here, I will apply an innovative strategy, ChAMPioN, to study the response of the MPN LSC to TKI in innovative pre-clinical laboratory models and directly in patients with MPN - up to the resolution of individual LSC. This work will reveal, for the first time, the molecular and clonal evolution of LSC during TKI therapies, thus enabling the development of more accurate predictions of TKI efficacy and resistance and rational approaches for curative drug therapies.
Max ERC Funding
3 005 818 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym Cholstim
Project Cholinergic modulation of immune homeostasis: new opportunities for treatment
Researcher (PI) Guy Eduard Elisabeth Boeckxstaens
Host Institution (HI) KATHOLIEKE UNIVERSITEIT LEUVEN
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary In the gastrointestinal tract, the balance between activation of the mucosal immune system and tolerance should be tightly regulated to maintain immune homeostasis to prevent chronic inflammation and tissue damage. Recently, the new concept was introduced that the vagus nerve plays an important role in modulating immune homeostasis as part of a so-called inflammatory reflex. We provided evidence for this concept in the gastrointestinal tract and showed that vagus nerve stimulation (VNS) reduced inflammation of the intestinal muscle layer. Moreover, we showed that this effect was mediated by activation of enteric cholinergic neurons (cholinergic tone) interacting with intestinal macrophages in the muscle layer. Of interest, we have collected exciting data that the vagus nerve (and thus the cholinergic tone) also significantly contributes to mucosal immune homeostasis. Mice that underwent vagotomy lost their ability to develop tolerance to oral feeding of an antigen, whereas VNS reduced mucosal inflammation in a model of food allergy. Based on these data, we hypothesize that the cholinergic tone is a major determinant of the tolerogenic microenvironment of the mucosal immune system, and want to further explore the immune-modulatory effect of the vagal innervation and enteric neurons on the macrophages residing in the lamina propria. In addition, we will further explore the therapeutic potential and the mechanisms involved of chronic VNS in colitis and food allergy. Finally, we will translate our preclinical findings to the human situation. The anti-inflammatory effect of VNS (applied during surgery) will be studied in human intestinal tissue whereas the therapeutic potential of chronic VNS in Crohn’s disease will be studied in a pilot trial.
The outcome of this project will be ground-breaking and will have an immense impact on clinical management as it will provide new therapeutic opportunities for the treatment of immune-mediated gastrointestinal disorders.
Summary
In the gastrointestinal tract, the balance between activation of the mucosal immune system and tolerance should be tightly regulated to maintain immune homeostasis to prevent chronic inflammation and tissue damage. Recently, the new concept was introduced that the vagus nerve plays an important role in modulating immune homeostasis as part of a so-called inflammatory reflex. We provided evidence for this concept in the gastrointestinal tract and showed that vagus nerve stimulation (VNS) reduced inflammation of the intestinal muscle layer. Moreover, we showed that this effect was mediated by activation of enteric cholinergic neurons (cholinergic tone) interacting with intestinal macrophages in the muscle layer. Of interest, we have collected exciting data that the vagus nerve (and thus the cholinergic tone) also significantly contributes to mucosal immune homeostasis. Mice that underwent vagotomy lost their ability to develop tolerance to oral feeding of an antigen, whereas VNS reduced mucosal inflammation in a model of food allergy. Based on these data, we hypothesize that the cholinergic tone is a major determinant of the tolerogenic microenvironment of the mucosal immune system, and want to further explore the immune-modulatory effect of the vagal innervation and enteric neurons on the macrophages residing in the lamina propria. In addition, we will further explore the therapeutic potential and the mechanisms involved of chronic VNS in colitis and food allergy. Finally, we will translate our preclinical findings to the human situation. The anti-inflammatory effect of VNS (applied during surgery) will be studied in human intestinal tissue whereas the therapeutic potential of chronic VNS in Crohn’s disease will be studied in a pilot trial.
The outcome of this project will be ground-breaking and will have an immense impact on clinical management as it will provide new therapeutic opportunities for the treatment of immune-mediated gastrointestinal disorders.
Max ERC Funding
2 495 200 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym CHROMOCOND
Project A molecular view of chromosome condensation
Researcher (PI) Frank Uhlmann
Host Institution (HI) CANCER RESEARCH UK LBG
Call Details Advanced Grant (AdG), LS3, ERC-2009-AdG
Summary Eukaryotic cells inherit much of their genomic information in the form of chromosomes during cell division. Centimetre-long DNA molecules are packed into micrometer-sized chromosomes to enable this process. How DNA is organised within mitotic chromosomes is still largely unknown. A key structural protein component of mitotic chromosomes, implicated in their compaction, is the condensin complex. In this proposal, we aim to elucidate the molecular architecture of mitotic chromosomes, taking advantage of new genomic techniques and the relatively simple genome organisation of yeast model systems. We will place particular emphasis on elucidating the contribution of the condensin complex, and the cell cycle regulation of its activities, in promoting chromosome condensation. Our previous work has provided genome-wide maps of condensin binding to budding and fission yeast chromosomes. We will continue to decipher the molecular determinants for condensin binding. To investigate how condensin mediates DNA compaction, we propose to generate chromosome-wide DNA/DNA proximity maps. Our approach will be an extension of the chromosome conformation capture (3C) technique. High throughput sequencing of interaction points has provided a first glimpse of the interactions that govern chromosome condensation. The role that condensin plays in promoting these interactions will be investigated. The contribution of condensin s ATP-dependent activities, and cell cycle-dependent post-translational modifications, will be studied. This will be complemented by mathematical modelling of the condensation process. In addition to chromosome condensation, condensin is required for resolution of sister chromatids in anaphase. We will develop an assay to study the catenation status of sister chromatids and how condensin may contribute to their topological resolution.
Summary
Eukaryotic cells inherit much of their genomic information in the form of chromosomes during cell division. Centimetre-long DNA molecules are packed into micrometer-sized chromosomes to enable this process. How DNA is organised within mitotic chromosomes is still largely unknown. A key structural protein component of mitotic chromosomes, implicated in their compaction, is the condensin complex. In this proposal, we aim to elucidate the molecular architecture of mitotic chromosomes, taking advantage of new genomic techniques and the relatively simple genome organisation of yeast model systems. We will place particular emphasis on elucidating the contribution of the condensin complex, and the cell cycle regulation of its activities, in promoting chromosome condensation. Our previous work has provided genome-wide maps of condensin binding to budding and fission yeast chromosomes. We will continue to decipher the molecular determinants for condensin binding. To investigate how condensin mediates DNA compaction, we propose to generate chromosome-wide DNA/DNA proximity maps. Our approach will be an extension of the chromosome conformation capture (3C) technique. High throughput sequencing of interaction points has provided a first glimpse of the interactions that govern chromosome condensation. The role that condensin plays in promoting these interactions will be investigated. The contribution of condensin s ATP-dependent activities, and cell cycle-dependent post-translational modifications, will be studied. This will be complemented by mathematical modelling of the condensation process. In addition to chromosome condensation, condensin is required for resolution of sister chromatids in anaphase. We will develop an assay to study the catenation status of sister chromatids and how condensin may contribute to their topological resolution.
Max ERC Funding
2 076 126 €
Duration
Start date: 2010-04-01, End date: 2015-03-31
Project acronym ChroNeuroRepair
Project Chromatin states in neurogenesis – from understanding chromatin loops to eliciting neurogenesis for repair
Researcher (PI) Magdalena Götz
Host Institution (HI) HELMHOLTZ ZENTRUM MUENCHEN DEUTSCHES FORSCHUNGSZENTRUM FUER GESUNDHEIT UND UMWELT GMBH
Call Details Advanced Grant (AdG), LS3, ERC-2013-ADG
Summary The mechanisms regulating neural stem cells and their progression to neurogenesis are important not only to understand brain development and evolution, but also to elicit neurogenesis after brain injury. Our recent findings imply novel chromatin-associated proteins in the regulation of neural stem cell fate and neurogenesis. Therefore this project aims to understand the molecular mechanisms of how these factors regulate neurogenesis in developing and adult mice (Aim1) and implement this knowledge for reprogramming glia into neurons after brain injury (Aim2). This will be pursued in mouse models in vivo (2.1) and with human glial cells derived from patient brain resections in vitro (2.2). It is well known that transcription factors need to alter the chromatin structure to achieve transcriptional regulation, but the factors involved in this regulation in neural stem and progenitor cells are still ill understood. Therefore the molecular function of the novel chromatin interacting protein Trnp1 with essential roles in neural stem cell (NSC) fate and the chromatin conformation mediated at neurogenic target genes by Pax6/Brg1-containing BAF complexes will be addressed in Aim1. Combined with genome-wide approaches to determine changes in chromatin conformation at neurogenic target gene sites this will greatly further our understanding of key roles of chromatin conformation in neural stem cells and neurogenesis. In Aim2 Trnp1 promoting neural stem cells fate and later acting neurogenic transcription factors will be used to improve neuronal reprogramming after stab wound injury in the murine brain in vivo and in patient-derived glial cells in vitro. Together with novel strategies to induce chromatin looping in a sequence-specific manner this project will not only advance our knowledge at the frontier of transcriptional regulation in neurogenesis, but also implement highly innovative approaches to utilize this knowledge for neuronal repair by direct reprogramming.
Summary
The mechanisms regulating neural stem cells and their progression to neurogenesis are important not only to understand brain development and evolution, but also to elicit neurogenesis after brain injury. Our recent findings imply novel chromatin-associated proteins in the regulation of neural stem cell fate and neurogenesis. Therefore this project aims to understand the molecular mechanisms of how these factors regulate neurogenesis in developing and adult mice (Aim1) and implement this knowledge for reprogramming glia into neurons after brain injury (Aim2). This will be pursued in mouse models in vivo (2.1) and with human glial cells derived from patient brain resections in vitro (2.2). It is well known that transcription factors need to alter the chromatin structure to achieve transcriptional regulation, but the factors involved in this regulation in neural stem and progenitor cells are still ill understood. Therefore the molecular function of the novel chromatin interacting protein Trnp1 with essential roles in neural stem cell (NSC) fate and the chromatin conformation mediated at neurogenic target genes by Pax6/Brg1-containing BAF complexes will be addressed in Aim1. Combined with genome-wide approaches to determine changes in chromatin conformation at neurogenic target gene sites this will greatly further our understanding of key roles of chromatin conformation in neural stem cells and neurogenesis. In Aim2 Trnp1 promoting neural stem cells fate and later acting neurogenic transcription factors will be used to improve neuronal reprogramming after stab wound injury in the murine brain in vivo and in patient-derived glial cells in vitro. Together with novel strategies to induce chromatin looping in a sequence-specific manner this project will not only advance our knowledge at the frontier of transcriptional regulation in neurogenesis, but also implement highly innovative approaches to utilize this knowledge for neuronal repair by direct reprogramming.
Max ERC Funding
2 376 560 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym CleverGenes
Project Novel Gene Therapy Based on the Activation of Endogenous Genes for the Treatment of Ischemia - Concepts of endogenetherapy, release of promoter pausing, promoter-targeted ncRNAs and nuclear RNAi
Researcher (PI) Seppo Ylä-Herttuala
Host Institution (HI) ITA-SUOMEN YLIOPISTO
Call Details Advanced Grant (AdG), LS7, ERC-2014-ADG
Summary Background: Therapeutic angiogenesis with vascular endothelial growth factors (VEGFs) has great potential to become a novel, minimally invasive new treatment for a large number of patients with severe myocardial ischemia. However, this requires development of new technology. Advancing state-of-the-art: We propose a paradigm shift in gene therapy for chronic ischemia by activating endogenous VEGF-A,-B and -C genes and angiogenic transcription programs from the native promoters instead of gene transfer of exogenous cDNA to target tissues. We will develop a new platform technology (endogenetherapy) based on our novel concept of the release of promoter pausing and new promoter-targeted upregulating short hairpinRNAs, tissue-specific superenhancerRNAs activating specific transcription centers involving gene clusters in different chromosomal regions, small circularRNAs formed from self-splicing exons-introns that can be regulated with oligonucleotides and small molecules such as metabolites, nuclear RNAi vectors and specific CRISPR/gRNAmutatedCas9-VP16 technology with an ability to target integration into genomic safe harbor sites. After preclinical studies in mice and in a newly developed chronic cardiac ischemia model in pigs with special emphasis on the analysis of clinically relevant blood flow, metabolic and functional outcomes based on MRI, ultrasound, photoacoustic and PET imaging, the best construct will be taken to a phase I clinical study in patients with severe myocardial ischemia. Since endogenetherapy also involves epigenetic changes, which are reversible and long-lasting, we expect to efficiently activate natural angiogenic programs. Significance: If successful, this approach will begin a new era in gene therapy. Since there is a clear lack of technology capable of targeted upregulation of endogenous genes, the novel endogenetherapy approach may become widely applicable beyond cardiovascular diseases also in other areas of medicine and biomedical research.
Summary
Background: Therapeutic angiogenesis with vascular endothelial growth factors (VEGFs) has great potential to become a novel, minimally invasive new treatment for a large number of patients with severe myocardial ischemia. However, this requires development of new technology. Advancing state-of-the-art: We propose a paradigm shift in gene therapy for chronic ischemia by activating endogenous VEGF-A,-B and -C genes and angiogenic transcription programs from the native promoters instead of gene transfer of exogenous cDNA to target tissues. We will develop a new platform technology (endogenetherapy) based on our novel concept of the release of promoter pausing and new promoter-targeted upregulating short hairpinRNAs, tissue-specific superenhancerRNAs activating specific transcription centers involving gene clusters in different chromosomal regions, small circularRNAs formed from self-splicing exons-introns that can be regulated with oligonucleotides and small molecules such as metabolites, nuclear RNAi vectors and specific CRISPR/gRNAmutatedCas9-VP16 technology with an ability to target integration into genomic safe harbor sites. After preclinical studies in mice and in a newly developed chronic cardiac ischemia model in pigs with special emphasis on the analysis of clinically relevant blood flow, metabolic and functional outcomes based on MRI, ultrasound, photoacoustic and PET imaging, the best construct will be taken to a phase I clinical study in patients with severe myocardial ischemia. Since endogenetherapy also involves epigenetic changes, which are reversible and long-lasting, we expect to efficiently activate natural angiogenic programs. Significance: If successful, this approach will begin a new era in gene therapy. Since there is a clear lack of technology capable of targeted upregulation of endogenous genes, the novel endogenetherapy approach may become widely applicable beyond cardiovascular diseases also in other areas of medicine and biomedical research.
Max ERC Funding
2 437 500 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym CODE
Project Coincidence detection of proteins and lipids in regulation of cellular membrane dynamics
Researcher (PI) Harald STENMARK
Host Institution (HI) UNIVERSITETET I OSLO
Call Details Advanced Grant (AdG), LS3, ERC-2017-ADG
Summary Specific recruitment of different proteins to distinct intracellular membranes is fundamental in the biology of eukaryotic cells, but the molecular basis for specificity is incompletely understood. This proposal investigates the hypothesis that coincidence detection of proteins and lipids constitutes a major mechanism for specific recruitment of proteins to intracellular membranes in order to control cellular membrane dynamics. CODE will establish and validate mathematical models for coincidence detection, identify and functionally characterise novel coincidence detectors, and engineer artificial coincidence detectors as novel tools in cell biology and biotechnology.
Summary
Specific recruitment of different proteins to distinct intracellular membranes is fundamental in the biology of eukaryotic cells, but the molecular basis for specificity is incompletely understood. This proposal investigates the hypothesis that coincidence detection of proteins and lipids constitutes a major mechanism for specific recruitment of proteins to intracellular membranes in order to control cellular membrane dynamics. CODE will establish and validate mathematical models for coincidence detection, identify and functionally characterise novel coincidence detectors, and engineer artificial coincidence detectors as novel tools in cell biology and biotechnology.
Max ERC Funding
2 500 000 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym CODIR
Project Colonic Disease Investigation by Robotic Hydro-colonoscopy
Researcher (PI) Alfred Cuschieri
Host Institution (HI) UNIVERSITY OF DUNDEE
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary CODIR proposes a new configuration of fundamental and applied biomedical and engineering multidisciplinary research to explore and characterise colon behavior necessary for the project and wider objectives. Scope and focus is on novel robotic hydro-colonoscopy (RHC), which stems from two considerations: (i) replacement of the flexible colonosocope with a patient-friendly system for inspection of the mucosal surface colon and (ii) the very recent concept of hydro-colonoscopy whereby water is used instead of traditional air insufflation. RHC can enable a breakthrough in patient-compliant complete endoscopic examination and biopsy of the colon for the further study of life threatening disorders of the colon commonly categorized as inflmmatory bowel disease, all of unknown aetilogy despite intensive research. CODIR will provide new insights for biomedical investigation and research applicable to many biomedical fields: biologic [absorption of water and electrolyte from the colon, characterisation of surface topograpgy of the colon, mechanical properties of colonic wall], imaging, mechatronics robot functionality and a novel colonic irrigation and filling system. The ambition is to develop a one-stop holistic system which cleans the colon of faecal debris and then introduces a tethered swimming/ submerging robot for inspection of the mucosal aspect of colon under the control of a clinician operating the endoluminal mini-robot from a control console. A secondary, very important outcome of CODIR is to increase patient compliance (currently 50%) for screening colonoscopy in early diagnosis of colorectal cancer, the worlds second commonest cancer. RHC can overcome major disadvantages of existing colonoscopy examination: discomfort, sedation, thus increasing compliance and enabling future research.
Summary
CODIR proposes a new configuration of fundamental and applied biomedical and engineering multidisciplinary research to explore and characterise colon behavior necessary for the project and wider objectives. Scope and focus is on novel robotic hydro-colonoscopy (RHC), which stems from two considerations: (i) replacement of the flexible colonosocope with a patient-friendly system for inspection of the mucosal surface colon and (ii) the very recent concept of hydro-colonoscopy whereby water is used instead of traditional air insufflation. RHC can enable a breakthrough in patient-compliant complete endoscopic examination and biopsy of the colon for the further study of life threatening disorders of the colon commonly categorized as inflmmatory bowel disease, all of unknown aetilogy despite intensive research. CODIR will provide new insights for biomedical investigation and research applicable to many biomedical fields: biologic [absorption of water and electrolyte from the colon, characterisation of surface topograpgy of the colon, mechanical properties of colonic wall], imaging, mechatronics robot functionality and a novel colonic irrigation and filling system. The ambition is to develop a one-stop holistic system which cleans the colon of faecal debris and then introduces a tethered swimming/ submerging robot for inspection of the mucosal aspect of colon under the control of a clinician operating the endoluminal mini-robot from a control console. A secondary, very important outcome of CODIR is to increase patient compliance (currently 50%) for screening colonoscopy in early diagnosis of colorectal cancer, the worlds second commonest cancer. RHC can overcome major disadvantages of existing colonoscopy examination: discomfort, sedation, thus increasing compliance and enabling future research.
Max ERC Funding
2 999 948 €
Duration
Start date: 2011-08-01, End date: 2016-07-31
Project acronym COGATIMABIO
Project Combined time domain and spectral domain coherence gating for imaging and biosensing
Researcher (PI) Adrian Podoleanu
Host Institution (HI) UNIVERSITY OF KENT
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary Revolutionary combination of principles of spectral domain and time domain coherence gating will be researched. The present proposal puts forward: (i) a novel class of optical interferometers, (ii) a novel class of wavefront sensors and (iii) combinations of imaging channels with the novel wavefront sensors. All these are driven by the needs to address the limitations in terms of speed of the time domain (TD) optical coherence tomography (OCT), in terms of range, resolution and focus of the spectral (SD) OCT methods and in terms of spatial resolution of wavefront sensors. A new class of OCT systems is researched, as a marriage between the TD-OCT and SD-OCT methods. The novel methods present the generality of being compatible with both TD-OCT and SD-OCT. It is envisaged that the research results will revolutionise the field of high resolution imaging and high sensitive sensing and open applications not currently possible with the present OCT, confocal microscopy or multiphoton microscopy technology. The method to be researched will allow versatile functionality in measurements, in 3D imaging of moving tissue and functional/low noise imaging by making use of angular compounding or polarisation. Novel directions are opened in the tracking of the axial position of objects (cornea or retina), automatic dispersion compensation as well as improvement in the synchronism between the coherence gate and the focus in axial scanning. Simultaneous measurements over multiple path lengths becomes feasible, with potential applications in high throughput sensing. The methods proposed open novel avenues in biosensing by amplification of tiny frequency shifts or tiny changes in the optical paths. Possible outcome are high sensitive biosensors, multiple imaging at different depths, fast and long range tracking, long axial scanning, coherence gated wavefront sensors with applications in vision sciences and microscopy, protein identification and contrast agents developments.
Summary
Revolutionary combination of principles of spectral domain and time domain coherence gating will be researched. The present proposal puts forward: (i) a novel class of optical interferometers, (ii) a novel class of wavefront sensors and (iii) combinations of imaging channels with the novel wavefront sensors. All these are driven by the needs to address the limitations in terms of speed of the time domain (TD) optical coherence tomography (OCT), in terms of range, resolution and focus of the spectral (SD) OCT methods and in terms of spatial resolution of wavefront sensors. A new class of OCT systems is researched, as a marriage between the TD-OCT and SD-OCT methods. The novel methods present the generality of being compatible with both TD-OCT and SD-OCT. It is envisaged that the research results will revolutionise the field of high resolution imaging and high sensitive sensing and open applications not currently possible with the present OCT, confocal microscopy or multiphoton microscopy technology. The method to be researched will allow versatile functionality in measurements, in 3D imaging of moving tissue and functional/low noise imaging by making use of angular compounding or polarisation. Novel directions are opened in the tracking of the axial position of objects (cornea or retina), automatic dispersion compensation as well as improvement in the synchronism between the coherence gate and the focus in axial scanning. Simultaneous measurements over multiple path lengths becomes feasible, with potential applications in high throughput sensing. The methods proposed open novel avenues in biosensing by amplification of tiny frequency shifts or tiny changes in the optical paths. Possible outcome are high sensitive biosensors, multiple imaging at different depths, fast and long range tracking, long axial scanning, coherence gated wavefront sensors with applications in vision sciences and microscopy, protein identification and contrast agents developments.
Max ERC Funding
1 999 241 €
Duration
Start date: 2010-05-01, End date: 2015-10-31
Project acronym COHESIN CONTROL
Project The mechanism by which cohesin controls gene expression
Researcher (PI) Kim Ashley Nasmyth
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Advanced Grant (AdG), LS3, ERC-2011-ADG_20110310
Summary How cells retain, lose, and regain developmental plasticity is poorly understood due to ignorance of the molecular mechanisms regulating gene expression. Each gene is regulated by a unique set of factors and as a consequence the trans-acting factors and cis-acting chromatin modification states regulating a given gene are extremely rare. Transcription is affected by events taking place many thousands of base pairs away from the start, a property enabling developmental and evolutionary plasticity, presumably made possible by DNA looping or translocation of factors along chromatin. Most factors regulating a given gene function at many other genes, complicating interpretation of the consequences of altering the activity of such factors. It is difficult to exclude the possibility that phenotypes are knock-on effects. This could be surmounted if it were possible to observe individual genes in real time in three-dimensional space and to analyse the immediate consequences of altering the activity of regulatory factors. Of these, those capable of inter-connecting DNAs or of translocating large distances along chromatin are of interest. Cohesin is such a factor, composed of three core subunits, a pair of Smc proteins and a kleisin subunit, that interact with each other to form a huge tripartite ring, within which it is thought chromatin fibres are entrapped. In proliferating cells, cohesin’s primary function is to connect sister chromatids during DNA replication until the onset of anaphase, possibly by virtue of co-entrapment within a single ring. However, cohesin is present in most quiescent cells and it is becoming clear that it also regulates gene expression and recombination. This proposal has two goals: To image gene expression on polytene chromosomes and to investigate cohesin’s role during ecdysone-induced transcription. The advantage of this system is that we can use micro-injection of TEV protease to inactivate cohesin. A second goal is to develop the TEV system to
Summary
How cells retain, lose, and regain developmental plasticity is poorly understood due to ignorance of the molecular mechanisms regulating gene expression. Each gene is regulated by a unique set of factors and as a consequence the trans-acting factors and cis-acting chromatin modification states regulating a given gene are extremely rare. Transcription is affected by events taking place many thousands of base pairs away from the start, a property enabling developmental and evolutionary plasticity, presumably made possible by DNA looping or translocation of factors along chromatin. Most factors regulating a given gene function at many other genes, complicating interpretation of the consequences of altering the activity of such factors. It is difficult to exclude the possibility that phenotypes are knock-on effects. This could be surmounted if it were possible to observe individual genes in real time in three-dimensional space and to analyse the immediate consequences of altering the activity of regulatory factors. Of these, those capable of inter-connecting DNAs or of translocating large distances along chromatin are of interest. Cohesin is such a factor, composed of three core subunits, a pair of Smc proteins and a kleisin subunit, that interact with each other to form a huge tripartite ring, within which it is thought chromatin fibres are entrapped. In proliferating cells, cohesin’s primary function is to connect sister chromatids during DNA replication until the onset of anaphase, possibly by virtue of co-entrapment within a single ring. However, cohesin is present in most quiescent cells and it is becoming clear that it also regulates gene expression and recombination. This proposal has two goals: To image gene expression on polytene chromosomes and to investigate cohesin’s role during ecdysone-induced transcription. The advantage of this system is that we can use micro-injection of TEV protease to inactivate cohesin. A second goal is to develop the TEV system to
Max ERC Funding
2 421 212 €
Duration
Start date: 2012-05-01, End date: 2018-04-30
Project acronym COREMA
Project Cell division and the origin of embryonic aneuploidy in preimplantation mouse development
Researcher (PI) Jan ELLENBERG
Host Institution (HI) EUROPEAN MOLECULAR BIOLOGY LABORATORY
Call Details Advanced Grant (AdG), LS3, ERC-2015-AdG
Summary Cell division is fundamental for development. In the early mammalian embryo it drives the rapid proliferation of totipotent cells, the basis for forming the fetus. Given its crucial importance, it is surprising that cell division is particularly error-prone at the beginning of mammalian life, resulting in spontaneous abortion or severe developmental retardation, the incidence of which is increasing with age of the mother. Why aneuploidy is so prevalent and how early embryonic development nevertheless achieves robustness is largely unknown. The goal of this project is a comprehensive analysis of cell divisions in the mouse preimplantation embryo to determine the molecular mechanisms underlying aneuploidy and its effects on normal development. Recent technological breakthroughs, including light sheet microscopy and rapid loss-of-function approaches in the mouse embryo will allow us for the first time to tackle the molecular mechanisms of aneuploidy generation and establish the preimplantation mouse embryo as a standard cell biological model system. For that purpose we will develop next generation light sheet microscopy to enable automated chromosome tracking in the whole embryo. Mapping of cell division errors will reveal when, where, and how aneuploidy occurs, what the fate of aneuploid cells is in the embryo, and how this changes with maternal age. We will then perform high resolution functional imaging assays to identify the mitotic pathways responsible for aneuploidy and understand why they do not fully function in early development. Key proteins will be functionally characterised in detail integrating light sheet imaging with single molecule biophysics in embryos from young and aged females to achieve a mechanistic understanding of the unique aspects of cell division underlying embryonic aneuploidy. The achieved knowledge gain will have an important impact for our understanding of mammalian, including human infertility.
Summary
Cell division is fundamental for development. In the early mammalian embryo it drives the rapid proliferation of totipotent cells, the basis for forming the fetus. Given its crucial importance, it is surprising that cell division is particularly error-prone at the beginning of mammalian life, resulting in spontaneous abortion or severe developmental retardation, the incidence of which is increasing with age of the mother. Why aneuploidy is so prevalent and how early embryonic development nevertheless achieves robustness is largely unknown. The goal of this project is a comprehensive analysis of cell divisions in the mouse preimplantation embryo to determine the molecular mechanisms underlying aneuploidy and its effects on normal development. Recent technological breakthroughs, including light sheet microscopy and rapid loss-of-function approaches in the mouse embryo will allow us for the first time to tackle the molecular mechanisms of aneuploidy generation and establish the preimplantation mouse embryo as a standard cell biological model system. For that purpose we will develop next generation light sheet microscopy to enable automated chromosome tracking in the whole embryo. Mapping of cell division errors will reveal when, where, and how aneuploidy occurs, what the fate of aneuploid cells is in the embryo, and how this changes with maternal age. We will then perform high resolution functional imaging assays to identify the mitotic pathways responsible for aneuploidy and understand why they do not fully function in early development. Key proteins will be functionally characterised in detail integrating light sheet imaging with single molecule biophysics in embryos from young and aged females to achieve a mechanistic understanding of the unique aspects of cell division underlying embryonic aneuploidy. The achieved knowledge gain will have an important impact for our understanding of mammalian, including human infertility.
Max ERC Funding
2 497 156 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym CrUCCial
Project Novel diagnostic and therapeutic approach to inflammatory bowel disease based on functional characterization of patients: the CrUCCial index
Researcher (PI) Severine VERMEIRE
Host Institution (HI) KATHOLIEKE UNIVERSITEIT LEUVEN
Call Details Advanced Grant (AdG), LS7, ERC-2015-AdG
Summary The clinical phenotype and the outcome of Crohn's disease (CD) and ulcerative colitis (UC), the opposite ends of chronic inflammatory bowel diseases (IBD), are heterogeneous and represent the result of a complex interplay of the gut microbiome with the immune system in genetically predisposed individuals. Disease management is much less heterogeneous as all patients are treated using non-specific anti-inflammatory agents, and only 30-50% achieve clinical and mucosal remission -the goal of therapy nowadays- therefore leaving large margins for improvement. The advances in knowledge about the factors triggering disease onset should be translated to approach the disease from a molecular angle. Key cellular pathways have emerged including bacterial recognition, autophagy, endoplasmic reticulum stress and intestinal barrier function. Functional/molecular characterization of these pathways in a given patient, correlation with meaningful clinical outcomes, and tailoring an individual therapeutic approach has never been attempted and will represent a breakthrough in the current paradigm of treating multifactorial inflammatory conditions. This project aims to functionally characterize patients with CD/UC for the major pathways by using integrated (epi)genetic, transcriptomic, immunologic, barrier integrity and metagenomic studies. From these readouts we will construct an index [the Crohn’s and Ulcerative Colitis Characterization and Intervention trial (CrUCCial) index], reflecting the proportional contribution of each of the pathogenic mechanisms in a given patient. We will next study the correlation of this index and its components to meaningful clinical outcomes and finally, the index will be tested in a pilot study of newly diagnosed patients in whom the disease will be targeted individually based on the components of the CrUCCial index. Our approach, from diagnosis over prognosis to therapy, will revolutionize the paradigm of disease management.
Summary
The clinical phenotype and the outcome of Crohn's disease (CD) and ulcerative colitis (UC), the opposite ends of chronic inflammatory bowel diseases (IBD), are heterogeneous and represent the result of a complex interplay of the gut microbiome with the immune system in genetically predisposed individuals. Disease management is much less heterogeneous as all patients are treated using non-specific anti-inflammatory agents, and only 30-50% achieve clinical and mucosal remission -the goal of therapy nowadays- therefore leaving large margins for improvement. The advances in knowledge about the factors triggering disease onset should be translated to approach the disease from a molecular angle. Key cellular pathways have emerged including bacterial recognition, autophagy, endoplasmic reticulum stress and intestinal barrier function. Functional/molecular characterization of these pathways in a given patient, correlation with meaningful clinical outcomes, and tailoring an individual therapeutic approach has never been attempted and will represent a breakthrough in the current paradigm of treating multifactorial inflammatory conditions. This project aims to functionally characterize patients with CD/UC for the major pathways by using integrated (epi)genetic, transcriptomic, immunologic, barrier integrity and metagenomic studies. From these readouts we will construct an index [the Crohn’s and Ulcerative Colitis Characterization and Intervention trial (CrUCCial) index], reflecting the proportional contribution of each of the pathogenic mechanisms in a given patient. We will next study the correlation of this index and its components to meaningful clinical outcomes and finally, the index will be tested in a pilot study of newly diagnosed patients in whom the disease will be targeted individually based on the components of the CrUCCial index. Our approach, from diagnosis over prognosis to therapy, will revolutionize the paradigm of disease management.
Max ERC Funding
2 494 500 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym CYCLODE
Project Cyclical and Linear Timing Modes in Development
Researcher (PI) Helge GROSSHANS
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Call Details Advanced Grant (AdG), LS3, ERC-2016-ADG
Summary Organismal development requires proper timing of events such as cell fate choices, but the mechanisms that control temporal patterning remain poorly understood. In particular, we know little of the cyclical timers, or ‘clocks’, that control recurring events such as vertebrate segmentation or nematode molting. Furthermore, it is unknown how cyclical timers are coordinated with the global, or linear, timing of development, e.g. to ensure an appropriate number of cyclical repeats. We propose to elucidate the components, wiring, and properties of a prototypic developmental clock by studying developmental timing in the roundworm C. elegans. We build on our recent discovery that nearly 20% of the worm’s transcriptome oscillates during larval development – an apparent manifestation of a clock that times the various recurring events that encompass each larval stage. Our aims are i) to identify components of this clock using genetic screens, ii) to gain insight into the system’s architecture and properties by employing specific perturbations such as food deprivation, and iii) to understand the coupling of this cyclic clock to the linear heterochronic timer through genetic manipulations. To achieve our ambitious goals, we will develop tools for mRNA sequencing of individual worms and for their developmental tracking and microchamber-based imaging. These important advances will increase temporal resolution, enhance signal-to-noise ratio, and achieve live tracking of oscillations in vivo. Our combination of genetic, genomic, imaging, and computational approaches will provide a detailed understanding of this clock, and biological timing mechanisms in general. As heterochronic genes and rhythmic gene expression are also important for controlling stem cell fates, we foresee that the results gained will additionally reveal regulatory mechanisms of stem cells, thus advancing our fundamental understanding of animal development and future applications in regenerative medicine.
Summary
Organismal development requires proper timing of events such as cell fate choices, but the mechanisms that control temporal patterning remain poorly understood. In particular, we know little of the cyclical timers, or ‘clocks’, that control recurring events such as vertebrate segmentation or nematode molting. Furthermore, it is unknown how cyclical timers are coordinated with the global, or linear, timing of development, e.g. to ensure an appropriate number of cyclical repeats. We propose to elucidate the components, wiring, and properties of a prototypic developmental clock by studying developmental timing in the roundworm C. elegans. We build on our recent discovery that nearly 20% of the worm’s transcriptome oscillates during larval development – an apparent manifestation of a clock that times the various recurring events that encompass each larval stage. Our aims are i) to identify components of this clock using genetic screens, ii) to gain insight into the system’s architecture and properties by employing specific perturbations such as food deprivation, and iii) to understand the coupling of this cyclic clock to the linear heterochronic timer through genetic manipulations. To achieve our ambitious goals, we will develop tools for mRNA sequencing of individual worms and for their developmental tracking and microchamber-based imaging. These important advances will increase temporal resolution, enhance signal-to-noise ratio, and achieve live tracking of oscillations in vivo. Our combination of genetic, genomic, imaging, and computational approaches will provide a detailed understanding of this clock, and biological timing mechanisms in general. As heterochronic genes and rhythmic gene expression are also important for controlling stem cell fates, we foresee that the results gained will additionally reveal regulatory mechanisms of stem cells, thus advancing our fundamental understanding of animal development and future applications in regenerative medicine.
Max ERC Funding
2 358 625 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym CYTRIX
Project Engineering Cytokines for Super-Affinity Binding to Matrix in Regenerative Medicine
Researcher (PI) Jeffrey Alan Hubbell
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary In physiological situations, the extracellular matrix (ECM) sequesters cytokines, localizes them, and modulates their signaling. Thus, physiological signaling from cytokines occurs primarily when the cytokines are interacting with the ECM. In therapeutic use of cytokines, however, this interaction and balance have not been respected; rather the growth factors are merely injected or applied as soluble molecules, perhaps in controlled release forms. This has led to modest efficacy and substantial concerns on safety. Here, we will develop a protein engineering design for second-generation cytokines to lead to their super-affinity binding to ECM molecules in the targeted tissues; this would allow application to a tissue site to yield a tight association with ECM molecules there, turning the tissue itself into a reservoir for cytokine sequestration and presentation. To accomplish this, we have undertaken preliminary work screening a library of cytokines for extraordinarily high affinity binding to a library of ECM molecules. We have thereby identified a small peptide domain within placental growth factor-2 (PlGF-2), namely PlGF-2123-144, that displays super-affinity for a number of ECM proteins. Also in preliminary work, we have demonstrated that recombinant fusion of this domain to low-affinity binding cytokines, namely VEGF-A, PDGF-BB and BMP-2, confers super-affinity binding to ECM molecules and accentuates their functionality in vivo in regenerative medicine models. In the proposed project, based on this preliminary data, we will push forward this protein engineering design, pursuing super-affinity variants of VEGF-A and PDGF-BB in chronic wounds, TGF-beta3 and CXCL11 in skin scar reduction, FGF-18 in osteoarthritic cartilage repair and CXCL12 in stem cell recruitment to ischemic cardiac muscle. Thus, we seek to demonstrate a fundamentally new concept and platform for second-generation growth factor protein engineering.
Summary
In physiological situations, the extracellular matrix (ECM) sequesters cytokines, localizes them, and modulates their signaling. Thus, physiological signaling from cytokines occurs primarily when the cytokines are interacting with the ECM. In therapeutic use of cytokines, however, this interaction and balance have not been respected; rather the growth factors are merely injected or applied as soluble molecules, perhaps in controlled release forms. This has led to modest efficacy and substantial concerns on safety. Here, we will develop a protein engineering design for second-generation cytokines to lead to their super-affinity binding to ECM molecules in the targeted tissues; this would allow application to a tissue site to yield a tight association with ECM molecules there, turning the tissue itself into a reservoir for cytokine sequestration and presentation. To accomplish this, we have undertaken preliminary work screening a library of cytokines for extraordinarily high affinity binding to a library of ECM molecules. We have thereby identified a small peptide domain within placental growth factor-2 (PlGF-2), namely PlGF-2123-144, that displays super-affinity for a number of ECM proteins. Also in preliminary work, we have demonstrated that recombinant fusion of this domain to low-affinity binding cytokines, namely VEGF-A, PDGF-BB and BMP-2, confers super-affinity binding to ECM molecules and accentuates their functionality in vivo in regenerative medicine models. In the proposed project, based on this preliminary data, we will push forward this protein engineering design, pursuing super-affinity variants of VEGF-A and PDGF-BB in chronic wounds, TGF-beta3 and CXCL11 in skin scar reduction, FGF-18 in osteoarthritic cartilage repair and CXCL12 in stem cell recruitment to ischemic cardiac muscle. Thus, we seek to demonstrate a fundamentally new concept and platform for second-generation growth factor protein engineering.
Max ERC Funding
2 368 170 €
Duration
Start date: 2014-05-01, End date: 2019-04-30
Project acronym DanioPattern
Project Development and Evolution of Colour Patterns in Danio species
Researcher (PI) Christiane NÜSSLEIN-VOLHARD
Host Institution (HI) MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN EV
Call Details Advanced Grant (AdG), LS3, ERC-2015-AdG
Summary Colour patterns are prominent features of many animals and have important functions in communication such as camouflage, kin recognition and mate selection. Colour patterns are highly variable and evolve rapidly leading to large diversities even within a single genus. As targets for natural as well as sexual selection, they are of high evolutionary significance. The zebrafish (Danio rerio), a vertebrate model organism for the study of development and disease, displays a conspicuous pattern of alternating blue and golden stripes on the body and on the anal- and tailfins. Mutants with spectacularly altered patterns have been analysed, and novel approaches in lineage tracing have provided first insights into the cellular and molecular basis of colour patterning. These studies revealed that the mechanisms at play are novel and of fundamental interest to the biology of pattern formation. Closely related Danio species have very divergent colour patterns in body and fins offering the unique opportunity to study development and evolution of colour patterns in vertebrates building on the thorough analysis of one model species. Our research in zebrafish will explore the basis of direct interactions between chromatophores mediated by channels and junctions. We will investigate the divergent mode of stripe formation in the fins and the molecular influence of the cellular environment on chromatophore interactions. In closely related Danio species, we will investigate the cellular interactions during pattern formation. We will analyse transcriptomes and genome sequences to identify candidate genes providing the molecular basis for pigment pattern diversity. These candidate genes will be tested by creating mutants and exchanging allelic variants using the CRISPR/Cas9 system. The work will lay the foundation to understand not only the genetic basis of variation in colour pattern formation between Danio species, but also the evolution of biodiversity in other vertebrates.
Summary
Colour patterns are prominent features of many animals and have important functions in communication such as camouflage, kin recognition and mate selection. Colour patterns are highly variable and evolve rapidly leading to large diversities even within a single genus. As targets for natural as well as sexual selection, they are of high evolutionary significance. The zebrafish (Danio rerio), a vertebrate model organism for the study of development and disease, displays a conspicuous pattern of alternating blue and golden stripes on the body and on the anal- and tailfins. Mutants with spectacularly altered patterns have been analysed, and novel approaches in lineage tracing have provided first insights into the cellular and molecular basis of colour patterning. These studies revealed that the mechanisms at play are novel and of fundamental interest to the biology of pattern formation. Closely related Danio species have very divergent colour patterns in body and fins offering the unique opportunity to study development and evolution of colour patterns in vertebrates building on the thorough analysis of one model species. Our research in zebrafish will explore the basis of direct interactions between chromatophores mediated by channels and junctions. We will investigate the divergent mode of stripe formation in the fins and the molecular influence of the cellular environment on chromatophore interactions. In closely related Danio species, we will investigate the cellular interactions during pattern formation. We will analyse transcriptomes and genome sequences to identify candidate genes providing the molecular basis for pigment pattern diversity. These candidate genes will be tested by creating mutants and exchanging allelic variants using the CRISPR/Cas9 system. The work will lay the foundation to understand not only the genetic basis of variation in colour pattern formation between Danio species, but also the evolution of biodiversity in other vertebrates.
Max ERC Funding
2 250 000 €
Duration
Start date: 2016-11-01, End date: 2021-04-30
Project acronym DEATHSWITCHING
Project Identifying genes and pathways that drive molecular switches and back-up mechanisms between apoptosis and autophagy
Researcher (PI) Adi Kimchi
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS3, ERC-2012-ADG_20120314
Summary A cell’s decision to die is governed by multiple input signals received from a complex network of programmed cell death (PCD) pathways, including apoptosis and programmed necrosis. Additionally, under some conditions, autophagy, whose function is mainly pro-survival, may act as a back-up death pathway. We propose to apply new approaches to study the molecular basis of two important questions that await resolution in the field: a) how the cell switches from a pro-survival autophagic response to an apoptotic response and b) whether and how pro-survival autophagy is converted to a death mechanism when apoptosis is blocked. To address the first issue, we will screen for direct physical interactions between autophagic and apoptotic proteins, using the protein fragment complementation assay. Validated pairs will be studied in depth to identify built-in molecular switches that activate apoptosis when autophagy fails to restore homeostasis. As a pilot case to address the concept of molecular ‘sensors’ and ‘switches’, we will focus on the previously identified Atg12/Bcl-2 interaction. In the second line of research we will categorize autophagy-dependent cell death triggers into those that directly result from autophagy-dependent degradation, either by excessive self-digestion or by selective protein degradation, and those that utilize the autophagy machinery to activate programmed necrosis. We will identify the genes regulating these scenarios by whole genome RNAi screens for increased cell survival. In parallel, we will use a cell library of annotated fluorescent-tagged proteins for measuring selective protein degradation. These will be the starting point for identification of the molecular pathways that convert survival autophagy to a death program. Finally, we will explore the physiological relevance of back-up death mechanisms and the newly identified molecular mechanisms to developmental PCD during the cavitation process in early stages of embryogenesis.
Summary
A cell’s decision to die is governed by multiple input signals received from a complex network of programmed cell death (PCD) pathways, including apoptosis and programmed necrosis. Additionally, under some conditions, autophagy, whose function is mainly pro-survival, may act as a back-up death pathway. We propose to apply new approaches to study the molecular basis of two important questions that await resolution in the field: a) how the cell switches from a pro-survival autophagic response to an apoptotic response and b) whether and how pro-survival autophagy is converted to a death mechanism when apoptosis is blocked. To address the first issue, we will screen for direct physical interactions between autophagic and apoptotic proteins, using the protein fragment complementation assay. Validated pairs will be studied in depth to identify built-in molecular switches that activate apoptosis when autophagy fails to restore homeostasis. As a pilot case to address the concept of molecular ‘sensors’ and ‘switches’, we will focus on the previously identified Atg12/Bcl-2 interaction. In the second line of research we will categorize autophagy-dependent cell death triggers into those that directly result from autophagy-dependent degradation, either by excessive self-digestion or by selective protein degradation, and those that utilize the autophagy machinery to activate programmed necrosis. We will identify the genes regulating these scenarios by whole genome RNAi screens for increased cell survival. In parallel, we will use a cell library of annotated fluorescent-tagged proteins for measuring selective protein degradation. These will be the starting point for identification of the molecular pathways that convert survival autophagy to a death program. Finally, we will explore the physiological relevance of back-up death mechanisms and the newly identified molecular mechanisms to developmental PCD during the cavitation process in early stages of embryogenesis.
Max ERC Funding
2 500 000 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym DENOVO-P
Project De novo Development of Polarity in Plant Cells
Researcher (PI) Liam DOLAN
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Advanced Grant (AdG), LS3, ERC-2017-ADG
Summary The polarity of the single cell from which many organisms develop determines the polarity of the body axis. However, the polarity of these single cells is often inherited. For example, zygote polarity is inherited from the polarized egg cell of Arabidopsis thaliana. By contrast, polarity is not pre-set in the spore cell that forms the Marchantia polymorpha (Marchantia) plant. An environmental cue – directional light – polarises the spore cell which, in turn, directs the formation of the first (apical-basal) axis and the fates of the two daughter cells formed when the spore cell divides. Using Marchantia, we will discover how cell polarity is established de novo in the developing spore cell and how this, in turn, directs the specification of the first major axis in the plant.
The proposed research is feasible because of the unique characteristics of the Marchantia system:
1. Isolated single apolar cells become polarized allowing us to exploit the real-time imaging with experimental manipulation of polarising cues at each stage of development.
2. Haploid genetics can be exploited to carry out genetic screens of unprecedented depth and we can identify mutant genes using a fully annotated genome sequence.
3. Gene expression can be measured with high temporal resolution during polarization.
We propose to:
1. Describe the cellular and morphogenetic events that occur as the spore cell polarizes, divides asymmetrically to form cells at either end of the apical-basal axis.
2. Define the mechanism underpinning the de novo establishment of polarity using a combination of forward and reverse genetics and determine if this mechanism is conserved among land plants.
3. Determine the role of auxin in transmitting spore cell polarity to the cells at both ends of the apical-basal axis.
This will describe, for the first time, the molecular mechanism controlling the de novo polarization of a single cell that develops into a plant.
Summary
The polarity of the single cell from which many organisms develop determines the polarity of the body axis. However, the polarity of these single cells is often inherited. For example, zygote polarity is inherited from the polarized egg cell of Arabidopsis thaliana. By contrast, polarity is not pre-set in the spore cell that forms the Marchantia polymorpha (Marchantia) plant. An environmental cue – directional light – polarises the spore cell which, in turn, directs the formation of the first (apical-basal) axis and the fates of the two daughter cells formed when the spore cell divides. Using Marchantia, we will discover how cell polarity is established de novo in the developing spore cell and how this, in turn, directs the specification of the first major axis in the plant.
The proposed research is feasible because of the unique characteristics of the Marchantia system:
1. Isolated single apolar cells become polarized allowing us to exploit the real-time imaging with experimental manipulation of polarising cues at each stage of development.
2. Haploid genetics can be exploited to carry out genetic screens of unprecedented depth and we can identify mutant genes using a fully annotated genome sequence.
3. Gene expression can be measured with high temporal resolution during polarization.
We propose to:
1. Describe the cellular and morphogenetic events that occur as the spore cell polarizes, divides asymmetrically to form cells at either end of the apical-basal axis.
2. Define the mechanism underpinning the de novo establishment of polarity using a combination of forward and reverse genetics and determine if this mechanism is conserved among land plants.
3. Determine the role of auxin in transmitting spore cell polarity to the cells at both ends of the apical-basal axis.
This will describe, for the first time, the molecular mechanism controlling the de novo polarization of a single cell that develops into a plant.
Max ERC Funding
2 499 224 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym DENOVOHSC
Project Blood regeneration: de novo development of human hematopoietic stem cells
Researcher (PI) Elaine Anne Dzierzak
Host Institution (HI) THE UNIVERSITY OF EDINBURGH
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary Hematopoietic stem cells (HSC) are used in clinical therapies for leukemia and blood-related genetic disorders. Whereas
the number of patients requiring treatment continues to increase, HSC transplantations are limited due to insufficient
patient-matched donor HSCs. The current challenge is to create more matched HSCs. As evidenced by the Nobel Prize
award this year, reprogramming of somatic cells to pluripotent stem cells (iPS) is one of the most important breakthroughs
of recent times. This innovative advance contributes to our ability to reprogram patient-specific cells not only to pluripotency, but also to directly program them to other desired cell lineages. The study of healthy and diseased patient cells in this context will have huge impact on the development of new drug and cell-based treatments. My research is uniquely positioned at the interface of fundamental and translational research at the University of Edinburgh Centre for Inflammation Research and Centre for Regenerative Medicine. Through more than a decade of HSC developmental research, my group has shown that HSCs arise from endothelial cells in a natural reprogramming event. We are one of the few groups worldwide that can isolate these special endothelial cells and show that they yield robust transplantable HSCs (the gold-standard for clinically relevant HSCs). Using our unique expertise I aim to foster new translational strategies to de novo generate human HSCs from patient somatic cells. My aims are to 1) mark and manipulate the program for HSC generation during the endothelial to HSC transition (EHT); 2) define extrinsic molecules affecting EHT and engineer novel niches; 3) reprogram human somatic cells or endothelial derived iPS cells directly to HSCs. These aims will be realized through novel multi-color reporter mouse and ES/iPS lines indicating EHT in real-time, allowing for the isolation and functional validation of de novo HSC generation. These novel models and cultures will significantly advance research and technology, to have major impact on the field.
Summary
Hematopoietic stem cells (HSC) are used in clinical therapies for leukemia and blood-related genetic disorders. Whereas
the number of patients requiring treatment continues to increase, HSC transplantations are limited due to insufficient
patient-matched donor HSCs. The current challenge is to create more matched HSCs. As evidenced by the Nobel Prize
award this year, reprogramming of somatic cells to pluripotent stem cells (iPS) is one of the most important breakthroughs
of recent times. This innovative advance contributes to our ability to reprogram patient-specific cells not only to pluripotency, but also to directly program them to other desired cell lineages. The study of healthy and diseased patient cells in this context will have huge impact on the development of new drug and cell-based treatments. My research is uniquely positioned at the interface of fundamental and translational research at the University of Edinburgh Centre for Inflammation Research and Centre for Regenerative Medicine. Through more than a decade of HSC developmental research, my group has shown that HSCs arise from endothelial cells in a natural reprogramming event. We are one of the few groups worldwide that can isolate these special endothelial cells and show that they yield robust transplantable HSCs (the gold-standard for clinically relevant HSCs). Using our unique expertise I aim to foster new translational strategies to de novo generate human HSCs from patient somatic cells. My aims are to 1) mark and manipulate the program for HSC generation during the endothelial to HSC transition (EHT); 2) define extrinsic molecules affecting EHT and engineer novel niches; 3) reprogram human somatic cells or endothelial derived iPS cells directly to HSCs. These aims will be realized through novel multi-color reporter mouse and ES/iPS lines indicating EHT in real-time, allowing for the isolation and functional validation of de novo HSC generation. These novel models and cultures will significantly advance research and technology, to have major impact on the field.
Max ERC Funding
2 500 000 €
Duration
Start date: 2015-01-01, End date: 2019-12-31
Project acronym DIDO
Project Innovative drugs targeting IDO molecular dynamics in autoimmunity and neoplasia
Researcher (PI) Ursula Grohmann
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PERUGIA
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary "Catabolism of amino acids is an ancient survival strategy that also controls immune responses in mammals. Indoleamine 2,3-dioxygenase (IDO), a tryptophan catabolizing enzyme, is recognized as an authentic regulator of immunity in several physiopathologic conditions, including autoimmune diseases, in which it is often defective, and neoplasia, in which it promotes immune unresponsiveness. The PI’s group recently revealed that IDO does not merely degrade tryptophan and produce immunoregulatory kynurenines but also acts as a signal-transducing molecule independently of its enzyme activity. IDO’s signaling function relies on the presence of phosphorylable motifs in a region (small IDO domain) distant from the catalytic site (large IDO domain). Preliminary data indicate that IDO, depending on microenvironmental conditions, can move among distinct cellular compartments. Thus IDO may be considered a ‘moonligthing’ protein, i.e., an ancestral metabolic molecule that, during evolution, has acquired the DYNAMIC feature of moving intracellularly and switching among distinct functions by changing its conformational state. By means of computational studies, Macchiarulo’s group (team member) has identified distinct conformations of IDO, some of which are associated with optimal catalytic activity of the enzyme whereas others may favor tyrosine phosphorylation of IDO’s small domain. A switch between distinct conformations can be induced by the use of ligands that bind either the catalytic site or an accessory pocket outside the IDO catalytic site. The first aim of DIDO is to decipher the relationships between IDO conformations and multiple functions of the enzyme. A second aim is to identify small molecules with drug-like properties capable of modulating distinct IDO’s molecular conformations in order to either potentiate (a new therapeutic approach in autoimmune diseases) or inhibit (more efficient anti-tumor therapeutic strategy) immunoregulatory signaling ability of IDO."
Summary
"Catabolism of amino acids is an ancient survival strategy that also controls immune responses in mammals. Indoleamine 2,3-dioxygenase (IDO), a tryptophan catabolizing enzyme, is recognized as an authentic regulator of immunity in several physiopathologic conditions, including autoimmune diseases, in which it is often defective, and neoplasia, in which it promotes immune unresponsiveness. The PI’s group recently revealed that IDO does not merely degrade tryptophan and produce immunoregulatory kynurenines but also acts as a signal-transducing molecule independently of its enzyme activity. IDO’s signaling function relies on the presence of phosphorylable motifs in a region (small IDO domain) distant from the catalytic site (large IDO domain). Preliminary data indicate that IDO, depending on microenvironmental conditions, can move among distinct cellular compartments. Thus IDO may be considered a ‘moonligthing’ protein, i.e., an ancestral metabolic molecule that, during evolution, has acquired the DYNAMIC feature of moving intracellularly and switching among distinct functions by changing its conformational state. By means of computational studies, Macchiarulo’s group (team member) has identified distinct conformations of IDO, some of which are associated with optimal catalytic activity of the enzyme whereas others may favor tyrosine phosphorylation of IDO’s small domain. A switch between distinct conformations can be induced by the use of ligands that bind either the catalytic site or an accessory pocket outside the IDO catalytic site. The first aim of DIDO is to decipher the relationships between IDO conformations and multiple functions of the enzyme. A second aim is to identify small molecules with drug-like properties capable of modulating distinct IDO’s molecular conformations in order to either potentiate (a new therapeutic approach in autoimmune diseases) or inhibit (more efficient anti-tumor therapeutic strategy) immunoregulatory signaling ability of IDO."
Max ERC Funding
2 442 078 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym DIRNDL
Project Directions in Development
Researcher (PI) Dolf WEIJERS
Host Institution (HI) WAGENINGEN UNIVERSITY
Call Details Advanced Grant (AdG), LS3, ERC-2018-ADG
Summary Cells in multicellular organisms organise along body and tissue axes. Cellular processes, such as division plane orientation, must be aligned with these polarity axes to generate functional 3-dimensional morphology, particularly in plants, where cell walls prevent cell migration. While some polarly localized plant proteins are known, molecular mechanisms of polarity establishment or its translation to division orientation are elusive, in part because regulators in animals and fungi appear to be missing from plant genomes. Cell polarity is first established in the embryo, but this has long been an intractable experimental model. My team has developed the genetic, cell biological and biochemical tools that now render the early Arabidopsis embryo an exquisite model for studying cell polarity and oriented division. Recent efforts already led to the unexpected identification of a novel family of deeply conserved polar plant proteins that share a structural domain with key animal polarity regulators. In the DIRNDL project, we will capitalize upon our unique position and foundational results, and use complementary approaches to discover the plant cell polarity and division orientation system. Firstly, we will address the function of the newly identified conserved polarity proteins, and determine mechanistic convergence of polarity regulators across multicellular kingdoms. Furthermore, we will use proteomic approaches to systematically identify polar proteins, and a genetic approach to identify regulators of polarity and division orientation, essential for embryogenesis. We will functionally analyse polar proteins and regulators both in Arabidopsis and the liverwort Marchantia to help prioritize conserved components, and to facilitate genetic analysis of protein function. Finally, we will use a cell-based system for engineering polarity de novo using the regulators identified in the project, and thus reveal the mechanisms that provide direction in plant development.
Summary
Cells in multicellular organisms organise along body and tissue axes. Cellular processes, such as division plane orientation, must be aligned with these polarity axes to generate functional 3-dimensional morphology, particularly in plants, where cell walls prevent cell migration. While some polarly localized plant proteins are known, molecular mechanisms of polarity establishment or its translation to division orientation are elusive, in part because regulators in animals and fungi appear to be missing from plant genomes. Cell polarity is first established in the embryo, but this has long been an intractable experimental model. My team has developed the genetic, cell biological and biochemical tools that now render the early Arabidopsis embryo an exquisite model for studying cell polarity and oriented division. Recent efforts already led to the unexpected identification of a novel family of deeply conserved polar plant proteins that share a structural domain with key animal polarity regulators. In the DIRNDL project, we will capitalize upon our unique position and foundational results, and use complementary approaches to discover the plant cell polarity and division orientation system. Firstly, we will address the function of the newly identified conserved polarity proteins, and determine mechanistic convergence of polarity regulators across multicellular kingdoms. Furthermore, we will use proteomic approaches to systematically identify polar proteins, and a genetic approach to identify regulators of polarity and division orientation, essential for embryogenesis. We will functionally analyse polar proteins and regulators both in Arabidopsis and the liverwort Marchantia to help prioritize conserved components, and to facilitate genetic analysis of protein function. Finally, we will use a cell-based system for engineering polarity de novo using the regulators identified in the project, and thus reveal the mechanisms that provide direction in plant development.
Max ERC Funding
2 500 000 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym DNADEMETHYLASE
Project Functions and mechanism of active DNA demethylation
Researcher (PI) Heinz Christof Niehrs
Host Institution (HI) INSTITUT FUR MOLEKULARE BIOLOGIE GGMBH
Call Details Advanced Grant (AdG), LS3, ERC-2009-AdG
Summary Epigenetic gene regulation is of central importance for development and disease. Despite dramatic progress in epigenetics during the past decade, DNA demethylation remains one of the last big frontiers and very little is known about it. DNA demethylation is a widespread phenomenon and occurs in plants as well as in animals, during development, in the adult, and during somatic cell reprogramming of pluripotency genes. The molecular identity of the DNA demethylase in animal cells remained unresolved and has hampered progress in the field for decades. In 2007 we published that Growth Arrest and DNA Damage 45 a (Gadd45a) is a key player in active DNA demethylation, which opened new avenues in the study of this elusive process. The goal of this project is to further analyze the mechanism of DNA demethylation as well as the role played by Gadd45 in development. Given the many unresolved questions in this burgeoning field, our work promises to be ground-breaking and therefore have a profound impact in unraveling one of the least understood processes of gene regulation. Specifically we will address the following points. I) The biological role of Gadd45 mediated DNA demethylation in mouse embryos and adults is unknown. We have obtained mouse mutants for Gadd45a,b, and g and we will analyze them for developmental defects and dissect the methylation regulation of relevant genes. II) The targeting mechanism by which Gadd45 is binding to and demethylating specific sites in the genome is a central unresolved issue. We have identified a candidate DNA binding protein interacting with Gadd45 and we will analyze its role in site specific targeting of DNA demethylation in vitro and in mouse. III) We found that Gadd45 is an RNA binding protein and we will therefore analyze how non-coding RNAs are involved in targeting and/or activating Gadd45 during DNA demethylation.
Summary
Epigenetic gene regulation is of central importance for development and disease. Despite dramatic progress in epigenetics during the past decade, DNA demethylation remains one of the last big frontiers and very little is known about it. DNA demethylation is a widespread phenomenon and occurs in plants as well as in animals, during development, in the adult, and during somatic cell reprogramming of pluripotency genes. The molecular identity of the DNA demethylase in animal cells remained unresolved and has hampered progress in the field for decades. In 2007 we published that Growth Arrest and DNA Damage 45 a (Gadd45a) is a key player in active DNA demethylation, which opened new avenues in the study of this elusive process. The goal of this project is to further analyze the mechanism of DNA demethylation as well as the role played by Gadd45 in development. Given the many unresolved questions in this burgeoning field, our work promises to be ground-breaking and therefore have a profound impact in unraveling one of the least understood processes of gene regulation. Specifically we will address the following points. I) The biological role of Gadd45 mediated DNA demethylation in mouse embryos and adults is unknown. We have obtained mouse mutants for Gadd45a,b, and g and we will analyze them for developmental defects and dissect the methylation regulation of relevant genes. II) The targeting mechanism by which Gadd45 is binding to and demethylating specific sites in the genome is a central unresolved issue. We have identified a candidate DNA binding protein interacting with Gadd45 and we will analyze its role in site specific targeting of DNA demethylation in vitro and in mouse. III) We found that Gadd45 is an RNA binding protein and we will therefore analyze how non-coding RNAs are involved in targeting and/or activating Gadd45 during DNA demethylation.
Max ERC Funding
2 376 000 €
Duration
Start date: 2010-06-01, End date: 2015-05-31
Project acronym DROSOPHILASIGNALING
Project Signaling Pathways Controlling Patterning, Growth and Final Size of Drosophila Limbs
Researcher (PI) Konrad Basler
Host Institution (HI) UNIVERSITAT ZURICH
Call Details Advanced Grant (AdG), LS3, ERC-2008-AdG
Summary Developmental biology seeks not only to learn more about the fundamental processes of growth and pattern per se, but to understand how they synergize to enable the morphogenesis of multicellular organisms. Our goal is to perform real-time analyses of these developmental processes in an intact developing organ. By applying a vital imaging approach, we can circumvent the normal limitations of inferring cellular dynamics from static images or molecular data, and obtain the real dynamic view of growth and patterning. The wing imaginal disc of Drosophila, which starts out as a simple epithelial structure and gives rise to a precisely structured adult limb, will serve as an ideal model system. This system has the combined advantages of relative simplicity and genetic tractability. We will create several innovations that expand the current toolkit and thus facilitate the detailed dissection of growth and patterning. A key early step will be to develop novel reporters to dynamically and faithfully monitor signaling cascades involved in growth and patterning, such as the Dpp and Hippo pathways. We will also implement quantification techniques that are currently being set up in collaboration with an experimental physicist, to deduce, and alter, the mechanical forces that develop in the cells of a growing tissue. The large amount of quantitative data that will be generated allow us derive computational models of the individual pathways and their interaction. The focus of the study will be to answer the following questions: 1) Is the Hippo pathway regulated spatially and temporally, and by what signaling pathways? 2) Do mechanical forces play a pivotal controlling role in organ morphogenesis? 3) What are the global effects on growth, when pathways controlling patterning, cell competition or compensatory proliferation are perturbed? The proposed project will bring the approaches taken to define the mechanisms underlying and controlling growth and patterning to the next level.
Summary
Developmental biology seeks not only to learn more about the fundamental processes of growth and pattern per se, but to understand how they synergize to enable the morphogenesis of multicellular organisms. Our goal is to perform real-time analyses of these developmental processes in an intact developing organ. By applying a vital imaging approach, we can circumvent the normal limitations of inferring cellular dynamics from static images or molecular data, and obtain the real dynamic view of growth and patterning. The wing imaginal disc of Drosophila, which starts out as a simple epithelial structure and gives rise to a precisely structured adult limb, will serve as an ideal model system. This system has the combined advantages of relative simplicity and genetic tractability. We will create several innovations that expand the current toolkit and thus facilitate the detailed dissection of growth and patterning. A key early step will be to develop novel reporters to dynamically and faithfully monitor signaling cascades involved in growth and patterning, such as the Dpp and Hippo pathways. We will also implement quantification techniques that are currently being set up in collaboration with an experimental physicist, to deduce, and alter, the mechanical forces that develop in the cells of a growing tissue. The large amount of quantitative data that will be generated allow us derive computational models of the individual pathways and their interaction. The focus of the study will be to answer the following questions: 1) Is the Hippo pathway regulated spatially and temporally, and by what signaling pathways? 2) Do mechanical forces play a pivotal controlling role in organ morphogenesis? 3) What are the global effects on growth, when pathways controlling patterning, cell competition or compensatory proliferation are perturbed? The proposed project will bring the approaches taken to define the mechanisms underlying and controlling growth and patterning to the next level.
Max ERC Funding
2 310 000 €
Duration
Start date: 2009-02-01, End date: 2014-01-31
Project acronym DYNASTEM
Project Dynamic, stem cell-mediated self-renewal in the Drosophila intestine
Researcher (PI) Bruce Alexander Edgar
Host Institution (HI) RUPRECHT-KARLS-UNIVERSITAET HEIDELBERG
Call Details Advanced Grant (AdG), LS3, ERC-2010-AdG_20100317
Summary Cells in intestinal epithelia turn over rapidly due to aging, damage, and toxins produced by the enteric microbiota. Gut homeostasis is maintained by intestinal stem cells (ISCs) that divide to renew the intestinal epithelium, but little is known about how ISC division and differentiation are coordinated with the loss of spent gut epithelial cells. This proposal addresses the mechanisms of dynamic self-renewal in the intestine of Drosophila. Our recent work has outlined a paradigm explaining intestinal homeostasis in Drosophila that could apply also to humans. A new lab is being established in Heidelberg where we wish to extend these studies. Our objectives are to understand: 1) How intestinal stem cell pool sizes are regulated; 2) How the cytokines and growth factors that mediate gut homeostasis are controlled; and 3) How these signals regulate the ISC cell cycle. Established genetic and cell biological methods will be applied, supported by molecular assays (microarrays, qPCR, ChIP/Seq) of gene control. New pathways of ISC control will be discovered via comprehensive genetic screens using transgenic RNAi and gene over-expression. In vitro culture of ISCs will be developed and used for live imaging and molecular analysis of the mechanisms controlling ISC proliferation and differentiation. These studies should elaborate a paradigm explaining intestinal homeostasis in flies that can guide studies in mammals, eventually contributing to the diagnosis and treatment for diseases in which gut homeostasis is disrupted, such as colorectal cancer and inflammatory bowel disease. Because stem cell biology is so highly relevant to wound healing, regeneration, cancer, aging and degenerative disease, this research could impact human health at many levels.
Summary
Cells in intestinal epithelia turn over rapidly due to aging, damage, and toxins produced by the enteric microbiota. Gut homeostasis is maintained by intestinal stem cells (ISCs) that divide to renew the intestinal epithelium, but little is known about how ISC division and differentiation are coordinated with the loss of spent gut epithelial cells. This proposal addresses the mechanisms of dynamic self-renewal in the intestine of Drosophila. Our recent work has outlined a paradigm explaining intestinal homeostasis in Drosophila that could apply also to humans. A new lab is being established in Heidelberg where we wish to extend these studies. Our objectives are to understand: 1) How intestinal stem cell pool sizes are regulated; 2) How the cytokines and growth factors that mediate gut homeostasis are controlled; and 3) How these signals regulate the ISC cell cycle. Established genetic and cell biological methods will be applied, supported by molecular assays (microarrays, qPCR, ChIP/Seq) of gene control. New pathways of ISC control will be discovered via comprehensive genetic screens using transgenic RNAi and gene over-expression. In vitro culture of ISCs will be developed and used for live imaging and molecular analysis of the mechanisms controlling ISC proliferation and differentiation. These studies should elaborate a paradigm explaining intestinal homeostasis in flies that can guide studies in mammals, eventually contributing to the diagnosis and treatment for diseases in which gut homeostasis is disrupted, such as colorectal cancer and inflammatory bowel disease. Because stem cell biology is so highly relevant to wound healing, regeneration, cancer, aging and degenerative disease, this research could impact human health at many levels.
Max ERC Funding
2 682 080 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym EDIP
Project Evolution of Development In Plants
Researcher (PI) Jane Alison Langdale
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Advanced Grant (AdG), LS3, ERC-2008-AdG
Summary Different morphologies evolve in different organisms in response to changing environments. As land plants evolved, developmental mechanisms were either generated de novo, or were recruited from existing toolkits and adapted to facilitate changes in form. Some of these changes occurred once, others on multiple occasions, and others were gained and then subsequently lost in a subset of lineages. Why have certain forms survived and others not? Why does a fern look different from a flowering plant, and why should developmental biologists care? By determining how many different ways there are to generate a particular morphology, we gain an understanding of whether a particular transition is constrained. This basic information allows an assessment of the extent to which genetic variation can modify developmental mechanisms and an indication of the degree of developmental plasticity that is possible and/or tolerated both within and between species. This proposal aims to characterize the developmental mechanisms that underpin the diverse shoot forms seen in extant plant species. The main goal is to compare developmental mechanisms that operate in vegetative shoots of bryophytes, lycophytes, ferns and angiosperms, with a view to understanding the constraints that limit morphological variation. Specifically, we will investigate the developmental basis of three major innovations that altered the morphology of vegetative shoots during land plant evolution: 1) formation of a multi-cellular embryo; 2) organization of apical growth centres and 3) patterning of leaves in distinct spatial arrangements along the shoot. To facilitate progress we also aim to develop transgenic methods, create mutant populations and generate digital transcriptomes for model species at key phylogenetic nodes. The proposed work will generate scenarios to explain how land plant form evolved and perhaps more importantly, how it could change in the future.
Summary
Different morphologies evolve in different organisms in response to changing environments. As land plants evolved, developmental mechanisms were either generated de novo, or were recruited from existing toolkits and adapted to facilitate changes in form. Some of these changes occurred once, others on multiple occasions, and others were gained and then subsequently lost in a subset of lineages. Why have certain forms survived and others not? Why does a fern look different from a flowering plant, and why should developmental biologists care? By determining how many different ways there are to generate a particular morphology, we gain an understanding of whether a particular transition is constrained. This basic information allows an assessment of the extent to which genetic variation can modify developmental mechanisms and an indication of the degree of developmental plasticity that is possible and/or tolerated both within and between species. This proposal aims to characterize the developmental mechanisms that underpin the diverse shoot forms seen in extant plant species. The main goal is to compare developmental mechanisms that operate in vegetative shoots of bryophytes, lycophytes, ferns and angiosperms, with a view to understanding the constraints that limit morphological variation. Specifically, we will investigate the developmental basis of three major innovations that altered the morphology of vegetative shoots during land plant evolution: 1) formation of a multi-cellular embryo; 2) organization of apical growth centres and 3) patterning of leaves in distinct spatial arrangements along the shoot. To facilitate progress we also aim to develop transgenic methods, create mutant populations and generate digital transcriptomes for model species at key phylogenetic nodes. The proposed work will generate scenarios to explain how land plant form evolved and perhaps more importantly, how it could change in the future.
Max ERC Funding
2 230 732 €
Duration
Start date: 2009-07-01, End date: 2015-06-30
Project acronym ELEGANSFUSION
Project Mechanisms of cell fusion in eukaryotes
Researcher (PI) Benjamin Podbilewicz
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Advanced Grant (AdG), LS3, ERC-2010-AdG_20100317
Summary Membrane fusion is a universal process essential inside cells (endoplasmic) and between cells in fertilization and organ formation (exoplasmic). With the exception of SNARE-mediated endoplasmic fusion the proteins that mediate cellular fusion (fusogens) are unknown. Despite many years of research, little is known about the mechanism of cell-cell fusion. Our studies of developmental cell fusion in the nematode C. elegans have led to the discovery of the first family of eukaryotic fusogens (FF). These fusogens, EFF-1 and AFF-1, are type I membrane glycoproteins that are essential for cell fusion and can fuse cells when ectopically expressed on the membranes of C. elegans and heterologous cells.
Our main goals are:
(1) To determine the physicochemical mechanism of cell membrane fusion mediated by FF proteins.
(2) To find the missing fusogens that act in cell fusion events across all kingdoms of life.
We hypothesize that FF proteins fuse membranes by a mechanism analogous to viral or endoplasmic fusogens and that unidentified fusogens fuse cells following the same principles as FF proteins.
Our specific aims are:
AIM 1 Determine the mechanism of FF-mediated cell fusion: A paradigm for cell membrane fusion
AIM 2 Find the sperm-egg fusion proteins (fusogens) in C. elegans
AIM 3 Identify the myoblast fusogens in mammals
AIM 4 Test fusogens using functional cell fusion assays in heterologous systems
Identifying critical domains required for FF fusion, intermediates in membrane remodeling, and atomic structures of FF proteins will advance the fundamental understanding of the mechanisms of eukaryotic cell fusion. We propose to find the Holy Grail of fertilization and mammalian myoblast fusion. We estimate that this project, if successful, will bring a breakthrough to the sperm-egg and muscle fusion fields with potential applications in basic and applied biomedical sciences.
Summary
Membrane fusion is a universal process essential inside cells (endoplasmic) and between cells in fertilization and organ formation (exoplasmic). With the exception of SNARE-mediated endoplasmic fusion the proteins that mediate cellular fusion (fusogens) are unknown. Despite many years of research, little is known about the mechanism of cell-cell fusion. Our studies of developmental cell fusion in the nematode C. elegans have led to the discovery of the first family of eukaryotic fusogens (FF). These fusogens, EFF-1 and AFF-1, are type I membrane glycoproteins that are essential for cell fusion and can fuse cells when ectopically expressed on the membranes of C. elegans and heterologous cells.
Our main goals are:
(1) To determine the physicochemical mechanism of cell membrane fusion mediated by FF proteins.
(2) To find the missing fusogens that act in cell fusion events across all kingdoms of life.
We hypothesize that FF proteins fuse membranes by a mechanism analogous to viral or endoplasmic fusogens and that unidentified fusogens fuse cells following the same principles as FF proteins.
Our specific aims are:
AIM 1 Determine the mechanism of FF-mediated cell fusion: A paradigm for cell membrane fusion
AIM 2 Find the sperm-egg fusion proteins (fusogens) in C. elegans
AIM 3 Identify the myoblast fusogens in mammals
AIM 4 Test fusogens using functional cell fusion assays in heterologous systems
Identifying critical domains required for FF fusion, intermediates in membrane remodeling, and atomic structures of FF proteins will advance the fundamental understanding of the mechanisms of eukaryotic cell fusion. We propose to find the Holy Grail of fertilization and mammalian myoblast fusion. We estimate that this project, if successful, will bring a breakthrough to the sperm-egg and muscle fusion fields with potential applications in basic and applied biomedical sciences.
Max ERC Funding
2 380 000 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym EMTASY
Project Common molecular pathways in epithelial-mesenchymal transition and left-right asymmetries
Researcher (PI) Maria Angela Nieto Toledano
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Advanced Grant (AdG), LS3, ERC-2012-ADG_20120314
Summary The majority of animals show an external bilateral symmetry, precluding the observation of multiple internal left-right (L/R) asymmetries which are fundamental for organ packaging and function. A prominent molecular pathway converging on and downstream of the Pitx2 transcription factor confers left-handed information in the left side of the embryo, with players expressed on the right ensuring that the left determinants are excluded. Therefore, conferring or excluding left identity in left and right hand sides, respectively, drives L/R asymmetry. Some indications suggest that a program actively specifying right–handed information could exist on the right. Our recent findings support this view. In a screening for novel regulators of the epithelial to mesenchymal transition (EMT), we have identified a transcription factor, EMT2, which similarly to well known factor Snail, it is an EMT inducer. The EMT is crucial for the development of tissues during embryonic development and for the progression of carcinomas to the invasive state. Strikingly, again as Snail, in addition to promote EMT, the EMT2 factor is predominantly expressed on the right side and may operate instructing L/R identity on the right-hand side of the embryo.
With this background, our knowledge of the EMT and a series of genome-wide high-throughput approaches and a comprehensive functional analysis using the chick, the fish and the mouse as model systems we propose to reveal the putative molecular pathways conveying right-handed information and to reveal commonalities between L/R pathways and the EMT. In the long run, we aim at better understanding human pathologies that involve these morphogenetic and cellular processes, including pathological situs conditions (i.e. altered organ positioning) and cancer progression.
Summary
The majority of animals show an external bilateral symmetry, precluding the observation of multiple internal left-right (L/R) asymmetries which are fundamental for organ packaging and function. A prominent molecular pathway converging on and downstream of the Pitx2 transcription factor confers left-handed information in the left side of the embryo, with players expressed on the right ensuring that the left determinants are excluded. Therefore, conferring or excluding left identity in left and right hand sides, respectively, drives L/R asymmetry. Some indications suggest that a program actively specifying right–handed information could exist on the right. Our recent findings support this view. In a screening for novel regulators of the epithelial to mesenchymal transition (EMT), we have identified a transcription factor, EMT2, which similarly to well known factor Snail, it is an EMT inducer. The EMT is crucial for the development of tissues during embryonic development and for the progression of carcinomas to the invasive state. Strikingly, again as Snail, in addition to promote EMT, the EMT2 factor is predominantly expressed on the right side and may operate instructing L/R identity on the right-hand side of the embryo.
With this background, our knowledge of the EMT and a series of genome-wide high-throughput approaches and a comprehensive functional analysis using the chick, the fish and the mouse as model systems we propose to reveal the putative molecular pathways conveying right-handed information and to reveal commonalities between L/R pathways and the EMT. In the long run, we aim at better understanding human pathologies that involve these morphogenetic and cellular processes, including pathological situs conditions (i.e. altered organ positioning) and cancer progression.
Max ERC Funding
2 460 000 €
Duration
Start date: 2013-05-01, End date: 2018-12-31
Project acronym ENCODE
Project Environmental Control of Development
Researcher (PI) Henrietta Leyser Day
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Advanced Grant (AdG), LS3, ERC-2011-ADG_20110310
Summary Plant development is highly plastic, with major variations in form determined by the environment. An excellent example is shoot branching, where the body plan of the shoot system conferred by one genotype can range from a single unbranched stem, to a highly ramified bush, depending on the growth conditions. In recent years we have investigated the hormonal network that allows environmentally sensitive changes in shoot branching in Arabidopsis. Through the analysis of a set of monogenic mutants with clear effects on both the number of shoot branches produced and on its responsiveness to environmental inputs, we have developed a model for shoot branching control involving interactions between three systemically transported plant hormones. In collaboration with Prusinkiewicz (Calgary), we have built a computational implementation of this model, which captures the phenotypes of wild-type plants and, through the manipulation of single biologically plausible model parameters, our mutant phenotypes. While there is still much to learn about individual network components, the mechanistic framework we have established is sufficiently well developed to allow network-level investigation. We therefore propose an ambitious project to use natural allelic variation in shoot branching and its environmental sensitivity as in vivo differently parameterized versions of the shoot branching regulatory network, which can be compared with parameter space exploration in our computational model. By investigating the properties of shoot branching in diverse genotypes and in the computational model parameter space, we will identify trait correlations that will contribute to understanding the architecture of the regulatory network. This approach will simultaneously test the validity of our current model and provide new hypotheses for investigation. Furthermore, the rapidly moving genomics tools available in Arabidopsis will allow us to elucidate the genetic basis for key network properties.
Summary
Plant development is highly plastic, with major variations in form determined by the environment. An excellent example is shoot branching, where the body plan of the shoot system conferred by one genotype can range from a single unbranched stem, to a highly ramified bush, depending on the growth conditions. In recent years we have investigated the hormonal network that allows environmentally sensitive changes in shoot branching in Arabidopsis. Through the analysis of a set of monogenic mutants with clear effects on both the number of shoot branches produced and on its responsiveness to environmental inputs, we have developed a model for shoot branching control involving interactions between three systemically transported plant hormones. In collaboration with Prusinkiewicz (Calgary), we have built a computational implementation of this model, which captures the phenotypes of wild-type plants and, through the manipulation of single biologically plausible model parameters, our mutant phenotypes. While there is still much to learn about individual network components, the mechanistic framework we have established is sufficiently well developed to allow network-level investigation. We therefore propose an ambitious project to use natural allelic variation in shoot branching and its environmental sensitivity as in vivo differently parameterized versions of the shoot branching regulatory network, which can be compared with parameter space exploration in our computational model. By investigating the properties of shoot branching in diverse genotypes and in the computational model parameter space, we will identify trait correlations that will contribute to understanding the architecture of the regulatory network. This approach will simultaneously test the validity of our current model and provide new hypotheses for investigation. Furthermore, the rapidly moving genomics tools available in Arabidopsis will allow us to elucidate the genetic basis for key network properties.
Max ERC Funding
2 483 870 €
Duration
Start date: 2012-01-01, End date: 2017-05-31
Project acronym EPIC
Project Enabling Precision Immuno-oncology in Colorectal cancer
Researcher (PI) Zlatko TRAJANOSKI
Host Institution (HI) MEDIZINISCHE UNIVERSITAT INNSBRUCK
Call Details Advanced Grant (AdG), LS7, ERC-2017-ADG
Summary Immunotherapy with checkpoints blockers is transforming the treatment of advanced cancers. Colorectal cancer (CRC), a cancer with 1.4 million new cases diagnosed annually worldwide, is refractory to immunotherapy (with the exception of a minority of tumors with microsatellite instability). This is somehow paradoxical as CRC is a cancer for which we have shown that it is under immunological control and that tumor infiltrating lymphocytes represent a strong independent predictor of survival. Thus, there is an urgent need to broaden the clinical benefits of immune checkpoint blockers to CRC by combining agents with synergistic mechanisms of action. An attractive approach to sensitize tumors to immunotherapy is to harness immunogenic effects induced by approved conventional or targeted agents.
Here I propose a new paradigm to identify molecular determinants of resistance to immunotherapy and develop personalized in silico and in vitro models for predicting response to combination therapy in CRC. The EPIC concept is based on three pillars: 1) emphasis on antitumor T cell activity; 2) systematic interrogation of tumor-immune cell interactions using data-driven modeling and knowledge-based mechanistic modeling, and 3) generation of key quantitative data to train and validate algorithms using perturbation experiments with patient-derived tumor organoids and cutting-edge technologies for multidimensional profiling. We will investigate three immunomodulatory processes: 1) immunostimulatory effects of chemotherapeutics, 2) rewiring of signaling networks induced by targeted drugs and their interference with immunity, and 3) metabolic reprogramming of T cells to enhance antitumor immunity.
The anticipated outcome of EPIC is a precision immuno-oncology platform that integrates tumor organoids with high-throughput and high-content data for testing drug combinations, and machine learning for making therapeutic recommendations for individual patients.
Summary
Immunotherapy with checkpoints blockers is transforming the treatment of advanced cancers. Colorectal cancer (CRC), a cancer with 1.4 million new cases diagnosed annually worldwide, is refractory to immunotherapy (with the exception of a minority of tumors with microsatellite instability). This is somehow paradoxical as CRC is a cancer for which we have shown that it is under immunological control and that tumor infiltrating lymphocytes represent a strong independent predictor of survival. Thus, there is an urgent need to broaden the clinical benefits of immune checkpoint blockers to CRC by combining agents with synergistic mechanisms of action. An attractive approach to sensitize tumors to immunotherapy is to harness immunogenic effects induced by approved conventional or targeted agents.
Here I propose a new paradigm to identify molecular determinants of resistance to immunotherapy and develop personalized in silico and in vitro models for predicting response to combination therapy in CRC. The EPIC concept is based on three pillars: 1) emphasis on antitumor T cell activity; 2) systematic interrogation of tumor-immune cell interactions using data-driven modeling and knowledge-based mechanistic modeling, and 3) generation of key quantitative data to train and validate algorithms using perturbation experiments with patient-derived tumor organoids and cutting-edge technologies for multidimensional profiling. We will investigate three immunomodulatory processes: 1) immunostimulatory effects of chemotherapeutics, 2) rewiring of signaling networks induced by targeted drugs and their interference with immunity, and 3) metabolic reprogramming of T cells to enhance antitumor immunity.
The anticipated outcome of EPIC is a precision immuno-oncology platform that integrates tumor organoids with high-throughput and high-content data for testing drug combinations, and machine learning for making therapeutic recommendations for individual patients.
Max ERC Funding
2 460 500 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym EPICHEART
Project Interplay of genetic, nutrient and lifestyle factors on incidence of coronary disease: EPIC-Heart
Researcher (PI) John Navid Danesh
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary A major new frontier in biomedicine is to identify interactions of specific components of nature and nurture in determining risk of complex diseases. Coronary heart disease (CHD) - the single leading cause of death in Europe - is jointly determined by genetic and lifestyle factors. It is unknown, however, how diet, nutrients, and lifestyle factors modulate genetic susceptibility. For scientific and public health reasons, there is a strategic need to study such interactions reliably. There is a unique opportunity for a cost-effective and powerful study because we have:
-conducted detailed assessment of diet and other lifestyle factors in 520K participants in a pan-European prospective study
-identified >12K incident CHD cases accrued during >5M person-years at risk
-identified a random sub-cohort of >15K participants without CHD to serve as referents
-commenced assay of lipid and other markers in 12K cases and 15K referents
-prepared DNA for genetic assay.
In 12K CHD cases and 15K referents, we will assay: >217K genetic variants contained in the state-of-the-art metabochip; 37 fatty acids; and vitamin D metabolites. Analyses will focus on: discovery of interactions; causal evaluation of biomarkers; CHD risk scores.
The findings should open new horizons and contribute towards: 1) explaining missing heritability 2) shaping nutritional guidelines 3) personalising preventive medicine 4) defining approaches to modify genetic susceptibility by avoiding deleterious lifestyles 5) identifying biomarkers that are priority therapeutic targets and 6) innovative prognostic tools.
Gene-lifestyle interplay is a major future growth area. This world-leading study will enhance Europes strengths in public health, nutrition and genetic epidemiology, and create a valuable future resource.
Summary
A major new frontier in biomedicine is to identify interactions of specific components of nature and nurture in determining risk of complex diseases. Coronary heart disease (CHD) - the single leading cause of death in Europe - is jointly determined by genetic and lifestyle factors. It is unknown, however, how diet, nutrients, and lifestyle factors modulate genetic susceptibility. For scientific and public health reasons, there is a strategic need to study such interactions reliably. There is a unique opportunity for a cost-effective and powerful study because we have:
-conducted detailed assessment of diet and other lifestyle factors in 520K participants in a pan-European prospective study
-identified >12K incident CHD cases accrued during >5M person-years at risk
-identified a random sub-cohort of >15K participants without CHD to serve as referents
-commenced assay of lipid and other markers in 12K cases and 15K referents
-prepared DNA for genetic assay.
In 12K CHD cases and 15K referents, we will assay: >217K genetic variants contained in the state-of-the-art metabochip; 37 fatty acids; and vitamin D metabolites. Analyses will focus on: discovery of interactions; causal evaluation of biomarkers; CHD risk scores.
The findings should open new horizons and contribute towards: 1) explaining missing heritability 2) shaping nutritional guidelines 3) personalising preventive medicine 4) defining approaches to modify genetic susceptibility by avoiding deleterious lifestyles 5) identifying biomarkers that are priority therapeutic targets and 6) innovative prognostic tools.
Gene-lifestyle interplay is a major future growth area. This world-leading study will enhance Europes strengths in public health, nutrition and genetic epidemiology, and create a valuable future resource.
Max ERC Funding
2 499 154 €
Duration
Start date: 2011-05-01, End date: 2017-04-30
Project acronym EPIROSE
Project Self-Organising Capacity of Stem Cells during Implantation and Early Post-implantation Development: Implications for Human Development
Researcher (PI) Magdalena Zernicka-goetz
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Advanced Grant (AdG), LS3, ERC-2014-ADG
Summary Embryonic development progresses through successive cell fate decisions and intricate three-dimensional morphogenetic transformations. Implantation is the defining event in mammalian pregnancy during which a fundamental morphogenetic transformation is initiated: the body axes are established and the embryonic germ layers created. Despite its importance, a comprehensive understanding of the molecular mechanisms, transcriptional pathways, cellular interactions, as well as the spatio-temporal development of the embryo at implantation stages is at present lacking, due to the embryo’s inaccessibility. To overcome these limitations, we generated a culture system that allows the development of mouse implanting embryos outside of the mother. This system provides the opportunity to address how architectural features and signaling events integrate to induce the emergence of the body plan. Combining this new technology with the analysis of genetically engineered mouse embryos, the aim of this research proposal is to fill the knowledge gap between pre and post-implantation development. Single cell sequencing, two-photon microscopy, high-content forward genetic screening, and modeling will be merged with a functional assessment of embryo development in vivo to reveal the determinants of implantation and early post-implantation development. This global understanding will be employed to explore the extent to which stem cells can recapitulate embryonic development, with tremendous potential for regenerative medicine. Knowledge of the cellular and molecular mechanisms that intertwine lineage specification, developmental potential, and tissue morphogenesis will offer novel insight on the pathological causes of embryo lethality and congenital disorders. The proposed studies will shed light on this crucial yet mysterious stage of development in the mouse and, by extrapolation, offer outstanding potential to advance our understanding of human development.
Summary
Embryonic development progresses through successive cell fate decisions and intricate three-dimensional morphogenetic transformations. Implantation is the defining event in mammalian pregnancy during which a fundamental morphogenetic transformation is initiated: the body axes are established and the embryonic germ layers created. Despite its importance, a comprehensive understanding of the molecular mechanisms, transcriptional pathways, cellular interactions, as well as the spatio-temporal development of the embryo at implantation stages is at present lacking, due to the embryo’s inaccessibility. To overcome these limitations, we generated a culture system that allows the development of mouse implanting embryos outside of the mother. This system provides the opportunity to address how architectural features and signaling events integrate to induce the emergence of the body plan. Combining this new technology with the analysis of genetically engineered mouse embryos, the aim of this research proposal is to fill the knowledge gap between pre and post-implantation development. Single cell sequencing, two-photon microscopy, high-content forward genetic screening, and modeling will be merged with a functional assessment of embryo development in vivo to reveal the determinants of implantation and early post-implantation development. This global understanding will be employed to explore the extent to which stem cells can recapitulate embryonic development, with tremendous potential for regenerative medicine. Knowledge of the cellular and molecular mechanisms that intertwine lineage specification, developmental potential, and tissue morphogenesis will offer novel insight on the pathological causes of embryo lethality and congenital disorders. The proposed studies will shed light on this crucial yet mysterious stage of development in the mouse and, by extrapolation, offer outstanding potential to advance our understanding of human development.
Max ERC Funding
2 477 951 €
Duration
Start date: 2016-01-01, End date: 2021-12-31
Project acronym EPISUSCEPTIBILITY
Project Epigenome and Cancer Susceptibility
Researcher (PI) Päivi Tuulikki Peltomäki
Host Institution (HI) HELSINGIN YLIOPISTO
Call Details Advanced Grant (AdG), LS7, ERC-2008-AdG
Summary Early detection is crucial for the outcome of most cancers. Prevention of cancer development is even more desirable. To facilitate these ultimate goals we aim to construct a comprehensive view of the stepwise process through which common human cancers, such as colorectal cancer, arise. In particular, we aim to identify novel mechanisms of cancer susceptibility by focusing on the epigenome, whose alterations may underlie several phenomena related to chronic adult-onset disease that are not explained by genetics alone. The stepwise process of carcinogenesis can be accelerated or halted for various reasons, including inherited susceptibility and diet. The human multi-organ cancer syndromes hereditary nonpolyposis colorectal cancer (HNPCC) and familial adenomatous polyposis (FAP) as well as their murine counterparts, the Mlh1+/- mouse and the ApcMin/+ mouse, will be used as shortcuts to study the interplay between the epigenome and genome in tumorigenesis and to identify biomarkers of cancer susceptibility, malignant transformation, and tumor progression. This will be achieved by molecular profiling of normal and tumor tissues, cell line studies, in vitro functional assays, and in silico approaches. Additionally, the role that the epigenome plays to mediate the effects of the Western type diet on colorectal tumorigenesis will be examined in the mouse. Unlike genetic changes, epigenetic alterations are potentially reversible, which makes them promising targets for preventive and therapeutic interventions.
Summary
Early detection is crucial for the outcome of most cancers. Prevention of cancer development is even more desirable. To facilitate these ultimate goals we aim to construct a comprehensive view of the stepwise process through which common human cancers, such as colorectal cancer, arise. In particular, we aim to identify novel mechanisms of cancer susceptibility by focusing on the epigenome, whose alterations may underlie several phenomena related to chronic adult-onset disease that are not explained by genetics alone. The stepwise process of carcinogenesis can be accelerated or halted for various reasons, including inherited susceptibility and diet. The human multi-organ cancer syndromes hereditary nonpolyposis colorectal cancer (HNPCC) and familial adenomatous polyposis (FAP) as well as their murine counterparts, the Mlh1+/- mouse and the ApcMin/+ mouse, will be used as shortcuts to study the interplay between the epigenome and genome in tumorigenesis and to identify biomarkers of cancer susceptibility, malignant transformation, and tumor progression. This will be achieved by molecular profiling of normal and tumor tissues, cell line studies, in vitro functional assays, and in silico approaches. Additionally, the role that the epigenome plays to mediate the effects of the Western type diet on colorectal tumorigenesis will be examined in the mouse. Unlike genetic changes, epigenetic alterations are potentially reversible, which makes them promising targets for preventive and therapeutic interventions.
Max ERC Funding
2 500 000 €
Duration
Start date: 2009-04-01, End date: 2014-09-30
Project acronym EPLORE
Project EPidemiological Left ventriclar Outcomes Research in Europe
Researcher (PI) Jan Albert Hendrik Staessen
Host Institution (HI) KATHOLIEKE UNIVERSITEIT LEUVEN
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Heart failure (HF) affects 15 million Europeans and entails higher mortality and health care costs than cancer. EPLORE addresses this issue by prospective epidemiological research in 4 European countries and by a proof-of-concept clinical trial. WP1 will for the first time at the population level document the incidence and progression of subclinical LV dysfunction and clarify whether asymptomatic LV dysfunction, as picked up by the newest echocardiographic techniques, predicts cardiovascular (CV) outcomes, including HF. WP2 will investigate the contribution of ventricular-arterial coupling disease and mechanical LV dyssynchrony to subclinical LV dysfunction. WP3 will identify a set of urinary polypeptides that signify early LV dysfunction and validate these biomarkers by showing that they predict deterioration of LV function, progression to HF and the incidence of CV complications over and beyond established risk factors. WP3 will also search for novel panels of circulating biomarkers, of which combined measurement will add information (accuracy, sensitivity and specificity) to established biomarkers (e.g., NT-proBNP) and identify genetic variants involved in the progression of LV dysfunction, either causally or as biomarker. WP4 consists of a randomised clinical trial to translate in a high-risk high-gain setting the results of WPs 1-3 into clinical practice and to identify a new treatment modality that potentially slows progression of diastolic LV dysfunction. Dissemination in WP5 will contribute to new guidelines for the prevention and treatment of HF. WP6 includes governance, monitoring research strategies and output, and protection of IPR. In conclusion, EPLORE will advance risk stratification and the early diagnosis of subclinical HF. The project will potentially result into specific treatments for diastolic LV dysfunction and inform guidelines for prevention and treatment of HF. It will benefit 20% of Europeans who currently have subclinical LV dysfunction.
Summary
Heart failure (HF) affects 15 million Europeans and entails higher mortality and health care costs than cancer. EPLORE addresses this issue by prospective epidemiological research in 4 European countries and by a proof-of-concept clinical trial. WP1 will for the first time at the population level document the incidence and progression of subclinical LV dysfunction and clarify whether asymptomatic LV dysfunction, as picked up by the newest echocardiographic techniques, predicts cardiovascular (CV) outcomes, including HF. WP2 will investigate the contribution of ventricular-arterial coupling disease and mechanical LV dyssynchrony to subclinical LV dysfunction. WP3 will identify a set of urinary polypeptides that signify early LV dysfunction and validate these biomarkers by showing that they predict deterioration of LV function, progression to HF and the incidence of CV complications over and beyond established risk factors. WP3 will also search for novel panels of circulating biomarkers, of which combined measurement will add information (accuracy, sensitivity and specificity) to established biomarkers (e.g., NT-proBNP) and identify genetic variants involved in the progression of LV dysfunction, either causally or as biomarker. WP4 consists of a randomised clinical trial to translate in a high-risk high-gain setting the results of WPs 1-3 into clinical practice and to identify a new treatment modality that potentially slows progression of diastolic LV dysfunction. Dissemination in WP5 will contribute to new guidelines for the prevention and treatment of HF. WP6 includes governance, monitoring research strategies and output, and protection of IPR. In conclusion, EPLORE will advance risk stratification and the early diagnosis of subclinical HF. The project will potentially result into specific treatments for diastolic LV dysfunction and inform guidelines for prevention and treatment of HF. It will benefit 20% of Europeans who currently have subclinical LV dysfunction.
Max ERC Funding
2 391 440 €
Duration
Start date: 2012-07-01, End date: 2017-06-30
Project acronym ERCOPE
Project The ER located master regulation of endosomal positioning and further movements
Researcher (PI) Jacobus (Jacques) NEEFJES
Host Institution (HI) ACADEMISCH ZIEKENHUIS LEIDEN
Call Details Advanced Grant (AdG), LS3, ERC-2015-AdG
Summary The endo-lysosomal system is critical to diverse processes, including protein homeostasis, signaling and antigen presentation. The vesicular compartment is organized as a collective unit wherein the bulk of endosomes derived from disparate origins resides in a cloud in the perinuclear region and extends outwards to include quickly moving vesicles in the periphery. At this busy intersection between the endocytic and biosynthetic pathways, lies the late endosomal compartment, responsible for protein degradation and antigen processing. In dendritic and other immune cells, this major constituent of the perinuclear cloud serves as a hub for MHC class II antigen loading. Previous work by us and others has elucidated key elements of MHC class II biology through the study of late endosomal transport to and from the cell periphery. It is clear that cell biology of endosomes is modulated by their proximity to other membrane compartments during transport, maturation, cargo selection and delivery and even during cytokinesis in cell division. However, how endosomal positioning in the perinuclear cloud and how their release for further transport is controlled remains largely unknown. The aim of this proposal is to define the molecular basis for endosomal positioning and then to interrogate the relationship between spatial regulation of the endocytic compartment and its functions with respect to i) MHC class II antigen presentation, ii) bacterial infection and iii) mitotic resolution. From a genome-wide siRNA screen for factors influencing MHC class II biology, we have identified a unique and previously uncharacterized ubiquitin ligase that resides in the ER membrane, from where it controls endosomal positioning and times their arrivals and departures as a function of its catalytic activity. On this basis, the work proposed herein is poised to resolve an entirely new molecular network in control of endosomal biology with implications for diverse biological processes.
Summary
The endo-lysosomal system is critical to diverse processes, including protein homeostasis, signaling and antigen presentation. The vesicular compartment is organized as a collective unit wherein the bulk of endosomes derived from disparate origins resides in a cloud in the perinuclear region and extends outwards to include quickly moving vesicles in the periphery. At this busy intersection between the endocytic and biosynthetic pathways, lies the late endosomal compartment, responsible for protein degradation and antigen processing. In dendritic and other immune cells, this major constituent of the perinuclear cloud serves as a hub for MHC class II antigen loading. Previous work by us and others has elucidated key elements of MHC class II biology through the study of late endosomal transport to and from the cell periphery. It is clear that cell biology of endosomes is modulated by their proximity to other membrane compartments during transport, maturation, cargo selection and delivery and even during cytokinesis in cell division. However, how endosomal positioning in the perinuclear cloud and how their release for further transport is controlled remains largely unknown. The aim of this proposal is to define the molecular basis for endosomal positioning and then to interrogate the relationship between spatial regulation of the endocytic compartment and its functions with respect to i) MHC class II antigen presentation, ii) bacterial infection and iii) mitotic resolution. From a genome-wide siRNA screen for factors influencing MHC class II biology, we have identified a unique and previously uncharacterized ubiquitin ligase that resides in the ER membrane, from where it controls endosomal positioning and times their arrivals and departures as a function of its catalytic activity. On this basis, the work proposed herein is poised to resolve an entirely new molecular network in control of endosomal biology with implications for diverse biological processes.
Max ERC Funding
2 383 625 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym ETAP
Project Tracing Evolution of Auxin Transport and Polarity in Plants
Researcher (PI) Jiri Friml
Host Institution (HI) INSTITUTE OF SCIENCE AND TECHNOLOGYAUSTRIA
Call Details Advanced Grant (AdG), LS3, ERC-2016-ADG
Summary Multicellularity in plants evolved independently from other eukaryotes and presents a unique, alternative way how to deal with challenges of life. A major plant developmental module is the directional transport for the plant hormone auxin. The crucial components are PIN auxin transporters, whose polar, subcellular localization determines directionality of auxin flow through tissues. PIN-dependent auxin transport represents a unique model for studying the functional link between basic cellular processes, such as vesicle trafficking and cell polarity, and their developmental outcome at the level of the multicellular organism. Despite decades of intensive research, the classical approaches in the established models are approaching their limits and many crucial questions remain unsolved, in particular related to PIN structure, regulatory motifs and evolutionary origin
I propose to start a new direction in my research using an evolutionary perspective. This promises to overcome current limitations and provides not only (i) interesting insights into PIN evolution and diversification, but also (ii) a unique opportunity to study how evolutionary conserved cellular mechanisms of e.g. endocytic trafficking evolved specific plug-ins to make them subject to plant-specific regulations. The characterization of (iii) prokaryotic PIN origin will provide a so urgently needed (iv) entry into PIN structural studies. To achieve these goals, we will also establish novel (v) genetic and cell biological models in the ancestral lineage of the land plants that will be of a great use for any plant evolutionary studies.
The intellectual and methodological challenges of such interdisciplinary strategy combining several lower and higher plant models are obvious, but our preliminary results at several fronts promise its feasibility and success to gain deeper understanding of exciting questions on evolution and mechanisms behind the coordination and specification of developmental programs.
Summary
Multicellularity in plants evolved independently from other eukaryotes and presents a unique, alternative way how to deal with challenges of life. A major plant developmental module is the directional transport for the plant hormone auxin. The crucial components are PIN auxin transporters, whose polar, subcellular localization determines directionality of auxin flow through tissues. PIN-dependent auxin transport represents a unique model for studying the functional link between basic cellular processes, such as vesicle trafficking and cell polarity, and their developmental outcome at the level of the multicellular organism. Despite decades of intensive research, the classical approaches in the established models are approaching their limits and many crucial questions remain unsolved, in particular related to PIN structure, regulatory motifs and evolutionary origin
I propose to start a new direction in my research using an evolutionary perspective. This promises to overcome current limitations and provides not only (i) interesting insights into PIN evolution and diversification, but also (ii) a unique opportunity to study how evolutionary conserved cellular mechanisms of e.g. endocytic trafficking evolved specific plug-ins to make them subject to plant-specific regulations. The characterization of (iii) prokaryotic PIN origin will provide a so urgently needed (iv) entry into PIN structural studies. To achieve these goals, we will also establish novel (v) genetic and cell biological models in the ancestral lineage of the land plants that will be of a great use for any plant evolutionary studies.
The intellectual and methodological challenges of such interdisciplinary strategy combining several lower and higher plant models are obvious, but our preliminary results at several fronts promise its feasibility and success to gain deeper understanding of exciting questions on evolution and mechanisms behind the coordination and specification of developmental programs.
Max ERC Funding
2 410 292 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym EU-rhythmy
Project Molecular strategies to treat inherited arrhythmias
Researcher (PI) Silvia Giuliana Priori
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PAVIA
Call Details Advanced Grant (AdG), LS7, ERC-2014-ADG
Summary Sudden cardiac death (SCD) is a leading cause of death in western countries: coronary artery disease is the major cause of SCD in older subjects while inherited arrhythmogenic diseases are the leading cause of SCD in younger individuals. After 25 years dedicated to research of the molecular bases of heritable arrhythmias, the PI of this proposal now intends to pioneer gene therapy for prevention of SCD: a virtually unexplored field. The development of molecular therapies for rhythm disturbances is a high risk effort however, if successful, it will be highly rewarding. The PI has envisioned an ambitious and comprehensive project to target two severe inherited arrhythmogenic diseases: dominant catecholaminergic polymorphic ventricular tachycardia (CPVT) and Long QT syndrome type 8 (LQT8). The availability of a clinically relevant model is critical to ensure clinical translation of results: the team will exploit an existing CPVT model and will engineer a knock-in pig to model LQT8. The PI and her team will investigate innovative strategies of gene-delivery, gene-silencing and gene-editing to the heart comparing efficacy of different constructs and promoters. The team will also carefully engineer novel gene-therapy approaches to avoid the development of regional inhomogeneity in protein expression that may facilitate proarrhythmic events. Such a comprehensive approach will provide a most valuable core of knowledge on the comparative efficacy of a broad range of molecular strategies on the electrical milieu of the heart. It is expected that these results will not only benefit CPVT and LQT8 but rather they will foster development of gene therapy for other inherited and acquired arrhythmias.
Summary
Sudden cardiac death (SCD) is a leading cause of death in western countries: coronary artery disease is the major cause of SCD in older subjects while inherited arrhythmogenic diseases are the leading cause of SCD in younger individuals. After 25 years dedicated to research of the molecular bases of heritable arrhythmias, the PI of this proposal now intends to pioneer gene therapy for prevention of SCD: a virtually unexplored field. The development of molecular therapies for rhythm disturbances is a high risk effort however, if successful, it will be highly rewarding. The PI has envisioned an ambitious and comprehensive project to target two severe inherited arrhythmogenic diseases: dominant catecholaminergic polymorphic ventricular tachycardia (CPVT) and Long QT syndrome type 8 (LQT8). The availability of a clinically relevant model is critical to ensure clinical translation of results: the team will exploit an existing CPVT model and will engineer a knock-in pig to model LQT8. The PI and her team will investigate innovative strategies of gene-delivery, gene-silencing and gene-editing to the heart comparing efficacy of different constructs and promoters. The team will also carefully engineer novel gene-therapy approaches to avoid the development of regional inhomogeneity in protein expression that may facilitate proarrhythmic events. Such a comprehensive approach will provide a most valuable core of knowledge on the comparative efficacy of a broad range of molecular strategies on the electrical milieu of the heart. It is expected that these results will not only benefit CPVT and LQT8 but rather they will foster development of gene therapy for other inherited and acquired arrhythmias.
Max ERC Funding
2 314 029 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym EVO500
Project Origin of a cell differentiation mechanism and its evolution over 500 million years of life on land
Researcher (PI) Liam Dolan
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Advanced Grant (AdG), LS3, ERC-2009-AdG
Summary The evolution of the first rooting systems approximately 470 million years ago was a critical event in the history of life on Earth because it allowed the growth of complex multicellular eukaryotic photosynthetic organisms – plants - on the surface of the land. Rooting systems are important because they facilitate the uptake of every chemical element in the plant body with the exception of carbon. The root systems of the
first land plants (liverworts) comprised a mass of unicellular tip-growing filaments (rhizoids) that grew from the plant surface into the soil. All root systems that evolved since then similarly comprise a system of tipgrowing filamentous cells located at the interface between the plant and the soil, indicating that the differentiation of filamentous root cells has been critical for root function for the past 470 million years. This proposal aims to characterize the origin and evolution of this essential cellular differentiation process. The proposed research is in three parts:
First we propose to define the mechanism that controlled the development of the first land plant root system by identifying genes that control liverwort rooting system (rhizoids) development and
characterizing their regulatory interactions.
Second we propose to determine if the mechanism that controlled the development of the first land
plant root system was inherited from algal ancestors.
Third we propose to characterize the mechanism that controls filamentous root hair growth in
Arabidopsis in response to environmental factors, and determine if it is conserved among land
plants.
In combination, these experiments will define the genetic mechanisms underpinning the development and evolution of one of the fundamental developmental processes in land plants.
Summary
The evolution of the first rooting systems approximately 470 million years ago was a critical event in the history of life on Earth because it allowed the growth of complex multicellular eukaryotic photosynthetic organisms – plants - on the surface of the land. Rooting systems are important because they facilitate the uptake of every chemical element in the plant body with the exception of carbon. The root systems of the
first land plants (liverworts) comprised a mass of unicellular tip-growing filaments (rhizoids) that grew from the plant surface into the soil. All root systems that evolved since then similarly comprise a system of tipgrowing filamentous cells located at the interface between the plant and the soil, indicating that the differentiation of filamentous root cells has been critical for root function for the past 470 million years. This proposal aims to characterize the origin and evolution of this essential cellular differentiation process. The proposed research is in three parts:
First we propose to define the mechanism that controlled the development of the first land plant root system by identifying genes that control liverwort rooting system (rhizoids) development and
characterizing their regulatory interactions.
Second we propose to determine if the mechanism that controlled the development of the first land
plant root system was inherited from algal ancestors.
Third we propose to characterize the mechanism that controls filamentous root hair growth in
Arabidopsis in response to environmental factors, and determine if it is conserved among land
plants.
In combination, these experiments will define the genetic mechanisms underpinning the development and evolution of one of the fundamental developmental processes in land plants.
Max ERC Funding
2 463 835 €
Duration
Start date: 2010-10-01, End date: 2015-09-30
Project acronym EYEGET
Project Gene therapy of inherited retinal diseases
Researcher (PI) Alberto AURICCHIO
Host Institution (HI) FONDAZIONE TELETHON
Call Details Advanced Grant (AdG), LS7, ERC-2015-AdG
Summary Inherited retinal degenerations (IRDs) are a major cause of blindness worldwide. IRD patients witness inexorable progressive vision loss as no therapy is currently available. In the last decade my group has significantly contributed to a change of this scenario by developing efficient adeno-associated viral (AAV) vectors for retinal gene therapy that are safe and effective in humans. The objective of EYEGET (EYE GEne Therapy) is to overcome some of the current major limitations in the field of retinal gene therapy to expand initial therapeutic successes to a larger number of IRDs. To achieve this, we propose to use four parallel, highly innovative and complementary approaches: i. expansion of the limited AAV cargo capacity by a novel methodology based on co-administration of multiple AAVs that reassemble in target retinal cells and reconstitute large genes; ii. targeting of frequent dominant gain-of-function mutations that cause RP using state-of-the-art AAV-mediated genome editing technologies; iii. induction of retinal cells clearance of toxic IRD products by AAV-mediated activation of autophagy and lysosomal function; iv. development of methodologies to directly convert fibroblasts to photoreceptors that can be transplanted in retinas from IRD patients with advanced PR loss and for whom in vivo gene therapy is no longer an option. We will use a combination of in vitro and in vivo state-of-the-art technologies including novel AAV vector design, high content screening of drugs that enhance AAV transduction, genome editing, and advanced in vivo retinal phenotyping to obtain proof-of-concept for each of these therapeutic strategies. The results from this study may impact the quality of life of millions of people worldwide by providing a cure based on gene and/or cell therapy for a large group of IRDs.
Summary
Inherited retinal degenerations (IRDs) are a major cause of blindness worldwide. IRD patients witness inexorable progressive vision loss as no therapy is currently available. In the last decade my group has significantly contributed to a change of this scenario by developing efficient adeno-associated viral (AAV) vectors for retinal gene therapy that are safe and effective in humans. The objective of EYEGET (EYE GEne Therapy) is to overcome some of the current major limitations in the field of retinal gene therapy to expand initial therapeutic successes to a larger number of IRDs. To achieve this, we propose to use four parallel, highly innovative and complementary approaches: i. expansion of the limited AAV cargo capacity by a novel methodology based on co-administration of multiple AAVs that reassemble in target retinal cells and reconstitute large genes; ii. targeting of frequent dominant gain-of-function mutations that cause RP using state-of-the-art AAV-mediated genome editing technologies; iii. induction of retinal cells clearance of toxic IRD products by AAV-mediated activation of autophagy and lysosomal function; iv. development of methodologies to directly convert fibroblasts to photoreceptors that can be transplanted in retinas from IRD patients with advanced PR loss and for whom in vivo gene therapy is no longer an option. We will use a combination of in vitro and in vivo state-of-the-art technologies including novel AAV vector design, high content screening of drugs that enhance AAV transduction, genome editing, and advanced in vivo retinal phenotyping to obtain proof-of-concept for each of these therapeutic strategies. The results from this study may impact the quality of life of millions of people worldwide by providing a cure based on gene and/or cell therapy for a large group of IRDs.
Max ERC Funding
2 499 564 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym EYELETS
Project A regenerative medicine approach in diabetes.
Researcher (PI) Per-Olof BERGGREN
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Advanced Grant (AdG), LS7, ERC-2018-ADG
Summary Pancreatic islet transplantation is essential for diabetes treatment. Outcome varies due to transplantation site, quality of islets and the fact that transplanted islets are affected by the same challenges as in situ islets. Tailor-making islets for transplantation by tissue engineering combined with a more favorable transplantation site that allows for both monitoring and local modulation of islet cells is thus instrumental. We have established the anterior chamber of the eye (ACE) as a favorable environment for long term survival of islet grafts and the cornea as a natural body window for non-invasive, longitudinal optical monitoring of islet function. ACE engrafted islets are able to maintain blood glucose homeostasis in diabetic animals. In addition to studies in non-human primates we are performing human clinical trials, the first patient already being transplanted. Tissue engineering of native islets is technically difficult. We will therefore apply genetically engineered islet organoids. This allows us to generate i) standardized material optimized for transplantation, function and survival, as well as ii) islet organoids suitable for monitoring (sensor islet organoids) and treating (metabolic islet organoids) insulin-dependent diabetes. We hypothesize that genetically engineered islet organoids transplanted to the ACE are superior to native pancreatic islets to monitor and treat insulin-dependent diabetes. Our overall aim is to create a platform allowing monitoring and treatment of insulin-dependent diabetes in mice that can be transferred to large animals for validation. The objective is to combine tissue engineering of islet cell organoids, transplantation to the ACE, synthetic biology, local pharmacological treatment strategies and the development of novel micro electronic/micro optical readout systems for islet cells. This regenerative medicine approach will follow our clinical trial programs and be transferred into the clinic to combat diabetes.
Summary
Pancreatic islet transplantation is essential for diabetes treatment. Outcome varies due to transplantation site, quality of islets and the fact that transplanted islets are affected by the same challenges as in situ islets. Tailor-making islets for transplantation by tissue engineering combined with a more favorable transplantation site that allows for both monitoring and local modulation of islet cells is thus instrumental. We have established the anterior chamber of the eye (ACE) as a favorable environment for long term survival of islet grafts and the cornea as a natural body window for non-invasive, longitudinal optical monitoring of islet function. ACE engrafted islets are able to maintain blood glucose homeostasis in diabetic animals. In addition to studies in non-human primates we are performing human clinical trials, the first patient already being transplanted. Tissue engineering of native islets is technically difficult. We will therefore apply genetically engineered islet organoids. This allows us to generate i) standardized material optimized for transplantation, function and survival, as well as ii) islet organoids suitable for monitoring (sensor islet organoids) and treating (metabolic islet organoids) insulin-dependent diabetes. We hypothesize that genetically engineered islet organoids transplanted to the ACE are superior to native pancreatic islets to monitor and treat insulin-dependent diabetes. Our overall aim is to create a platform allowing monitoring and treatment of insulin-dependent diabetes in mice that can be transferred to large animals for validation. The objective is to combine tissue engineering of islet cell organoids, transplantation to the ACE, synthetic biology, local pharmacological treatment strategies and the development of novel micro electronic/micro optical readout systems for islet cells. This regenerative medicine approach will follow our clinical trial programs and be transferred into the clinic to combat diabetes.
Max ERC Funding
2 500 000 €
Duration
Start date: 2020-01-01, End date: 2024-12-31
Project acronym FAST
Project Investigating new therapeutic approaches to Friedreich's Ataxia
Researcher (PI) Roberto Testi
Host Institution (HI) UNIVERSITA DEGLI STUDI DI ROMA TOR VERGATA
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Friedreich’s Ataxia (FRDA) is a devastating degenerative disease with no specific therapy. It is passed by autosomal recessive inheritance and affects 1:30,000 individuals in Caucasian populations. Symptoms appear in the first decade of life and include progressive and unremitting lack of movement coordination, leading to complete inability, and dilated cardiomyopathy leading to congestive heart failure, the most common cause of premature death. FRDA is due to the insufficient transcription of the gene coding for the mitochondrial protein frataxin. Reduced cellular levels of frataxin cause impaired mitochondrial function and increased sensitivity to oxidative stress, leading to accelerated cell death in critical tissues.
Severity of the disease critically depends on residual frataxin levels. Therapeutic efforts are mostly focused on increasing cellular frataxin . We found that frataxin is normally degraded by the ubiquitin-proteasome system. We identified the lysine responsible for the ubiquitination of frataxin and, by computational screening followed by experimental validation, we identified and validated a series of small molecules, called ubiquitin-competing molecules (UCM), that prevent frataxin ubiquitination and induce frataxin accumulation in cells derived from FRDA patients. Moreover, treatment with UCM partially rescues aconitase and ATP production defects in cells derived from FRDA patients.
Our goal is two fold: 1) submit a set of leads we already identified, as well as their new and more complex derivatives, to preclinical testing in FRDA mice 2) identify the E3 ligase that is responsible for frataxin ubiquitination, and investigate the possibility to use it as a druggable target for small molecules to prevent frataxin degradation.
Summary
Friedreich’s Ataxia (FRDA) is a devastating degenerative disease with no specific therapy. It is passed by autosomal recessive inheritance and affects 1:30,000 individuals in Caucasian populations. Symptoms appear in the first decade of life and include progressive and unremitting lack of movement coordination, leading to complete inability, and dilated cardiomyopathy leading to congestive heart failure, the most common cause of premature death. FRDA is due to the insufficient transcription of the gene coding for the mitochondrial protein frataxin. Reduced cellular levels of frataxin cause impaired mitochondrial function and increased sensitivity to oxidative stress, leading to accelerated cell death in critical tissues.
Severity of the disease critically depends on residual frataxin levels. Therapeutic efforts are mostly focused on increasing cellular frataxin . We found that frataxin is normally degraded by the ubiquitin-proteasome system. We identified the lysine responsible for the ubiquitination of frataxin and, by computational screening followed by experimental validation, we identified and validated a series of small molecules, called ubiquitin-competing molecules (UCM), that prevent frataxin ubiquitination and induce frataxin accumulation in cells derived from FRDA patients. Moreover, treatment with UCM partially rescues aconitase and ATP production defects in cells derived from FRDA patients.
Our goal is two fold: 1) submit a set of leads we already identified, as well as their new and more complex derivatives, to preclinical testing in FRDA mice 2) identify the E3 ligase that is responsible for frataxin ubiquitination, and investigate the possibility to use it as a druggable target for small molecules to prevent frataxin degradation.
Max ERC Funding
1 496 200 €
Duration
Start date: 2012-03-01, End date: 2015-02-28
Project acronym FitteR-CATABOLIC
Project Survival of the Fittest: On how to enhance recovery from critical illness through learning from evolutionary conserved catabolic pathways
Researcher (PI) Greta Herman VAN DEN BERGHE
Host Institution (HI) KATHOLIEKE UNIVERSITEIT LEUVEN
Call Details Advanced Grant (AdG), LS7, ERC-2017-ADG
Summary Since a few decades, human patients who suffer from severe illnesses or multiple trauma, conditions that were previously lethal, are being treated in intensive care units (ICUs). Modern intensive care medicine bridges patients from life-threatening conditions to recovery with use of mechanical devices, vasoactive drugs and powerful anti-microbial agents. By postponing death, a new unnatural condition, intensive-care-dependent prolonged (>1 week) critical illness, has been created. About 25% of ICU patients today require prolonged intensive care, sometimes for weeks or months, and these patients are at high risk of death while consuming 75% of resources. Although the primary insult was adequately dealt with, many long-stay patients typically suffer from hypercatabolism, ICU-acquired brain dysfunction and polyneuropathy/myopathy leading to severe muscle weakness, further increasing the risk of late death. As hypercatabolism was considered the culprit, several anabolic interventions were tested, but these showed harm instead of benefit. We previously showed that fasting early during illness is superior to forceful feeding, pointing to certain benefits of catabolic responses. In healthy humans, fasting activates catabolism to provide substrates essential to protect and maintain brain and muscle function. This proposal aims to investigate whether evolutionary conserved catabolic fasting pathways, specifically lipolysis and ketogenesis, can be exploited in the search for prevention of brain dysfunction and muscle weakness in long-stay ICU patients, with the goal to identify a new metabolic intervention to enhance their recovery. The project builds further on our experience with bi-directional translational research - using human material whenever possible and a validated mouse model of sepsis-induced critical illness for objectives that cannot be addressed in patients - and aims to close the loop, from a novel concept to a large randomized controlled trial in patients.
Summary
Since a few decades, human patients who suffer from severe illnesses or multiple trauma, conditions that were previously lethal, are being treated in intensive care units (ICUs). Modern intensive care medicine bridges patients from life-threatening conditions to recovery with use of mechanical devices, vasoactive drugs and powerful anti-microbial agents. By postponing death, a new unnatural condition, intensive-care-dependent prolonged (>1 week) critical illness, has been created. About 25% of ICU patients today require prolonged intensive care, sometimes for weeks or months, and these patients are at high risk of death while consuming 75% of resources. Although the primary insult was adequately dealt with, many long-stay patients typically suffer from hypercatabolism, ICU-acquired brain dysfunction and polyneuropathy/myopathy leading to severe muscle weakness, further increasing the risk of late death. As hypercatabolism was considered the culprit, several anabolic interventions were tested, but these showed harm instead of benefit. We previously showed that fasting early during illness is superior to forceful feeding, pointing to certain benefits of catabolic responses. In healthy humans, fasting activates catabolism to provide substrates essential to protect and maintain brain and muscle function. This proposal aims to investigate whether evolutionary conserved catabolic fasting pathways, specifically lipolysis and ketogenesis, can be exploited in the search for prevention of brain dysfunction and muscle weakness in long-stay ICU patients, with the goal to identify a new metabolic intervention to enhance their recovery. The project builds further on our experience with bi-directional translational research - using human material whenever possible and a validated mouse model of sepsis-induced critical illness for objectives that cannot be addressed in patients - and aims to close the loop, from a novel concept to a large randomized controlled trial in patients.
Max ERC Funding
2 500 000 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym FliesCan
Project Modelling Cancer Traits in Drosophila
Researcher (PI) Cayetano Gonzalez Hernandez
Host Institution (HI) FUNDACIO INSTITUT DE RECERCA BIOMEDICA (IRB BARCELONA)
Call Details Advanced Grant (AdG), LS3, ERC-2011-ADG_20110310
Summary Despite significant advance, cancer treatment remains suboptimal. Anatomical and physiological differences between humans and simple model organisms like Drosophila are many and major, and preclude the modelling of key aspects of the disease as it proceeds in vertebrates. However, malignant tumors in vertebrates and flies are made of cells that have derailed from their normal course of development, grow out of control, become immortal, invasive, and kill the host. Moreover, like most solid human tumors, Drosophila malignant tumors display chromosomal instability and copy number variation. In addition, some of them are characterized by the upregulation of germline genes, a distinct feature of certain human cancers. Drosophila tumor models offer an unprecedented opportunity to study these basic malignant traits, which characterize human tumors, in a genetically tractable organism, applying sophisticated genome-wide and comprehensive functional assays at a rate and with a level of detail that are not possible in vertebrates. The goal of this project is twofold: (1) to identify new paths of intervention to inhibit tumor growth, and (2) to determine the origin and function of aneuploidy and changes in gene copy number in malignant growth. We are expectant that the results obtained during the course of this project might eventually have a real impact in human health.
Summary
Despite significant advance, cancer treatment remains suboptimal. Anatomical and physiological differences between humans and simple model organisms like Drosophila are many and major, and preclude the modelling of key aspects of the disease as it proceeds in vertebrates. However, malignant tumors in vertebrates and flies are made of cells that have derailed from their normal course of development, grow out of control, become immortal, invasive, and kill the host. Moreover, like most solid human tumors, Drosophila malignant tumors display chromosomal instability and copy number variation. In addition, some of them are characterized by the upregulation of germline genes, a distinct feature of certain human cancers. Drosophila tumor models offer an unprecedented opportunity to study these basic malignant traits, which characterize human tumors, in a genetically tractable organism, applying sophisticated genome-wide and comprehensive functional assays at a rate and with a level of detail that are not possible in vertebrates. The goal of this project is twofold: (1) to identify new paths of intervention to inhibit tumor growth, and (2) to determine the origin and function of aneuploidy and changes in gene copy number in malignant growth. We are expectant that the results obtained during the course of this project might eventually have a real impact in human health.
Max ERC Funding
2 406 000 €
Duration
Start date: 2012-07-01, End date: 2017-06-30
Project acronym FLUPLAN
Project Novel strategies to combat future influenza pandemics
Researcher (PI) Albertus Dominicus Marcellinus Erasmus Osterhaus
Host Institution (HI) ERASMUS UNIVERSITAIR MEDISCH CENTRUM ROTTERDAM
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary The Mexican influenza A virus (H1N1) reminds us that the threat of an influenza pandemic is real. The 1918 Spanish flu virus, also started as a low pathogenic virus that mutated into a highly pathogenic virus within months, causing more than 50 million deaths. The Mexican influenza A virus (H1N1) may follow the same path. FLUPLAN will expand our knowledge of the packaging signals that govern reassortment events between influenza A viruses in general and between the Mexican influenza A virus (H1N1) and circulating human, porcine and highly pathogenic avian influenza A viruses in particular. FLUPLAN will thus lead to fundamental insights in the mechanisms that govern reassortment phenomena, providing a risk assessment concerning the pandemic potential of circulating avian and mammalian influenza A viruses. This will provide us with a panel of possible reassortant viruses of potentially pandemic nature. The MVA vaccine vector system that efficiently induced broad protective immunity against HPAI-H5N1 viruses in macaques, will be used for the preparation of a repository of MVA-H based pandemic vaccine seed viruses.The selection will be based on the reassortant viruses mentioned above, and on a repository of avian influenza viruses of the 16HA subtypes including the Mexican influenza A virus (H1N1) of avian/swine origin. The added value of including a relevant MVA-NP in the immunization schedule to obtain broader and longer protection will be determined in a macaque infection model. Collectively these studies will provide us with a highly versatile system that anticipates on future pandemic events by having seed viruses for vaccine development ready to go on the shelf, for the rapid production of broadly protective pandemic vaccines, which will save time and thus lives.
Summary
The Mexican influenza A virus (H1N1) reminds us that the threat of an influenza pandemic is real. The 1918 Spanish flu virus, also started as a low pathogenic virus that mutated into a highly pathogenic virus within months, causing more than 50 million deaths. The Mexican influenza A virus (H1N1) may follow the same path. FLUPLAN will expand our knowledge of the packaging signals that govern reassortment events between influenza A viruses in general and between the Mexican influenza A virus (H1N1) and circulating human, porcine and highly pathogenic avian influenza A viruses in particular. FLUPLAN will thus lead to fundamental insights in the mechanisms that govern reassortment phenomena, providing a risk assessment concerning the pandemic potential of circulating avian and mammalian influenza A viruses. This will provide us with a panel of possible reassortant viruses of potentially pandemic nature. The MVA vaccine vector system that efficiently induced broad protective immunity against HPAI-H5N1 viruses in macaques, will be used for the preparation of a repository of MVA-H based pandemic vaccine seed viruses.The selection will be based on the reassortant viruses mentioned above, and on a repository of avian influenza viruses of the 16HA subtypes including the Mexican influenza A virus (H1N1) of avian/swine origin. The added value of including a relevant MVA-NP in the immunization schedule to obtain broader and longer protection will be determined in a macaque infection model. Collectively these studies will provide us with a highly versatile system that anticipates on future pandemic events by having seed viruses for vaccine development ready to go on the shelf, for the rapid production of broadly protective pandemic vaccines, which will save time and thus lives.
Max ERC Funding
2 187 758 €
Duration
Start date: 2010-06-01, End date: 2015-05-31
Project acronym FUNMETA
Project Metabolomics of fungal diseases: a systems biology approach for biomarkers discovery and therapy
Researcher (PI) Luigina Romani
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PERUGIA
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Humans have evolved intimate symbiotic relationships with a consortium of gut microbes (including fungi) and individual variations in the microbiome influence host health and disease. The fact that fungi are capable of colonizing almost every niche within the human body suggests that they must possess particular immune adaptation mechanisms, the breakdown of which may result in fatal fungal infections and severe fungal diseases. Traditional reductionist approaches of the past have not been sufficient to address these new challenges in the pathogenesis of fungal diseases. Here, I propose an integrated, systems biology approach to understand the role of L-tryptophan (trp) metabolic pathways in multilevel host−fungus interactions. Present in mammals as well as in fungi, pathways of trp metabolic pathways are exploited by the host and the fungal biota for survival and immune adaptation. A variety of indole derivatives, generated through conversion from dietary trp by symbiotic bacteria, activate the aryl hydrocarbon receptor/IL-22 pathway that provides antifungal resistance and tissue repair. Harmful inflammatory responses to fungi are instead tamed by kynurenines generated via the enzyme indoleamine 2,3–dioxygenase (IDO) of the trp pathway. Through high-throughput wet-lab ‘omics’ techniques combined with computational techniques, the project aims at defining the molecular basis of mammalian and fungal IDO activity and a metabolic network linking the metabolic phenotype (metabotype) to immune adaptations and its possible breakdown in experimental and human fungal infections. The project will provide ideal post-graduate training focussed on the development of metabolomics for diagnosis of fungal diseases and optimization of current antifungal therapy and diet that are of relevance to public health care solutions.
Summary
Humans have evolved intimate symbiotic relationships with a consortium of gut microbes (including fungi) and individual variations in the microbiome influence host health and disease. The fact that fungi are capable of colonizing almost every niche within the human body suggests that they must possess particular immune adaptation mechanisms, the breakdown of which may result in fatal fungal infections and severe fungal diseases. Traditional reductionist approaches of the past have not been sufficient to address these new challenges in the pathogenesis of fungal diseases. Here, I propose an integrated, systems biology approach to understand the role of L-tryptophan (trp) metabolic pathways in multilevel host−fungus interactions. Present in mammals as well as in fungi, pathways of trp metabolic pathways are exploited by the host and the fungal biota for survival and immune adaptation. A variety of indole derivatives, generated through conversion from dietary trp by symbiotic bacteria, activate the aryl hydrocarbon receptor/IL-22 pathway that provides antifungal resistance and tissue repair. Harmful inflammatory responses to fungi are instead tamed by kynurenines generated via the enzyme indoleamine 2,3–dioxygenase (IDO) of the trp pathway. Through high-throughput wet-lab ‘omics’ techniques combined with computational techniques, the project aims at defining the molecular basis of mammalian and fungal IDO activity and a metabolic network linking the metabolic phenotype (metabotype) to immune adaptations and its possible breakdown in experimental and human fungal infections. The project will provide ideal post-graduate training focussed on the development of metabolomics for diagnosis of fungal diseases and optimization of current antifungal therapy and diet that are of relevance to public health care solutions.
Max ERC Funding
2 299 200 €
Duration
Start date: 2012-04-01, End date: 2018-03-31
Project acronym FUNSEL
Project Generation of AAV-based, arrayed genetic libraries for in vivo functional selection: an innovative approach to identify secreted factors and microRNAs against degenerative disorders
Researcher (PI) Mauro Giacca
Host Institution (HI) INTERNATIONAL CENTRE FOR GENETIC ENGINEERING AND BIOTECHNOLOGY
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary A foremost health problem stems from the burden of degenerative diseases, including heart failure, neurodegeneration, retinal degeneration and diabetes, essentially linked to the aging of the human population and the incapacity of post-mitotic tissues to undergo efficient repair. This is an ambitious, highly innovative project aimed at developing an in vivo selection procedure, based on gene transfer of two genetic libraries cloned into Adeno-Associated Virus (AAV)-based vectors, for the identification of novel secreted factors or microRNAs providing benefit against various degenerative diseases. Two arrayed libraries will be generated, one coding for ~1,300 cDNAs from the mouse secretome, the other for all known microRNAs (~800 genes). Pools of vectors from each library will be obtained with serotypes suitable for in vivo transduction of different organs. The vectors will be injected in a series of mouse models of degenerative disorders involving damage to cardiomyocytes,, neurodegeneration, retinal degeneration and loss of beta-cells in the pancreas. The degenerative conditions will drive the selection for secreted factors or miRNA putatively preventing cell apoptosis, enhancing residual cell function or, in the best possible scenario, promoting tissue regeneration. This in vivo selection approach, which is supported by very encouraging preliminary results, has never been attempted before and is rendered possible by the property of AAV vectors to be produced at high titers, infect tissues at high multiplicity, persist in the transduced cells for prolonged period of times and efficiently express their transgenes in vivo. In addition to its final goal of identifying novel biotherapeutics, the project entails the successful achievement of several intermediate objectives and is expected to extend both technology and knowledge beyond the state-of-the art.
Summary
A foremost health problem stems from the burden of degenerative diseases, including heart failure, neurodegeneration, retinal degeneration and diabetes, essentially linked to the aging of the human population and the incapacity of post-mitotic tissues to undergo efficient repair. This is an ambitious, highly innovative project aimed at developing an in vivo selection procedure, based on gene transfer of two genetic libraries cloned into Adeno-Associated Virus (AAV)-based vectors, for the identification of novel secreted factors or microRNAs providing benefit against various degenerative diseases. Two arrayed libraries will be generated, one coding for ~1,300 cDNAs from the mouse secretome, the other for all known microRNAs (~800 genes). Pools of vectors from each library will be obtained with serotypes suitable for in vivo transduction of different organs. The vectors will be injected in a series of mouse models of degenerative disorders involving damage to cardiomyocytes,, neurodegeneration, retinal degeneration and loss of beta-cells in the pancreas. The degenerative conditions will drive the selection for secreted factors or miRNA putatively preventing cell apoptosis, enhancing residual cell function or, in the best possible scenario, promoting tissue regeneration. This in vivo selection approach, which is supported by very encouraging preliminary results, has never been attempted before and is rendered possible by the property of AAV vectors to be produced at high titers, infect tissues at high multiplicity, persist in the transduced cells for prolonged period of times and efficiently express their transgenes in vivo. In addition to its final goal of identifying novel biotherapeutics, the project entails the successful achievement of several intermediate objectives and is expected to extend both technology and knowledge beyond the state-of-the art.
Max ERC Funding
1 824 000 €
Duration
Start date: 2010-04-01, End date: 2015-03-31
Project acronym FUSIMAGINE
Project A new neuroimaging modality: from bench to bedside
Researcher (PI) Mickaël Tanter
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary "NeuroImaging systems are invaluable tools in the understanding of the brain both for fundamental research and clinical diagnosis. However, recent improvements in deep brain imaging technology have been somewhat limited because most of them are based on incremental innovation of mature techniques (EEG, PET and fMRI) instead of breakthrough.
In FUSIMAGINE, a genuinely new functional brain imaging modality will be developed and validated whose performances could have a major impact in neuroscience from fundamental research to clinical applications.
This new modality is based on the use of ultrafast ultrasound scanners able to reach more than 10 000 frames per second (fps) compared to the usual 50 fps in conventional ultrasound scanners. This concept relies on compounded plane wave transmissions introduced by my team and demonstrates up to 100-fold increase in the sensitivity of blood flow measurements. It enables to image the subtle hemodynamic changes in small brain vessels and thus brain activity thanks to neurovascular coupling. Functional Ultrasound (fUS by analogy to fMRI) is a real breakthrough in brain imaging as our project will demonstrate that:
in neuroscience, fUS provides a unique real time, portable and deep brain functional imaging technique for awake and even freely moving small animal imaging, moreover with unprecedented spatiotemporal resolution (~100µm, 50ms).
in clinical diagnosis, fUS provides a unique bedside neuro-imaging system of newborns brain activity through the fontanel window. Such real time system will permit to monitor and better understand neonatal seizures and hemorrhages. On adults, fUS provides a unique functional imaging modality during neurosurgery to predict the cortical mapping remodeling resulting of tumor development (such as low-grade gliomas). Finally, new adaptive skull bone correction techniques implemented on the system will enable us to perform non invasive transcranial fUS imaging on human adults through the temple bone."
Summary
"NeuroImaging systems are invaluable tools in the understanding of the brain both for fundamental research and clinical diagnosis. However, recent improvements in deep brain imaging technology have been somewhat limited because most of them are based on incremental innovation of mature techniques (EEG, PET and fMRI) instead of breakthrough.
In FUSIMAGINE, a genuinely new functional brain imaging modality will be developed and validated whose performances could have a major impact in neuroscience from fundamental research to clinical applications.
This new modality is based on the use of ultrafast ultrasound scanners able to reach more than 10 000 frames per second (fps) compared to the usual 50 fps in conventional ultrasound scanners. This concept relies on compounded plane wave transmissions introduced by my team and demonstrates up to 100-fold increase in the sensitivity of blood flow measurements. It enables to image the subtle hemodynamic changes in small brain vessels and thus brain activity thanks to neurovascular coupling. Functional Ultrasound (fUS by analogy to fMRI) is a real breakthrough in brain imaging as our project will demonstrate that:
in neuroscience, fUS provides a unique real time, portable and deep brain functional imaging technique for awake and even freely moving small animal imaging, moreover with unprecedented spatiotemporal resolution (~100µm, 50ms).
in clinical diagnosis, fUS provides a unique bedside neuro-imaging system of newborns brain activity through the fontanel window. Such real time system will permit to monitor and better understand neonatal seizures and hemorrhages. On adults, fUS provides a unique functional imaging modality during neurosurgery to predict the cortical mapping remodeling resulting of tumor development (such as low-grade gliomas). Finally, new adaptive skull bone correction techniques implemented on the system will enable us to perform non invasive transcranial fUS imaging on human adults through the temple bone."
Max ERC Funding
2 497 603 €
Duration
Start date: 2014-05-01, End date: 2019-04-30
Project acronym FUTUREGENES
Project Gene transfer techniques in the treatment of cardiovascular diseases and malignant glioma
Researcher (PI) Seppo Yla-Herttuala
Host Institution (HI) ITA-SUOMEN YLIOPISTO
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary Background: Poor angiogenesis and collateral vessel formation lead to coronary heart disease, claudication, infarctions and amputations while malignant glioma is one of the most aggressive proangiogenic tumors leading to death in a few months. For these diseases either stimulation or blocking, respectively, of angiogenesis may provide novel treatment options. Advancing State-of-the-Art: Our hypothesis is that in ischemia it will be possible to support natural growth of blood vessels with Therapeutic angiogenesis and lymphangiogenesis by using local gene transfer of the new members of vascular endothelial growth factor (VEGF) family and their receptors. New co-receptors, designer mutants and PCR suffling products of VEGFs will be used. New vector technology will be used to achieve long-lasting effects of VEGFs. We aim to develop novel site-specifically integrating, targeted, regulated vectors to precisely express the new VEGFs, their soluble decoy receptors and single-chain therapeutic antibodies (scFv) for pro- and anti-angiogenic purposes. As novel approaches, we have developed metabolically biotinylated lenti- and adenoviruses suitable for targeting and Epigenetherapy where siRNA/miRNAs and short nuclear RNAs regulate endogenous gene expression at the VEGF promoter level via modification of histone code. scFv library for endothelial cells and lentivirus-siRNA library directed to all human and mouse kinases will be screened to identify new mediators of angiogenesis in order to develop next generation pro- and antiangiogenic therapies. Based on our strong track record in Clinical applications, the best new pro- and antiangiogenic approaches will be taken to phase I clinical studies in myocardial ischemia and malignant glioma. Significance: This work should lead to significant advances and new therapies for severe ischemia and malignant glioma. Epigenetherapy and new site-specifically integrating, regulated vectors should be widely applicable in medicine.
Summary
Background: Poor angiogenesis and collateral vessel formation lead to coronary heart disease, claudication, infarctions and amputations while malignant glioma is one of the most aggressive proangiogenic tumors leading to death in a few months. For these diseases either stimulation or blocking, respectively, of angiogenesis may provide novel treatment options. Advancing State-of-the-Art: Our hypothesis is that in ischemia it will be possible to support natural growth of blood vessels with Therapeutic angiogenesis and lymphangiogenesis by using local gene transfer of the new members of vascular endothelial growth factor (VEGF) family and their receptors. New co-receptors, designer mutants and PCR suffling products of VEGFs will be used. New vector technology will be used to achieve long-lasting effects of VEGFs. We aim to develop novel site-specifically integrating, targeted, regulated vectors to precisely express the new VEGFs, their soluble decoy receptors and single-chain therapeutic antibodies (scFv) for pro- and anti-angiogenic purposes. As novel approaches, we have developed metabolically biotinylated lenti- and adenoviruses suitable for targeting and Epigenetherapy where siRNA/miRNAs and short nuclear RNAs regulate endogenous gene expression at the VEGF promoter level via modification of histone code. scFv library for endothelial cells and lentivirus-siRNA library directed to all human and mouse kinases will be screened to identify new mediators of angiogenesis in order to develop next generation pro- and antiangiogenic therapies. Based on our strong track record in Clinical applications, the best new pro- and antiangiogenic approaches will be taken to phase I clinical studies in myocardial ischemia and malignant glioma. Significance: This work should lead to significant advances and new therapies for severe ischemia and malignant glioma. Epigenetherapy and new site-specifically integrating, regulated vectors should be widely applicable in medicine.
Max ERC Funding
2 200 000 €
Duration
Start date: 2010-06-01, End date: 2015-05-31
Project acronym GALECTCOMPART
Project Endocytic Membrane Compartmentalization by Galectins
Researcher (PI) Ludger Johannes
Host Institution (HI) INSTITUT CURIE
Call Details Advanced Grant (AdG), LS3, ERC-2013-ADG
Summary The contribution of clathrin-independent endocytosis to the cellular entry of signaling receptors, cell adhesion factors, and other cell surface molecules is well documented. However, how this process is initiated is still unknown. We have recently found that a cellular lectin, galectin-3 (Gal3), entered cells via morphologically distinct tubular structures, so-called clathrin-independent carriers (CLICs); Gal3 endocytosis was required for the uptake of the CLIC cargo CD44, a cell adhesion/migration factor; CD44 and Gal3 uptake required glycosphingolipids (GSLs); Gal3 induced tubular membrane invaginations in a GSL-dependent manner. Based on these findings we propose the groundbreaking hypothesis that Gal3 is an adaptor that clusters cargo proteins carrying defined carbohydrate modifications together with specific GSLs into nanoscale membrane environments whose mechanical properties drive the clathrin-independent formation of endocytic pits. In this program, we will first establish the compositional topology of endocytic galectin processes (Aim 1). The adaptor hypothesis will then be tested using innovative chemical biology tools (Aim 2). Quantitative models of cargo clustering and membrane shape changes will be developed on the basis of biophysical measurements and coarse grain simulations (Aim 3). Intravital imaging of endocytosis and cell migration in mice will finally explore how the functional link between galectins and GSLs contributes to wound healing in the colon (Aim 4). The molecular functions of galectins and GSLs in endocytosis — major unresolved questions in cellular membrane biology — will thereby be established, providing details from atomic arrangements via multi-molecular complexes and meso-scaled membrane domains to in vivo physiology. I am confident that this program will lead to the discovery of a new clathrin-independent endocytic mechanism by which different types of cargo are sorted to and internalized from specific membrane domains.
Summary
The contribution of clathrin-independent endocytosis to the cellular entry of signaling receptors, cell adhesion factors, and other cell surface molecules is well documented. However, how this process is initiated is still unknown. We have recently found that a cellular lectin, galectin-3 (Gal3), entered cells via morphologically distinct tubular structures, so-called clathrin-independent carriers (CLICs); Gal3 endocytosis was required for the uptake of the CLIC cargo CD44, a cell adhesion/migration factor; CD44 and Gal3 uptake required glycosphingolipids (GSLs); Gal3 induced tubular membrane invaginations in a GSL-dependent manner. Based on these findings we propose the groundbreaking hypothesis that Gal3 is an adaptor that clusters cargo proteins carrying defined carbohydrate modifications together with specific GSLs into nanoscale membrane environments whose mechanical properties drive the clathrin-independent formation of endocytic pits. In this program, we will first establish the compositional topology of endocytic galectin processes (Aim 1). The adaptor hypothesis will then be tested using innovative chemical biology tools (Aim 2). Quantitative models of cargo clustering and membrane shape changes will be developed on the basis of biophysical measurements and coarse grain simulations (Aim 3). Intravital imaging of endocytosis and cell migration in mice will finally explore how the functional link between galectins and GSLs contributes to wound healing in the colon (Aim 4). The molecular functions of galectins and GSLs in endocytosis — major unresolved questions in cellular membrane biology — will thereby be established, providing details from atomic arrangements via multi-molecular complexes and meso-scaled membrane domains to in vivo physiology. I am confident that this program will lead to the discovery of a new clathrin-independent endocytic mechanism by which different types of cargo are sorted to and internalized from specific membrane domains.
Max ERC Funding
2 270 054 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym GEMELLI
Project Gene networks controlling embryonic polarity, regulation and twinning
Researcher (PI) Claudio Daniel Stern
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Advanced Grant (AdG), LS3, ERC-2009-AdG
Summary Much of what we know about how embryos determine their axes of symmetry comes from research in invertebrates (mainly Drosophila) and cold-blooded vertebrates (mainly Xenopus). In both cases, polarity is set up by the localisation of maternal determinants in the cytoplasm of the fertilised egg. These determinants are inherited differentially by daughter cells, leading them to acquire different fates, which effectively fixes the axes of the embryo by the 8 cell stage. In contrast, in amniotes (reptiles, birds and mammals) embryonic polarity remains plastic until much later, just before gastrulation, when the embryo may contain as many as 50,000 cells. If an embryo at this stage is cut into fragments, each fragment can generate a complete embryo. This property, called "embryonic regulation", is thought to be responsible for the generation of monozygotic (identical) and conjoined ( Siamese ) twins in humans and other amniotes. We know almost nothing about how polarity is determined in higher vertebrates or about the mechanisms of embryonic regulation and twinning. This project uses a multi-disciplinary systems approach to reveal the gene interaction network controlling polarity, regulation and twinning. The project will also generate a mathematical model of early development or "virtual embryo", allowing prediction of experimental outcomes and clinical scenarios.
Summary
Much of what we know about how embryos determine their axes of symmetry comes from research in invertebrates (mainly Drosophila) and cold-blooded vertebrates (mainly Xenopus). In both cases, polarity is set up by the localisation of maternal determinants in the cytoplasm of the fertilised egg. These determinants are inherited differentially by daughter cells, leading them to acquire different fates, which effectively fixes the axes of the embryo by the 8 cell stage. In contrast, in amniotes (reptiles, birds and mammals) embryonic polarity remains plastic until much later, just before gastrulation, when the embryo may contain as many as 50,000 cells. If an embryo at this stage is cut into fragments, each fragment can generate a complete embryo. This property, called "embryonic regulation", is thought to be responsible for the generation of monozygotic (identical) and conjoined ( Siamese ) twins in humans and other amniotes. We know almost nothing about how polarity is determined in higher vertebrates or about the mechanisms of embryonic regulation and twinning. This project uses a multi-disciplinary systems approach to reveal the gene interaction network controlling polarity, regulation and twinning. The project will also generate a mathematical model of early development or "virtual embryo", allowing prediction of experimental outcomes and clinical scenarios.
Max ERC Funding
1 997 899 €
Duration
Start date: 2010-06-01, End date: 2016-02-29
Project acronym GENE FOR CURE
Project Expanding and extending gene therapy of monogenic diseases of the haematopoietic system
Researcher (PI) Marina Cavazzana
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2015-AdG
Summary Given that (i) not all patients with a monogenic disease affecting the lymphohaematopoietic system have an HLA-genoidentical sibling donor and (ii) severe immunological complications worsen the outcome in HLA-partially-matched hematopoietic stem cell transplantation (HSCT), the genetic modification of autologous hematopoietic stem cells (HSCs) has become a powerful curative treatment option for these individuals. The present project seeks to further consolidate the rationale for replacing HLA-partially-matched HSCT with a gene therapy approach. Wiskott-Aldrich syndrome is a primary immunodeficiency whose severity is due to impairment of both lymphoid and myeloid cell function. We have reported robust evidence showing that the infusion of gene-corrected autologous HSCs enables the restoration of the T cell function. However, we are still cautious with regard to claiming the stable correction of autoimmunity and thrombocytopenia. Accordingly, we plan to thoroughly monitor long-term B cell functional outcomes and the platelet count in our treated patients. Moreover, we wish to extend the gene therapy approach to the SCID caused by mutations in the DLREIC gene, since the long-term post-HSCT outcomes are particularly poor. The preclinical work has been completed; initiation of a clinical protocol is the next step. Immune-dysregulation polyendocrinopathy enteropathy X-linked (IPEX) and sickle cell anaemia (SCA) are the last two target diseases. IPEX is a devastating disease caused by mutation of FOXP3 transcription factor; it may be possible to correct it by infusing gene-modified CD4+Tcells. We intend to accumulate the data required to prove our working hypothesis. SCA is a worldwide public health issue. We are seeking to improve the conventional gene therapy procedure and to evaluate the efficacy of CrisP/Cas9-mediated disruption of the CIS-regulatory elements required for HbF silencing. This disruption may provide a cure for SCA.
Summary
Given that (i) not all patients with a monogenic disease affecting the lymphohaematopoietic system have an HLA-genoidentical sibling donor and (ii) severe immunological complications worsen the outcome in HLA-partially-matched hematopoietic stem cell transplantation (HSCT), the genetic modification of autologous hematopoietic stem cells (HSCs) has become a powerful curative treatment option for these individuals. The present project seeks to further consolidate the rationale for replacing HLA-partially-matched HSCT with a gene therapy approach. Wiskott-Aldrich syndrome is a primary immunodeficiency whose severity is due to impairment of both lymphoid and myeloid cell function. We have reported robust evidence showing that the infusion of gene-corrected autologous HSCs enables the restoration of the T cell function. However, we are still cautious with regard to claiming the stable correction of autoimmunity and thrombocytopenia. Accordingly, we plan to thoroughly monitor long-term B cell functional outcomes and the platelet count in our treated patients. Moreover, we wish to extend the gene therapy approach to the SCID caused by mutations in the DLREIC gene, since the long-term post-HSCT outcomes are particularly poor. The preclinical work has been completed; initiation of a clinical protocol is the next step. Immune-dysregulation polyendocrinopathy enteropathy X-linked (IPEX) and sickle cell anaemia (SCA) are the last two target diseases. IPEX is a devastating disease caused by mutation of FOXP3 transcription factor; it may be possible to correct it by infusing gene-modified CD4+Tcells. We intend to accumulate the data required to prove our working hypothesis. SCA is a worldwide public health issue. We are seeking to improve the conventional gene therapy procedure and to evaluate the efficacy of CrisP/Cas9-mediated disruption of the CIS-regulatory elements required for HbF silencing. This disruption may provide a cure for SCA.
Max ERC Funding
2 445 268 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym GENECADD
Project GEnetic NEtworks as a tool for anti-CAncer Drug Development
Researcher (PI) Ulf Thomas Edvard Helleday
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary Although several therapies target cellular pathways, current small molecules drug discovery is based on identification of inhibitors to single proteins, without knowledge of whether they are the most advantageous target. The objective of this proposal is to develop a novel method for drug discovery, combining phenotypic cell based screens with functional genetic networks to determine the molecular mechanisms of numerous small molecule inhibitors. This method will enable identification of numerous distinct inhibitors of a particular pathway, as well as providing their molecular mechanism.
Cancer cells harbour gene mutations that make them more reliant on other cellular pathways for survival. Such cellular pathways can be targeted to selectively kill the cancer cells using the concept of synthetic lethality. In this project we want to identify inhibitors of homologous recombination to target cancer using synthetic lethality.
To establish a functional genetic network for homologous recombination, we will first identify all recombination proteins using multiple genome-wide RNAi screens. Then the synthetic sick or lethal interaction map between all recombination proteins is determined by co-depletion of these. Such synthetic sick or lethal network will identify numerous putative targets for anti-cancer treatment. Importantly, using this network for chemical-genetic functional interactions will assist in determinating of the molecular mechanisms of inhibitors. Chemical-genetic networks based on synthetic sickness or lethality can potentially change future drug discovery methods as well as providing new mechanistic insights into the field of toxicology.
Summary
Although several therapies target cellular pathways, current small molecules drug discovery is based on identification of inhibitors to single proteins, without knowledge of whether they are the most advantageous target. The objective of this proposal is to develop a novel method for drug discovery, combining phenotypic cell based screens with functional genetic networks to determine the molecular mechanisms of numerous small molecule inhibitors. This method will enable identification of numerous distinct inhibitors of a particular pathway, as well as providing their molecular mechanism.
Cancer cells harbour gene mutations that make them more reliant on other cellular pathways for survival. Such cellular pathways can be targeted to selectively kill the cancer cells using the concept of synthetic lethality. In this project we want to identify inhibitors of homologous recombination to target cancer using synthetic lethality.
To establish a functional genetic network for homologous recombination, we will first identify all recombination proteins using multiple genome-wide RNAi screens. Then the synthetic sick or lethal interaction map between all recombination proteins is determined by co-depletion of these. Such synthetic sick or lethal network will identify numerous putative targets for anti-cancer treatment. Importantly, using this network for chemical-genetic functional interactions will assist in determinating of the molecular mechanisms of inhibitors. Chemical-genetic networks based on synthetic sickness or lethality can potentially change future drug discovery methods as well as providing new mechanistic insights into the field of toxicology.
Max ERC Funding
2 500 000 €
Duration
Start date: 2011-03-01, End date: 2016-02-29
Project acronym GLYCOTREAT
Project Novel vaccine generation for the treatment of cancer. A glyco-nanomedial approach instructing T cells
Researcher (PI) Yvette Van Kooyk
Host Institution (HI) STICHTING VUMC
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary There is an urgent need to develop vaccines for the induction of CD8+ T-cell immunity to treat cancer and infectious diseases. Dendritic Cells (DC) have shown potential to induce antigen specific CD8+ T-cell responses with the help of CD4+ T cells, yet the efficacy by which the induction is achieved still has its limitations. The main challenge is: (a) to increase targeting efficacy to the complete repertoire of DC subsets; (b) to trigger T-cell responses by the DC that is powerful enough to eliminate a tumour (c) to implement novel human read-out systems, that mimic he human body response to evaluate vaccine efficacy.
The aim of this research project is to develop new glycan-based nanomedicines targeted to DC to induce powerful T-cell responses. Within the scope of this research project these new glycan-based nanomedicines will be tested to (i) trigger a strong T-cell response to pathogens, (ii) induce a powerful and adequate T-cell response to self antigen in a tumour induced immune suppressive environment and (iii) render fundamental insights to establish a vaccine platform relevant for the treatment of cancer and infectious diseases.
GlycoTreat employs an unconventional, novel glycan biotechnology approach to target a multitude of DC subsets in the human skin to validate the groundbreaking hypothesis that the local administration and molecular size and glycan valency of the targeting compound affect the efficiency of the T-cell stimulating vaccine. This research project joins the chemical design of glyco-nanomedical vaccines with immunological outcomes in our advanced in-vitro, in-situ human skin and in-vivo mouse DC model systems. While crossing the established disciplinary boundaries between chemistry, biology and medicine, Prof. van Kooyk will generate a new field of expertise in vaccine development applied in the field of cancer treatment.
Summary
There is an urgent need to develop vaccines for the induction of CD8+ T-cell immunity to treat cancer and infectious diseases. Dendritic Cells (DC) have shown potential to induce antigen specific CD8+ T-cell responses with the help of CD4+ T cells, yet the efficacy by which the induction is achieved still has its limitations. The main challenge is: (a) to increase targeting efficacy to the complete repertoire of DC subsets; (b) to trigger T-cell responses by the DC that is powerful enough to eliminate a tumour (c) to implement novel human read-out systems, that mimic he human body response to evaluate vaccine efficacy.
The aim of this research project is to develop new glycan-based nanomedicines targeted to DC to induce powerful T-cell responses. Within the scope of this research project these new glycan-based nanomedicines will be tested to (i) trigger a strong T-cell response to pathogens, (ii) induce a powerful and adequate T-cell response to self antigen in a tumour induced immune suppressive environment and (iii) render fundamental insights to establish a vaccine platform relevant for the treatment of cancer and infectious diseases.
GlycoTreat employs an unconventional, novel glycan biotechnology approach to target a multitude of DC subsets in the human skin to validate the groundbreaking hypothesis that the local administration and molecular size and glycan valency of the targeting compound affect the efficiency of the T-cell stimulating vaccine. This research project joins the chemical design of glyco-nanomedical vaccines with immunological outcomes in our advanced in-vitro, in-situ human skin and in-vivo mouse DC model systems. While crossing the established disciplinary boundaries between chemistry, biology and medicine, Prof. van Kooyk will generate a new field of expertise in vaccine development applied in the field of cancer treatment.
Max ERC Funding
2 498 736 €
Duration
Start date: 2014-02-01, End date: 2019-07-31
Project acronym GROLEO
Project The Genetics and Physiology of Growth and Size Determination
Researcher (PI) Pierre Leopold
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Advanced Grant (AdG), LS3, ERC-2010-AdG_20100317
Summary Animal growth is a complex process that is intimately linked to the developmental program in order to form fit adults with proper size and proportions. Genetics is an important determinant of growth, exemplified by the role of local diffusible molecules in setting organ proportions for a given species. In addition to this genetic control, organisms use adaptation mechanisms allowing modulating the size of individuals according to environmental cues, among which nutrition. Therefore, sophisticated cross-talks between local and global cues are at play for the determination of the final size of an individual. The major objective of this project is to tackle the mechanisms involved in coupling growth control with environmental cues, as well as the mechanisms participating in growth arrest and the determination of final size.
Our project proposes a blend of physiological and genetic approaches on the Drosophila model, with the use of tissue-targeted loss-of-function to unravel some of the important cross-talks existing between organs for the control of growth at the global level. We will develop these approaches to (i) unravel the molecular nature of tissue cross-talks involved in nutrient sensing and the control of insulin/IGF secretion; (ii) tackle the feed-back mechanisms linking the developmental clock to the growing state of tissues and organs.
These projects should bring new contributions in two separate fields related to growth control, Developmental Biology and Physiology, in an attempt to merge these complementary approaches into a broader vision of this fascinating biological question.
Summary
Animal growth is a complex process that is intimately linked to the developmental program in order to form fit adults with proper size and proportions. Genetics is an important determinant of growth, exemplified by the role of local diffusible molecules in setting organ proportions for a given species. In addition to this genetic control, organisms use adaptation mechanisms allowing modulating the size of individuals according to environmental cues, among which nutrition. Therefore, sophisticated cross-talks between local and global cues are at play for the determination of the final size of an individual. The major objective of this project is to tackle the mechanisms involved in coupling growth control with environmental cues, as well as the mechanisms participating in growth arrest and the determination of final size.
Our project proposes a blend of physiological and genetic approaches on the Drosophila model, with the use of tissue-targeted loss-of-function to unravel some of the important cross-talks existing between organs for the control of growth at the global level. We will develop these approaches to (i) unravel the molecular nature of tissue cross-talks involved in nutrient sensing and the control of insulin/IGF secretion; (ii) tackle the feed-back mechanisms linking the developmental clock to the growing state of tissues and organs.
These projects should bring new contributions in two separate fields related to growth control, Developmental Biology and Physiology, in an attempt to merge these complementary approaches into a broader vision of this fascinating biological question.
Max ERC Funding
2 500 000 €
Duration
Start date: 2011-07-01, End date: 2016-06-30
Project acronym GT-SKIN
Project Gene Therapy for Inherited Skin Adhesion Disorders
Researcher (PI) Fulvio Mavilio
Host Institution (HI) ASSOCIATION GENETHON
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary Epidermolysis bullosa (EB) is a family of severe skin adhesion defects characterized by disfiguring blistering, infections, visual impairment, and a high risk of cancer. We showed through pre-clinical and clinical investigation that transplantation of genetically corrected epithelia leads to long-term functional correction of EB. The current vectors, however, integrate in an uncontrolled fashion in the human genome, a cause of genotoxicity and potentially severe adverse events. The objective of this project is the development and pre-clinical evaluation of new gene targeting and gene correction technology, aimed at integrating therapeutic transgenes at specific loci or at correcting genetic defects by homologous recombination. Epidermal stem cells (EpSCs) and EB are ideal targets for the design and development of such technology, which is expected to have an impact on gene therapy of many other genetic diseases. We will design and test novel viral vectors for homologous recombination based on site-specific integrases and Zn-finger nucleases, and non-viral vectors for integrating large genes and complex regulatory sequences based on vertebrate transposons. All vectors will be tested in repopulating human EpSCs in vitro and in vivo, in a pre-clinical model of xenotransplantation of human skin on immunodeficient mice. A parallel, basic research program will aim at applying existing and novel genomic approaches to the definition of transcription factors, regulatory regions and gene expression programs involved in self renewal, commitment and differentiation of EpSCs. The anticipated output of the project is the development of next-generation technology for gene transfer, and the establishment of a knowledge base for a better utilization of EpSCs in gene therapy.
Summary
Epidermolysis bullosa (EB) is a family of severe skin adhesion defects characterized by disfiguring blistering, infections, visual impairment, and a high risk of cancer. We showed through pre-clinical and clinical investigation that transplantation of genetically corrected epithelia leads to long-term functional correction of EB. The current vectors, however, integrate in an uncontrolled fashion in the human genome, a cause of genotoxicity and potentially severe adverse events. The objective of this project is the development and pre-clinical evaluation of new gene targeting and gene correction technology, aimed at integrating therapeutic transgenes at specific loci or at correcting genetic defects by homologous recombination. Epidermal stem cells (EpSCs) and EB are ideal targets for the design and development of such technology, which is expected to have an impact on gene therapy of many other genetic diseases. We will design and test novel viral vectors for homologous recombination based on site-specific integrases and Zn-finger nucleases, and non-viral vectors for integrating large genes and complex regulatory sequences based on vertebrate transposons. All vectors will be tested in repopulating human EpSCs in vitro and in vivo, in a pre-clinical model of xenotransplantation of human skin on immunodeficient mice. A parallel, basic research program will aim at applying existing and novel genomic approaches to the definition of transcription factors, regulatory regions and gene expression programs involved in self renewal, commitment and differentiation of EpSCs. The anticipated output of the project is the development of next-generation technology for gene transfer, and the establishment of a knowledge base for a better utilization of EpSCs in gene therapy.
Max ERC Funding
2 210 000 €
Duration
Start date: 2011-12-01, End date: 2017-05-31
Project acronym GTNCTV
Project Gene therapy and nanotechnology based CNS targeted vectors
Researcher (PI) Mimoun Azzouz
Host Institution (HI) THE UNIVERSITY OF SHEFFIELD
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Targeting therapeutic genes selectively into the central nervous system (CNS) is a crucial precondition for translation of gene therapy strategies into human trials. The current multidisciplinary proposal integrates expertise identified as essential in the effective acceleration of research to overcome bottlenecks in the field including: 1) Inefficiency of therapy delivery to the CNS because of factors like the blood-brain barrier (BBB); 2) Poor understanding of disease mechanisms at the molecular and cellular levels. These problems must be overcome to develop fully effective treatments for neurological disorders. Currently the adeno-associated (AAV)-based system is one of the most refined and effective gene delivery systems for neuronal cells. In contrast to all other systems, it has been possible to engineer AAV9 to deliver genes through the BBB to the CNS by intravascular (IV) administration. However, following IV delivery, these vectors also target liver and other tissues, with significant potential for untoward effects. This has prompted us to adopt two major strategies: i) targeting of AAV9 vectors at the level of transcription by insertion of hybrid motor neuron specific promoters into the vector genome; ii) development of a CNS-targeted delivery approach based on state-of-the art nanoparticle-mediated encapsulation of AAV9 vectors. We anticipate that engineering strategies with the ability to restrict transgene expression to CNS tissue will significantly overcome various existing hurdles in CNS gene therapy development. Our objectives are to: 1) explore mechanisms leading to penetration of scAAV9 vectors through BBB since the exact mechanism of AAV9 diffusion through BBB is unknown; 2) design novel targeted strategies with enhanced tropism to CNS; 3) use CNS targeted vectors to investigate mechanisms of motor neuron death linked to mutations in RNA processing genes; 4) utilise CNS-targeted systems to test therapeutic strategies for motor neuron diseases.
Summary
Targeting therapeutic genes selectively into the central nervous system (CNS) is a crucial precondition for translation of gene therapy strategies into human trials. The current multidisciplinary proposal integrates expertise identified as essential in the effective acceleration of research to overcome bottlenecks in the field including: 1) Inefficiency of therapy delivery to the CNS because of factors like the blood-brain barrier (BBB); 2) Poor understanding of disease mechanisms at the molecular and cellular levels. These problems must be overcome to develop fully effective treatments for neurological disorders. Currently the adeno-associated (AAV)-based system is one of the most refined and effective gene delivery systems for neuronal cells. In contrast to all other systems, it has been possible to engineer AAV9 to deliver genes through the BBB to the CNS by intravascular (IV) administration. However, following IV delivery, these vectors also target liver and other tissues, with significant potential for untoward effects. This has prompted us to adopt two major strategies: i) targeting of AAV9 vectors at the level of transcription by insertion of hybrid motor neuron specific promoters into the vector genome; ii) development of a CNS-targeted delivery approach based on state-of-the art nanoparticle-mediated encapsulation of AAV9 vectors. We anticipate that engineering strategies with the ability to restrict transgene expression to CNS tissue will significantly overcome various existing hurdles in CNS gene therapy development. Our objectives are to: 1) explore mechanisms leading to penetration of scAAV9 vectors through BBB since the exact mechanism of AAV9 diffusion through BBB is unknown; 2) design novel targeted strategies with enhanced tropism to CNS; 3) use CNS targeted vectors to investigate mechanisms of motor neuron death linked to mutations in RNA processing genes; 4) utilise CNS-targeted systems to test therapeutic strategies for motor neuron diseases.
Max ERC Funding
2 499 959 €
Duration
Start date: 2012-03-01, End date: 2017-10-31
Project acronym HealthierWomen
Project A woman's reproductive experience: Long-term implications for chronic disease and death
Researcher (PI) Rolv SKJAERVEN
Host Institution (HI) UNIVERSITETET I BERGEN
Call Details Advanced Grant (AdG), LS7, ERC-2018-ADG
Summary Pregnancy complications such as preeclampsia and preterm birth are known to affect infant health, but their influence on mothers’ long-term health is not well understood. Most previous studies are seriously limited by their reliance on information from the first pregnancy. Often they lack the data to study women’s complete reproductive histories. Without a complete reproductive history, the relationship between pregnancy complications and women’s long-term health cannot be reliably studied. The Medical Birth Registry of Norway, covering all births from 1967-, includes information on more than 3 million births and 1.5 million sibships. Linking this to population based death and cancer registries provides a worldwide unique source of population-based data which can be analysed to identify heterogeneities in risk by lifetime parity and the cumulative experience of pregnancy complications. Having worked in this field of research for many years, I see many erroneous conclusions in studies based on insufficient data. For instance, both after preeclampsia and after a stillbirth, the high risk of heart disease observed in one-child mothers is strongly attenuated in women with subsequent pregnancies. I will study different patterns of pregnancy complications that occur alone or in combination across pregnancies, and analyse their associations with cause specific maternal mortality. Using this unique methodology, I will challenge the idea that placental dysfunction is the origin of preeclampsia and test the hypothesis that pregnancy complications may cause direct long-term effects on maternal health. The findings of this research have the potential to advance our understanding of how pregnancy complications affect the long-term maternal health and help to develop more effective chronic disease prevention strategies.
Summary
Pregnancy complications such as preeclampsia and preterm birth are known to affect infant health, but their influence on mothers’ long-term health is not well understood. Most previous studies are seriously limited by their reliance on information from the first pregnancy. Often they lack the data to study women’s complete reproductive histories. Without a complete reproductive history, the relationship between pregnancy complications and women’s long-term health cannot be reliably studied. The Medical Birth Registry of Norway, covering all births from 1967-, includes information on more than 3 million births and 1.5 million sibships. Linking this to population based death and cancer registries provides a worldwide unique source of population-based data which can be analysed to identify heterogeneities in risk by lifetime parity and the cumulative experience of pregnancy complications. Having worked in this field of research for many years, I see many erroneous conclusions in studies based on insufficient data. For instance, both after preeclampsia and after a stillbirth, the high risk of heart disease observed in one-child mothers is strongly attenuated in women with subsequent pregnancies. I will study different patterns of pregnancy complications that occur alone or in combination across pregnancies, and analyse their associations with cause specific maternal mortality. Using this unique methodology, I will challenge the idea that placental dysfunction is the origin of preeclampsia and test the hypothesis that pregnancy complications may cause direct long-term effects on maternal health. The findings of this research have the potential to advance our understanding of how pregnancy complications affect the long-term maternal health and help to develop more effective chronic disease prevention strategies.
Max ERC Funding
2 500 000 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym HEPASPHER
Project Mimicking liver disease and regeneration in vitro for drug development and liver transplantation
Researcher (PI) Magnus INGELMAN-SUNDBERG
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary The liver is a vital organ for synthesis and detoxification. The most significant liver diseases are hepatitis, non alcoholic fatty liver disease (NAFLD), non-alcoholic fatty liver steatohepatitis (NASH), carcinoma and cirrhosis. An additional and important cause of liver injury is adverse drug reactions (ADRs). In particular NAFLD is the most common liver disease affecting between 20% and 44% of European adults and 43-70% of patients with type 2 diabetes, and is one prime cause for chronic and end-stage liver disease, such as cirrhosis and primary hepatocellular carcinoma.
This proposal is based on recent findings in the laboratory: The development of novel 3D spheroid system with chemically defined media allowing studies of chronic drug toxicity, relevant liver disease and liver function for 5 weeks in vitro, the finding of the role of miRNA in hepatocyte dedifferentiation and that hepatocytes during spheroid formation first de-differentiate but later in spheroids re-differentiate to an in vivo relevant phenotype. This forms the basis for the main objectives: i) to study diseased liver in vitro with identification of mechanisms, biomarkers and novel drug candidates for treatment of NAFLD and fibrosis, ii) evaluate drug toxicity sensitivity and mechanisms in diseased liver systems and iii) further develop methods for hepatocyte proliferation and regeneration in vitro for transplantation purposes, including genetic editing in cases of hepatocytes obtained from patients with genetically inherited liver diseases.
This work is carried out in close contact with the Hepatology unit at the Karolinska Hospital partly using resources at the Science for Life Laboratory at Karolinska. It is anticipated that the project can provide with novel mechanisms, biomarkers and new targets for treatment of liver disease as well as novel methods for clinically applicable liver regeneration without the use of stem cells or transformed cells.
Summary
The liver is a vital organ for synthesis and detoxification. The most significant liver diseases are hepatitis, non alcoholic fatty liver disease (NAFLD), non-alcoholic fatty liver steatohepatitis (NASH), carcinoma and cirrhosis. An additional and important cause of liver injury is adverse drug reactions (ADRs). In particular NAFLD is the most common liver disease affecting between 20% and 44% of European adults and 43-70% of patients with type 2 diabetes, and is one prime cause for chronic and end-stage liver disease, such as cirrhosis and primary hepatocellular carcinoma.
This proposal is based on recent findings in the laboratory: The development of novel 3D spheroid system with chemically defined media allowing studies of chronic drug toxicity, relevant liver disease and liver function for 5 weeks in vitro, the finding of the role of miRNA in hepatocyte dedifferentiation and that hepatocytes during spheroid formation first de-differentiate but later in spheroids re-differentiate to an in vivo relevant phenotype. This forms the basis for the main objectives: i) to study diseased liver in vitro with identification of mechanisms, biomarkers and novel drug candidates for treatment of NAFLD and fibrosis, ii) evaluate drug toxicity sensitivity and mechanisms in diseased liver systems and iii) further develop methods for hepatocyte proliferation and regeneration in vitro for transplantation purposes, including genetic editing in cases of hepatocytes obtained from patients with genetically inherited liver diseases.
This work is carried out in close contact with the Hepatology unit at the Karolinska Hospital partly using resources at the Science for Life Laboratory at Karolinska. It is anticipated that the project can provide with novel mechanisms, biomarkers and new targets for treatment of liver disease as well as novel methods for clinically applicable liver regeneration without the use of stem cells or transformed cells.
Max ERC Funding
2 413 449 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym HEPCENT
Project Molecular Analysis of Hepatitis C Virus Neutralization and Entry For the Development of Novel Antiviral Immunopreventive Strategies
Researcher (PI) François-Loic Cosset
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2008-AdG
Summary Hepatitis C virus (HCV) infection is a leading cause of chronic liver disease world-wide. HCV-induced end-stage liver disease such as liver cirrhosis and hepatocellular carcinoma represent a major concern in global health. Treatment options for chronic hepatitis C are limited and no vaccine against HCV infection is available. Vaccine development is hampered by several obstacles. High viral variability and escape from host immune responses render antigen selection a major challenge. Antigen selection requires thorough studies to identify conserved T cell and neutralization epitopes and to decipher neutralization mechanisms, aiming to discover the optimal viral target for immune responses counteracting HCV escape strategies. At the same time it is important to develop antigen presentation systems that are efficient in patients with impaired antiviral immune responses, as often observed during chronic hepatitis C. While most vaccine development programs are based on improving HCV cellular immunity, it is essential to associate, in a same vaccine formulation, immunogens able to induce broad spectrums neutralizing and cellular responses. Owing to recent progresses in the field, here we propose a project aiming to overcome the current limitations in vaccine development by addressing the improvement of B cell responses targeting HCV infection. This will be achieved by a detailed investigation of: 1) mechanisms of antibody-mediated neutralization and escape, 2) impact of lipoproteins associating with the viral particle during assembly/release and counteracting neutralization and 3) cell entry steps that can potentially be targeted by antibodies, including those that are not induced naturally. Thus, through the combined expertise of the team in molecular virology, immunology, clinical hepatology and vectorology, we aim to rationalize the development of B cell immunogens and neutralizing antibodies for novel antiviral immunopreventive strategies targeting HCV infection.
Summary
Hepatitis C virus (HCV) infection is a leading cause of chronic liver disease world-wide. HCV-induced end-stage liver disease such as liver cirrhosis and hepatocellular carcinoma represent a major concern in global health. Treatment options for chronic hepatitis C are limited and no vaccine against HCV infection is available. Vaccine development is hampered by several obstacles. High viral variability and escape from host immune responses render antigen selection a major challenge. Antigen selection requires thorough studies to identify conserved T cell and neutralization epitopes and to decipher neutralization mechanisms, aiming to discover the optimal viral target for immune responses counteracting HCV escape strategies. At the same time it is important to develop antigen presentation systems that are efficient in patients with impaired antiviral immune responses, as often observed during chronic hepatitis C. While most vaccine development programs are based on improving HCV cellular immunity, it is essential to associate, in a same vaccine formulation, immunogens able to induce broad spectrums neutralizing and cellular responses. Owing to recent progresses in the field, here we propose a project aiming to overcome the current limitations in vaccine development by addressing the improvement of B cell responses targeting HCV infection. This will be achieved by a detailed investigation of: 1) mechanisms of antibody-mediated neutralization and escape, 2) impact of lipoproteins associating with the viral particle during assembly/release and counteracting neutralization and 3) cell entry steps that can potentially be targeted by antibodies, including those that are not induced naturally. Thus, through the combined expertise of the team in molecular virology, immunology, clinical hepatology and vectorology, we aim to rationalize the development of B cell immunogens and neutralizing antibodies for novel antiviral immunopreventive strategies targeting HCV infection.
Max ERC Funding
2 447 357 €
Duration
Start date: 2009-04-01, End date: 2014-12-31
Project acronym HEPCIR
Project Cell circuits as targets and biomarkers for liver disease and cancer prevention
Researcher (PI) Thomas Baumert
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2014-ADG
Summary Chronic liver diseases such as liver cirrhosis and hepatocellular carcinoma (HCC) are major challenges for global health. HCC is the second leading and fastest rising cause of cancer death worldwide. The limited availability of therapeutic options reflects our poor understanding of the molecular and clinical mechanisms involved in progression of liver disease. Chronic hepatitis C virus (HCV) infection is a main risk factor for HCC. Although HCC may be avoided by addressing the underlying cause in early stage disease, strategies to prevent HCC in patients with established cirrhosis and advanced fibrosis, in which the risk of HCC persists despite treatment of the underlying cause are lacking. Indeed, even HCV cure does not eliminate the risk of HCC development when advanced fibrosis is already present. Since fibrosis/cirrhosis-driven carcinogenesis is the mechanism of HCC development common to all major etiologies, we propose to use HCV-induced liver disease as a model to decipher the pan-etiology sequence of molecular events underlying disease progression and HCC. Our own data provide solid evidence that HCV infection alters pathways implicated in liver disease progression, including cirrhosis deterioration, HCC development, and overall and liver-specific death. Thus, the molecular investigation of these pathways will identify key cell circuits for the understanding of the pathogenesis of liver disease and HCC in general, and as broadly applicable pan-etiology diagnostic and therapeutic targets. Using a novel patient-derived cell culture model system for liver disease biology combined with advanced functional genomics, novel animal models and clinical investigation, we aim to uncover the cell circuits that are of clinical relevance for liver disease progression and cancer. By providing novel targets and biomarkers for liver disease and HCC prevention, this proposal will have a marked impact on the management and prognosis of patients with liver disease and HCC.
Summary
Chronic liver diseases such as liver cirrhosis and hepatocellular carcinoma (HCC) are major challenges for global health. HCC is the second leading and fastest rising cause of cancer death worldwide. The limited availability of therapeutic options reflects our poor understanding of the molecular and clinical mechanisms involved in progression of liver disease. Chronic hepatitis C virus (HCV) infection is a main risk factor for HCC. Although HCC may be avoided by addressing the underlying cause in early stage disease, strategies to prevent HCC in patients with established cirrhosis and advanced fibrosis, in which the risk of HCC persists despite treatment of the underlying cause are lacking. Indeed, even HCV cure does not eliminate the risk of HCC development when advanced fibrosis is already present. Since fibrosis/cirrhosis-driven carcinogenesis is the mechanism of HCC development common to all major etiologies, we propose to use HCV-induced liver disease as a model to decipher the pan-etiology sequence of molecular events underlying disease progression and HCC. Our own data provide solid evidence that HCV infection alters pathways implicated in liver disease progression, including cirrhosis deterioration, HCC development, and overall and liver-specific death. Thus, the molecular investigation of these pathways will identify key cell circuits for the understanding of the pathogenesis of liver disease and HCC in general, and as broadly applicable pan-etiology diagnostic and therapeutic targets. Using a novel patient-derived cell culture model system for liver disease biology combined with advanced functional genomics, novel animal models and clinical investigation, we aim to uncover the cell circuits that are of clinical relevance for liver disease progression and cancer. By providing novel targets and biomarkers for liver disease and HCC prevention, this proposal will have a marked impact on the management and prognosis of patients with liver disease and HCC.
Max ERC Funding
2 305 000 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym HERA
Project Host-environment interactions in the protection from asthma and allergies
Researcher (PI) Erika Von Mutius
Host Institution (HI) LUDWIG-MAXIMILIANS-UNIVERSITAET MUENCHEN
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary Asthma and allergies are chronic conditions affecting billions of Europeans. These complex diseases are determined by interplay of genetic and environmental factors. Treatments can control symptoms, but cannot cure or prevent the diseases. I, and my team, have shown that children are strongly protected from asthma and allergies when growing up in a farming environment rich in microbial exposures: the prevalence of asthma and hay fever is reduced over 5 fold. We have shown that environmental exposure to microbial compounds is inversely related to asthma and allergies. We have isolated microbes from animal sheds which protect mice from allergic airway inflammation. My team is now at a critical point to move this work forward to the next step, which is to systematically identify the microbes and their immuno-stimulatory compounds that protect from asthma and allergies. We have key resources in hand. In previous population based studies large numbers of environmental samples from farm and non farm children with and without asthma and allergies have been stored in biobanks. Genome wide genotyping data have also been gathered. The HERA project aims at applying the latest innovative high throughput sequencing techniques to comprehensively characterize the microbial ecology of these environmental samples. New methods for assessing microbial immuno-stimulatory substances will be used. These innovations will allow the HERA team to identify distinct asthma and allergy protective microbial exposures taking each individual s genetic susceptibility into account. Once protective microbial exposures have been identified, the responsible substances can be isolated. These substances can be developed into novel and effective prevention strategies to combat the asthma and allergy epidemic.
Summary
Asthma and allergies are chronic conditions affecting billions of Europeans. These complex diseases are determined by interplay of genetic and environmental factors. Treatments can control symptoms, but cannot cure or prevent the diseases. I, and my team, have shown that children are strongly protected from asthma and allergies when growing up in a farming environment rich in microbial exposures: the prevalence of asthma and hay fever is reduced over 5 fold. We have shown that environmental exposure to microbial compounds is inversely related to asthma and allergies. We have isolated microbes from animal sheds which protect mice from allergic airway inflammation. My team is now at a critical point to move this work forward to the next step, which is to systematically identify the microbes and their immuno-stimulatory compounds that protect from asthma and allergies. We have key resources in hand. In previous population based studies large numbers of environmental samples from farm and non farm children with and without asthma and allergies have been stored in biobanks. Genome wide genotyping data have also been gathered. The HERA project aims at applying the latest innovative high throughput sequencing techniques to comprehensively characterize the microbial ecology of these environmental samples. New methods for assessing microbial immuno-stimulatory substances will be used. These innovations will allow the HERA team to identify distinct asthma and allergy protective microbial exposures taking each individual s genetic susceptibility into account. Once protective microbial exposures have been identified, the responsible substances can be isolated. These substances can be developed into novel and effective prevention strategies to combat the asthma and allergy epidemic.
Max ERC Funding
2 155 697 €
Duration
Start date: 2010-05-01, End date: 2015-04-30
Project acronym HITSCIL
Project How intraflagellar transport shapes the cilium: a single-molecule systems study
Researcher (PI) Erwin J G PETERMAN
Host Institution (HI) STICHTING VU
Call Details Advanced Grant (AdG), LS3, ERC-2017-ADG
Summary Sensory cilia are organelles extending like antennas from many eukaryotic cells, with crucial functions in sensing and signalling. Cilia consist of an axoneme built of microtubules, enveloped by a specialized membrane. Ciliary development and maintenance depend critically on a specific, microtubule-based intracellular transport mechanism, intraflagellar transport (IFT). In my laboratory, we study the chemosensory cilia of C. elegans, which sense water-soluble molecules in the animal’s environment for chemotaxis. Over the past years, we have developed a unique set of quantitative, single-molecule fluorescence microscopy tools that allow us to visualize and quantify IFT dynamics with unprecedented detail in living animals. So far, our focus has been on the cooperation of the motor proteins driving IFT. The overall objective of my current proposal is to zoom out and shed light on the connection between ciliary structure, chemosensory function and IFT, from a systems perspective. Recent work has indicated that axoneme length is controlled by IFT. Preliminary results from my laboratory show that axoneme length changes dynamically in response to perturbations of IFT or cilia. Furthermore, we have shown that IFT is substantially affected upon exposure of animals to known repellent solutions. The four major aims in my proposal are to:
• determine how directional changes in IFT are regulated and are affected by external disturbances,
• understand the dynamics of the axonemal microtubules and how IFT affects these dynamics and vice versa,
• study how sensory ciliary function affects IFT and ciliary structure,
• further develop our (single-molecule) fluorescence microscopy toolbox by improving instrumentation and using better fluorescent probes and sensors.
These experiments will place my lab in a unique position to push forward our understanding of the relationship between structure, function and dynamics of transport of this fascinating and fundamental organelle.
Summary
Sensory cilia are organelles extending like antennas from many eukaryotic cells, with crucial functions in sensing and signalling. Cilia consist of an axoneme built of microtubules, enveloped by a specialized membrane. Ciliary development and maintenance depend critically on a specific, microtubule-based intracellular transport mechanism, intraflagellar transport (IFT). In my laboratory, we study the chemosensory cilia of C. elegans, which sense water-soluble molecules in the animal’s environment for chemotaxis. Over the past years, we have developed a unique set of quantitative, single-molecule fluorescence microscopy tools that allow us to visualize and quantify IFT dynamics with unprecedented detail in living animals. So far, our focus has been on the cooperation of the motor proteins driving IFT. The overall objective of my current proposal is to zoom out and shed light on the connection between ciliary structure, chemosensory function and IFT, from a systems perspective. Recent work has indicated that axoneme length is controlled by IFT. Preliminary results from my laboratory show that axoneme length changes dynamically in response to perturbations of IFT or cilia. Furthermore, we have shown that IFT is substantially affected upon exposure of animals to known repellent solutions. The four major aims in my proposal are to:
• determine how directional changes in IFT are regulated and are affected by external disturbances,
• understand the dynamics of the axonemal microtubules and how IFT affects these dynamics and vice versa,
• study how sensory ciliary function affects IFT and ciliary structure,
• further develop our (single-molecule) fluorescence microscopy toolbox by improving instrumentation and using better fluorescent probes and sensors.
These experiments will place my lab in a unique position to push forward our understanding of the relationship between structure, function and dynamics of transport of this fascinating and fundamental organelle.
Max ERC Funding
2 499 580 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym Homo.symbiosus
Project Assessing, preserving and restoring man-microbes symbiosis
Researcher (PI) JOEL DORE
Host Institution (HI) INSTITUT NATIONAL DE LA RECHERCHE AGRONOMIQUE
Call Details Advanced Grant (AdG), LS7, ERC-2017-ADG
Summary The microbiomics revolution has favoured the recognition of the gut as a true organ and the importance of man-microbes symbiosis in health and disease. Derived from a long co-evolution the latter has been challenged by numerous environmental triggers, modern lifestyles, changes in birth modalities, nutritional transition and therapeutic attitudes. A large fraction of the human population has tentatively entered a man-microbes dysbiotic state characterized by altered interactions between microbiome and host features with auto-aggravating crosstalk signals. The result is increased incidence of incurable immune-mediated diseases of modern societies that affect a third the human population on earth today and for which current therapeutics only address symptoms alleviation, rather than considering man as a holobiont.
In this context and its resulting threat for the human species, I will carry out a project geared to open a new era of individualized preventive care and novel gut ecology-based therapeutic approaches. The project will assemble insights and contributions from theoretical to experimental ecology, quantitative and functional microbiomics, preclinical work, cohort studies and clinical trials, so as to:
• Validate the concept of critical transition and alternative stable state as it applies to a shift from man-microbes symbiosis to disease-prone man-microbes dysbiosis
• Assess the potential of diet alone to promote such a shift
• Model the symbiosis-to-dysbiosis transitions and derive predictors of tipping points
• Propose counter-measures that may allow to break vicious circles and restore a balanced, health-prone, man-microbes symbiosis by concomitantly acting upon microbiome and host features
• Validate strategies to reinforce ecological robustness and restore man-microbes symbiosis
Based on a paradigm shift, the proposed work will set the grounds for future personalized preventive nutrition and clinical management considering man as a true holobiont.
Summary
The microbiomics revolution has favoured the recognition of the gut as a true organ and the importance of man-microbes symbiosis in health and disease. Derived from a long co-evolution the latter has been challenged by numerous environmental triggers, modern lifestyles, changes in birth modalities, nutritional transition and therapeutic attitudes. A large fraction of the human population has tentatively entered a man-microbes dysbiotic state characterized by altered interactions between microbiome and host features with auto-aggravating crosstalk signals. The result is increased incidence of incurable immune-mediated diseases of modern societies that affect a third the human population on earth today and for which current therapeutics only address symptoms alleviation, rather than considering man as a holobiont.
In this context and its resulting threat for the human species, I will carry out a project geared to open a new era of individualized preventive care and novel gut ecology-based therapeutic approaches. The project will assemble insights and contributions from theoretical to experimental ecology, quantitative and functional microbiomics, preclinical work, cohort studies and clinical trials, so as to:
• Validate the concept of critical transition and alternative stable state as it applies to a shift from man-microbes symbiosis to disease-prone man-microbes dysbiosis
• Assess the potential of diet alone to promote such a shift
• Model the symbiosis-to-dysbiosis transitions and derive predictors of tipping points
• Propose counter-measures that may allow to break vicious circles and restore a balanced, health-prone, man-microbes symbiosis by concomitantly acting upon microbiome and host features
• Validate strategies to reinforce ecological robustness and restore man-microbes symbiosis
Based on a paradigm shift, the proposed work will set the grounds for future personalized preventive nutrition and clinical management considering man as a true holobiont.
Max ERC Funding
2 491 014 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym HOPE
Project Host Protective Engineering of Cancer Immunity by Targeting the Intracellular Immune Checkpoint NR2F6
Researcher (PI) Gottfried BAIER
Host Institution (HI) MEDIZINISCHE UNIVERSITAT INNSBRUCK
Call Details Advanced Grant (AdG), LS7, ERC-2017-ADG
Summary "Because of its biological complexity, cancer is still poorly understood. Chronic inflammation has been shown, both experimentally and epidemiologically, to be a predisposition to, and also an inseparable aspect of clinically prevalent cancer entities. Therefore, a detailed understanding of both tumour and immune cell functions in cancer progression is a prerequisite for more successful therapeutic startegies. My team was the first to reveal the lymphocyte-intrinsic PKC/NR2F6 axis as an essential signalling node at the crossroads between inflammation and cancer. It is the mission of this project to identify molecular signatures that influence the risk of developing tumours employing established research tools and state-of-the-art genetic, biochemical, proteomic and transcriptomic as well as large scale CRISPR/Cas9 perturbation screening-based functional genomic technologies. Defining this as yet poorly elucidated effector pathway with its profoundly relevant role would enable development of preventive and immune-therapeutic strategies against NSCLC lung cancer and potentially also against other entities. Our three-pronged approach to achieve this goal is to: (i) delineate biological and clinical properties of the immunological PKC/NR2F6 network, (ii) validate NR2F6 as an immune-oncology combination target needed to overcome limitations to ""first generation anti-PD-1 checkpoint inhibitors"" rendering T cells capable of rejecting tumours and their metastases at distal organs and (iii) exploit human combinatorial T cell therapy concepts for prevention of immune-related adverse events as well as of tumour recurrence by reducing opportunities for the tumour to develop resistance in the clinic. Insight into the functions of NR2F6 pathway and involved mechanisms is a prerequisite for understanding how the microenvironment at the tumour site either supports tumour growth and spread or prevents tumour initiation and progression, the latter by host-protective cancer immunity."
Summary
"Because of its biological complexity, cancer is still poorly understood. Chronic inflammation has been shown, both experimentally and epidemiologically, to be a predisposition to, and also an inseparable aspect of clinically prevalent cancer entities. Therefore, a detailed understanding of both tumour and immune cell functions in cancer progression is a prerequisite for more successful therapeutic startegies. My team was the first to reveal the lymphocyte-intrinsic PKC/NR2F6 axis as an essential signalling node at the crossroads between inflammation and cancer. It is the mission of this project to identify molecular signatures that influence the risk of developing tumours employing established research tools and state-of-the-art genetic, biochemical, proteomic and transcriptomic as well as large scale CRISPR/Cas9 perturbation screening-based functional genomic technologies. Defining this as yet poorly elucidated effector pathway with its profoundly relevant role would enable development of preventive and immune-therapeutic strategies against NSCLC lung cancer and potentially also against other entities. Our three-pronged approach to achieve this goal is to: (i) delineate biological and clinical properties of the immunological PKC/NR2F6 network, (ii) validate NR2F6 as an immune-oncology combination target needed to overcome limitations to ""first generation anti-PD-1 checkpoint inhibitors"" rendering T cells capable of rejecting tumours and their metastases at distal organs and (iii) exploit human combinatorial T cell therapy concepts for prevention of immune-related adverse events as well as of tumour recurrence by reducing opportunities for the tumour to develop resistance in the clinic. Insight into the functions of NR2F6 pathway and involved mechanisms is a prerequisite for understanding how the microenvironment at the tumour site either supports tumour growth and spread or prevents tumour initiation and progression, the latter by host-protective cancer immunity."
Max ERC Funding
2 484 325 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym HYGIENE
Project THE HYGIENE HYPOTHESIS : REVISITING THE CONCEPT BY INTEGRATING EPIDEMIOLOGY AND MECHANISTIC STUDIES
Researcher (PI) Jean-François Bach
Host Institution (HI) INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary The hygiene hypothesis postulating the paradoxical protective role of infections on immune-mediated
diseases including atopy (i.e. atopic dermatitis, rhinitis, asthma) and more recently autoimmune diseases has
been the matter of extensive investigation. The aim of the present project is to validate this hypothesis
integrating epidemiological and experimental studies.
We will review in a meta-analysis published studies of direct and indirect markers of infections and atopic
diseases, and conduct a case-control study to analyse the association between infections and atopy using
atopic dermatitis as a prototypic model. This epidemiological study will assess the occurrence of different
infections and other risk factors related to the incidence of atopic dermatitis in children under age five in
Italy (300 cases and 600 controls).
This epidemiological study will be supported by experimental approaches addressing mechanistic questions
raised by the hygiene hypothesis. Experimental models will include induction of acute and chronic
bronchoconstriction/asthma, atopic dermatitis. In addition the project will aim at devising new mouse models
of atopy. The nature of infections providing protection against allergic diseases will be investigated to
characterise the difference at the molecular level between protective and non protective pathogens or their
derivatives. The underlying immune mechanisms notably homeostasis imbalance, antigenic competition,
stimulation of regulatory immune cells and Toll-like receptor involvement will be analysed. In addition to
developing integrated in vivo models, including the use of transgenic animals, efforts will be focussed on the
study of available synthetic pathogen-derived compounds showing a protective activity to better approach
their cellular and molecular mode of action. These studies may pave the way to novel and safe therapies that
could advantageously substitute for the “protective” immune stimulation induced by major infections.
Summary
The hygiene hypothesis postulating the paradoxical protective role of infections on immune-mediated
diseases including atopy (i.e. atopic dermatitis, rhinitis, asthma) and more recently autoimmune diseases has
been the matter of extensive investigation. The aim of the present project is to validate this hypothesis
integrating epidemiological and experimental studies.
We will review in a meta-analysis published studies of direct and indirect markers of infections and atopic
diseases, and conduct a case-control study to analyse the association between infections and atopy using
atopic dermatitis as a prototypic model. This epidemiological study will assess the occurrence of different
infections and other risk factors related to the incidence of atopic dermatitis in children under age five in
Italy (300 cases and 600 controls).
This epidemiological study will be supported by experimental approaches addressing mechanistic questions
raised by the hygiene hypothesis. Experimental models will include induction of acute and chronic
bronchoconstriction/asthma, atopic dermatitis. In addition the project will aim at devising new mouse models
of atopy. The nature of infections providing protection against allergic diseases will be investigated to
characterise the difference at the molecular level between protective and non protective pathogens or their
derivatives. The underlying immune mechanisms notably homeostasis imbalance, antigenic competition,
stimulation of regulatory immune cells and Toll-like receptor involvement will be analysed. In addition to
developing integrated in vivo models, including the use of transgenic animals, efforts will be focussed on the
study of available synthetic pathogen-derived compounds showing a protective activity to better approach
their cellular and molecular mode of action. These studies may pave the way to novel and safe therapies that
could advantageously substitute for the “protective” immune stimulation induced by major infections.
Max ERC Funding
2 099 999 €
Duration
Start date: 2010-08-01, End date: 2015-07-31
Project acronym HYPOXIMMUNO
Project Tackling the Achilles Heel of Immunotherapy: Validating imaging biomarkers and targeting the immunological niche of tumour hypoxia
Researcher (PI) Philippe LAMBIN
Host Institution (HI) UNIVERSITEIT MAASTRICHT
Call Details Advanced Grant (AdG), LS7, ERC-2015-AdG
Summary Lung cancer is the most common cause of death from cancer worldwide with 1.59 million deaths annually and it places the highest economic burden of all cancers on the EU at €18.8billion. These figures call for a major transformation of the therapeutic management of lung cancer. The recent developments in immunotherapy give new hope. At present a significant minority of patients benefit from immune therapy, which remains not curative when delivered alone. It is thus clear that improving the efficacy of immune therapy is needed as well as the identification of predictive biomarkers. Combining immunotherapy with radiotherapy improves effectiveness, but resistance is still common, most probably because of hypoxia. In HYPOXIMMUNO, we will open up a new paradigm in treating metastatic NSCLC. I will provide for the first time (pre)clinical proof of concept of (i) highly innovative CT-based radiomics and PET-hypoxia imaging patient stratification tools and (ii) a tri-modal curative hypoxia-targeting treatment strategy that complements current immunotherapy and radiotherapy approaches. This tri-modal curative treatment strategy is based on (1) high precision Stereotactic Radiotherapy (SABR) to kill cancer cells and trigger the immune system, combined with (2) a hypoxia-activated prodrug to target the hypoxic tumour cells resistant to immunotherapy and (3) a promising and innovative tumour specific antibody (Ab)-based immunotherapy (immunocytokine L19-IL2 to “push the accelerator” with or without a checkpoint inhibitor to “release the break”) to form a powerful synergistic immuno-oncology strategy. In HYPOXIMMUNO, I envision to lay the foundation for the next breakthrough in oncology, by improving patient selection and developing highly innovative cancer therapies that target hypoxia, to improve the quality of life and prolong survival of cancer patients, in particular patients with metastatic NSCLC (watch the animation summarising the project: https://youtu.be/oP9Gp4a0b_Q).
Summary
Lung cancer is the most common cause of death from cancer worldwide with 1.59 million deaths annually and it places the highest economic burden of all cancers on the EU at €18.8billion. These figures call for a major transformation of the therapeutic management of lung cancer. The recent developments in immunotherapy give new hope. At present a significant minority of patients benefit from immune therapy, which remains not curative when delivered alone. It is thus clear that improving the efficacy of immune therapy is needed as well as the identification of predictive biomarkers. Combining immunotherapy with radiotherapy improves effectiveness, but resistance is still common, most probably because of hypoxia. In HYPOXIMMUNO, we will open up a new paradigm in treating metastatic NSCLC. I will provide for the first time (pre)clinical proof of concept of (i) highly innovative CT-based radiomics and PET-hypoxia imaging patient stratification tools and (ii) a tri-modal curative hypoxia-targeting treatment strategy that complements current immunotherapy and radiotherapy approaches. This tri-modal curative treatment strategy is based on (1) high precision Stereotactic Radiotherapy (SABR) to kill cancer cells and trigger the immune system, combined with (2) a hypoxia-activated prodrug to target the hypoxic tumour cells resistant to immunotherapy and (3) a promising and innovative tumour specific antibody (Ab)-based immunotherapy (immunocytokine L19-IL2 to “push the accelerator” with or without a checkpoint inhibitor to “release the break”) to form a powerful synergistic immuno-oncology strategy. In HYPOXIMMUNO, I envision to lay the foundation for the next breakthrough in oncology, by improving patient selection and developing highly innovative cancer therapies that target hypoxia, to improve the quality of life and prolong survival of cancer patients, in particular patients with metastatic NSCLC (watch the animation summarising the project: https://youtu.be/oP9Gp4a0b_Q).
Max ERC Funding
2 499 399 €
Duration
Start date: 2016-12-01, End date: 2021-11-30
Project acronym I-MIRNOME
Project Lymphocyte microRNAs in health and disease: Understanding lymphocyte functions through the identification of microRNA target genes and exploiting serum microRNA signatures to monitor immune responses
Researcher (PI) Sergio Abrignani
Host Institution (HI) UNIVERSITA DEGLI STUDI DI MILANO
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary Background: CD4+ T lymphocyte subsets orchestrate immune responses in health and disease. Little is known on control of T cell differentiation exerted by microRNA that affect mRNA translation. The identification of microRNA and their targets that regulate differentiation of T cell subsets may provide new therapeutic targets for immune-mediated diseases. Since microRNA are released in exosomes and circulate in blood, activities of tissue-derived lymphocytes could be assessed by microRNA signatures in the serum. We have defined microRNAs present in resting lymphocyte subsets from peripheral blood and measured lymphocyte-derived microRNAs in the serum. We have also solved important challenges for the identification of microRNA targets, the definition of signatures of activated T cells and their monitoring in the serum, which form the key topics of this application. Advancing State-of-the-Art and objectives: We will identify microRNA of CD4+ T cell subsets purified from inflamed organs and investigate microRNA target network that regulates T cell differentiation. We will exploit this knowledge to profile signatures of in vivo activated T cells and to map genes that could improve understanding of T cell commitment. We will also develop quantitative assays to monitor microRNA signatures in the serum and provide functional evidence of key genes targeted by microRNA which could be targets of immunomodulatory drugs. Significance: This application addresses important challenges at the frontiers of immunology and could lead to significant advances in immunotherapies and diagnostic tools for patients with immune mediated diseases. New ways of identifying microRNA targets and techniques to quantify microRNA signatures in the serum, could be widely applicable in biomedical research.
Summary
Background: CD4+ T lymphocyte subsets orchestrate immune responses in health and disease. Little is known on control of T cell differentiation exerted by microRNA that affect mRNA translation. The identification of microRNA and their targets that regulate differentiation of T cell subsets may provide new therapeutic targets for immune-mediated diseases. Since microRNA are released in exosomes and circulate in blood, activities of tissue-derived lymphocytes could be assessed by microRNA signatures in the serum. We have defined microRNAs present in resting lymphocyte subsets from peripheral blood and measured lymphocyte-derived microRNAs in the serum. We have also solved important challenges for the identification of microRNA targets, the definition of signatures of activated T cells and their monitoring in the serum, which form the key topics of this application. Advancing State-of-the-Art and objectives: We will identify microRNA of CD4+ T cell subsets purified from inflamed organs and investigate microRNA target network that regulates T cell differentiation. We will exploit this knowledge to profile signatures of in vivo activated T cells and to map genes that could improve understanding of T cell commitment. We will also develop quantitative assays to monitor microRNA signatures in the serum and provide functional evidence of key genes targeted by microRNA which could be targets of immunomodulatory drugs. Significance: This application addresses important challenges at the frontiers of immunology and could lead to significant advances in immunotherapies and diagnostic tools for patients with immune mediated diseases. New ways of identifying microRNA targets and techniques to quantify microRNA signatures in the serum, could be widely applicable in biomedical research.
Max ERC Funding
2 496 000 €
Duration
Start date: 2011-06-01, End date: 2016-05-31
Project acronym ImageLink
Project Multiparametric tumor imaging and beyond: Towards understanding in vivo signals
Researcher (PI) Bernd Jürgen Pichler
Host Institution (HI) EBERHARD KARLS UNIVERSITAET TUEBINGEN
Call Details Advanced Grant (AdG), LS7, ERC-2012-ADG_20120314
Summary Non-invasive preclinical and clinical imaging is a powerful tool and has a huge potential, specifically in the realm of oncology. Recently, our laboratory developed a novel multimodality imaging system, which combines positron emission tomography (PET) and magnetic resonance imaging (MRI), yielding temporally and spatially matched data. However, the molecular PET and functional MRI signals are very complex and are often not fully understood. Thus, we will cross-validate the complementary PET/MRI information with proteomics and metabolomics data to gain a better understanding of the in vivo image data and yield finally an accurate holistic tumor profile. The cross-validation will be supported by image-guided accurately dissected tumor substructures. Tumor metabolism, receptor status, hypoxia, perfusion, apoptosis and angiogenesis will be investigated by established PET tracers. In the same imaging session, functional parameters of the tumor, such as perfusion, oxygenation and morphology will be assessed by MRI. Beyond this, novel imaging ligands for senescence, tumor stroma, and fatty acid synthase, which have been recently recognized as emerging key-players in tumor progression and therapy resistance, will be developed. The individual in vivo and in vitro parameters will be fed into a data mining utilizing a computer learning approach with regression and classification methods to detect common patterns and the related pharmacokinetics behind the in vivo imaging parameters. Analysis of the dynamic PET data will be performed by compartment analysis and kinetic modelling. Overall aim is to gain a better understanding of imaging data, provide an accurate holistic in vivo tumor profile to support prognostic parameters for tumor progression and therapy response. Finally, the revealed information will lead to a more accurate selection of imaging biomarkers for diagnosis and therapy control and will provide input for new strategies in tumor-specific tracer development.
Summary
Non-invasive preclinical and clinical imaging is a powerful tool and has a huge potential, specifically in the realm of oncology. Recently, our laboratory developed a novel multimodality imaging system, which combines positron emission tomography (PET) and magnetic resonance imaging (MRI), yielding temporally and spatially matched data. However, the molecular PET and functional MRI signals are very complex and are often not fully understood. Thus, we will cross-validate the complementary PET/MRI information with proteomics and metabolomics data to gain a better understanding of the in vivo image data and yield finally an accurate holistic tumor profile. The cross-validation will be supported by image-guided accurately dissected tumor substructures. Tumor metabolism, receptor status, hypoxia, perfusion, apoptosis and angiogenesis will be investigated by established PET tracers. In the same imaging session, functional parameters of the tumor, such as perfusion, oxygenation and morphology will be assessed by MRI. Beyond this, novel imaging ligands for senescence, tumor stroma, and fatty acid synthase, which have been recently recognized as emerging key-players in tumor progression and therapy resistance, will be developed. The individual in vivo and in vitro parameters will be fed into a data mining utilizing a computer learning approach with regression and classification methods to detect common patterns and the related pharmacokinetics behind the in vivo imaging parameters. Analysis of the dynamic PET data will be performed by compartment analysis and kinetic modelling. Overall aim is to gain a better understanding of imaging data, provide an accurate holistic in vivo tumor profile to support prognostic parameters for tumor progression and therapy response. Finally, the revealed information will lead to a more accurate selection of imaging biomarkers for diagnosis and therapy control and will provide input for new strategies in tumor-specific tracer development.
Max ERC Funding
2 494 800 €
Duration
Start date: 2013-06-01, End date: 2018-05-31
Project acronym IMAGO
Project Imaging regulatory pathways of angiogenesis
Researcher (PI) Michal Neeman
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS7, ERC-2008-AdG
Summary Homeostasis of multicellular tissues relies on accurate match of vascular supply and drain to the needs of the tissue. Multiple pathways are involved in detection, signalling and execution of the required steps involved in organization of blood and lymphatic vessels during embryonic development. Similar mechanisms are utilized for overcoming changes in tissue requirements also in adult tissues and in pathological processes. The goal of this work is to reveal the dynamic forces that shape the blood vessels during angiogenesis. In particular, we would like to explore the impact of interstitial convective flow in dynamic imprinting of growth factor signalling, thereby regulating vascular patterning. Angiogenesis is explored here as an example for a possible general role for interstitial convection of growth factors in determination of the fine spatial patterning of tissue morphogenesis in vertebrates. To achieve this goal, we will develop multi-modality tools for imaging the regulation of vascular patterning. In vivo imaging will then be utilized for mapping vascular patterning in pathological and physiological angiogenesis including tumours, wound repair, the preovulatory ovarian follicle and foetal implantation sites. Whole body optical, CT, ultrasound and MRI will be applied for non-invasive imaging of deep organs. Microscopic morphometric and molecular information will be derived from the macroscopic imaging data, using selective molecular imaging approaches and functional imaging tools with specific pharmacological models that will be developed to account for interstitial convective flow. Intravital two photon microscopy and fluorescence endoscopy will be used for high resolution evaluation of vascular patterning. The evaluation of novel mechanisms for spatial regulation of intercellular growth factor signalling, will allow us to define new potential targets for intervention, and to develop new tools for preclinical and clinical imaging of angiogenesis.
Summary
Homeostasis of multicellular tissues relies on accurate match of vascular supply and drain to the needs of the tissue. Multiple pathways are involved in detection, signalling and execution of the required steps involved in organization of blood and lymphatic vessels during embryonic development. Similar mechanisms are utilized for overcoming changes in tissue requirements also in adult tissues and in pathological processes. The goal of this work is to reveal the dynamic forces that shape the blood vessels during angiogenesis. In particular, we would like to explore the impact of interstitial convective flow in dynamic imprinting of growth factor signalling, thereby regulating vascular patterning. Angiogenesis is explored here as an example for a possible general role for interstitial convection of growth factors in determination of the fine spatial patterning of tissue morphogenesis in vertebrates. To achieve this goal, we will develop multi-modality tools for imaging the regulation of vascular patterning. In vivo imaging will then be utilized for mapping vascular patterning in pathological and physiological angiogenesis including tumours, wound repair, the preovulatory ovarian follicle and foetal implantation sites. Whole body optical, CT, ultrasound and MRI will be applied for non-invasive imaging of deep organs. Microscopic morphometric and molecular information will be derived from the macroscopic imaging data, using selective molecular imaging approaches and functional imaging tools with specific pharmacological models that will be developed to account for interstitial convective flow. Intravital two photon microscopy and fluorescence endoscopy will be used for high resolution evaluation of vascular patterning. The evaluation of novel mechanisms for spatial regulation of intercellular growth factor signalling, will allow us to define new potential targets for intervention, and to develop new tools for preclinical and clinical imaging of angiogenesis.
Max ERC Funding
2 278 344 €
Duration
Start date: 2009-01-01, End date: 2013-12-31
Project acronym IMCCA
Project Innovative Modelling to Optimise Control of Childhood Anaemia across Africa
Researcher (PI) Penelope Vounatsou
Host Institution (HI) SCHWEIZERISCHES TROPEN- UND PUBLIC HEALTH-INSTITUT
Call Details Advanced Grant (AdG), LS7, ERC-2012-ADG_20120314
Summary "Anaemia affects two-third of all preschool-aged children in Africa. Malnutrition, malaria and helminth infections are among the main factors contributing to anaemia in this age group, however their relative contribution across the continent is not well understood. Spatially explicit estimates of anaemia risk are important measures of child morbidity and mortality.
The goal of the project is to develop Bayesian geostatistical methodology for very large, non-stationary data and employ it to (i) determine the relative contribution of malaria, helminth infections and malnutrition on anaemia and severe anaemia burden among pre-school children; (ii) obtain spatially explicit estimates of the risk of anaemia, severe anaemia and number of affected children; (iii) characterize co-endemicity patterns of anaemia, malaria, helminth infection and malnutrition; and (iv) quantify the contribution of severe anaemia to child mortality.
This research will contribute novel statistical methodologies in (i) spatial analysis of very large non-stationary geostatistical data, (ii) variable selection within a non-stationary model for very large geostatistical data, (iii) modelling disease co-endemicity from spatially misaligned surveys arising from independent regression models and (iv) meta-analyses of heterogeneous large spatial data by coupling geostatistical with mathematical transmission models.
Applications of geostatistical methodology will help optimising interventions to combat anaemia by generating (i) the first anaemia risk map and number of affected preschool children across Africa which will guide efficient allocation of nutrient supplements and fortified foods; (ii) anaemia co-endemicity maps and estimates of the relative contribution of anaemia risk factors to design integrated interventions based on local conditions; and (ii) estimates of the anaemia-related mortality across Africa."
Summary
"Anaemia affects two-third of all preschool-aged children in Africa. Malnutrition, malaria and helminth infections are among the main factors contributing to anaemia in this age group, however their relative contribution across the continent is not well understood. Spatially explicit estimates of anaemia risk are important measures of child morbidity and mortality.
The goal of the project is to develop Bayesian geostatistical methodology for very large, non-stationary data and employ it to (i) determine the relative contribution of malaria, helminth infections and malnutrition on anaemia and severe anaemia burden among pre-school children; (ii) obtain spatially explicit estimates of the risk of anaemia, severe anaemia and number of affected children; (iii) characterize co-endemicity patterns of anaemia, malaria, helminth infection and malnutrition; and (iv) quantify the contribution of severe anaemia to child mortality.
This research will contribute novel statistical methodologies in (i) spatial analysis of very large non-stationary geostatistical data, (ii) variable selection within a non-stationary model for very large geostatistical data, (iii) modelling disease co-endemicity from spatially misaligned surveys arising from independent regression models and (iv) meta-analyses of heterogeneous large spatial data by coupling geostatistical with mathematical transmission models.
Applications of geostatistical methodology will help optimising interventions to combat anaemia by generating (i) the first anaemia risk map and number of affected preschool children across Africa which will guide efficient allocation of nutrient supplements and fortified foods; (ii) anaemia co-endemicity maps and estimates of the relative contribution of anaemia risk factors to design integrated interventions based on local conditions; and (ii) estimates of the anaemia-related mortality across Africa."
Max ERC Funding
2 494 800 €
Duration
Start date: 2013-10-01, End date: 2018-09-30