Project acronym AngioGenesHD
Project Epistasis analysis of angiogenes with high cellular definition
Researcher (PI) Rui Miguel Dos Santos Benedito
Host Institution (HI) CENTRO NACIONAL DE INVESTIGACIONESCARDIOVASCULARES CARLOS III (F.S.P.)
Call Details Starting Grant (StG), LS4, ERC-2014-STG
Summary Blood and lymphatic vessels have been the subject of intense investigation due to their important role in cancer development and in cardiovascular diseases. The significant advance in the methods used to modify and analyse gene function have allowed us to obtain a much better understanding of the molecular mechanisms involved in the regulation of the biology of blood vessels. However, there are two key aspects that significantly diminish our capacity to understand the function of gene networks and their intersections in vivo. One is the long time that is usually required to generate a given double mutant vertebrate tissue, and the other is the lack of single-cell genetic and phenotypic resolution. We have recently performed an in vivo comparative transcriptome analysis of highly angiogenic endothelial cells experiencing different VEGF and Notch signalling levels. These are two of the most important molecular mechanisms required for the adequate differentiation, proliferation and sprouting of endothelial cells. Using the information generated from this analysis, the overall aim of the proposed project is to characterize the vascular function of some of the previously identified genes and determine how they functionally interact with these two signalling pathways. We propose to use novel inducible genetic tools that will allow us to generate a spatially and temporally regulated fluorescent cell mosaic matrix for quantitative analysis. This will enable us to analyse with unprecedented speed and resolution the function of several different genes simultaneously, during vascular development, homeostasis or associated diseases. Understanding the genetic epistatic interactions that control the differentiation and behaviour of endothelial cells, in different contexts, and with high cellular definition, has the potential to unveil new mechanisms with high biological and therapeutic relevance.
Summary
Blood and lymphatic vessels have been the subject of intense investigation due to their important role in cancer development and in cardiovascular diseases. The significant advance in the methods used to modify and analyse gene function have allowed us to obtain a much better understanding of the molecular mechanisms involved in the regulation of the biology of blood vessels. However, there are two key aspects that significantly diminish our capacity to understand the function of gene networks and their intersections in vivo. One is the long time that is usually required to generate a given double mutant vertebrate tissue, and the other is the lack of single-cell genetic and phenotypic resolution. We have recently performed an in vivo comparative transcriptome analysis of highly angiogenic endothelial cells experiencing different VEGF and Notch signalling levels. These are two of the most important molecular mechanisms required for the adequate differentiation, proliferation and sprouting of endothelial cells. Using the information generated from this analysis, the overall aim of the proposed project is to characterize the vascular function of some of the previously identified genes and determine how they functionally interact with these two signalling pathways. We propose to use novel inducible genetic tools that will allow us to generate a spatially and temporally regulated fluorescent cell mosaic matrix for quantitative analysis. This will enable us to analyse with unprecedented speed and resolution the function of several different genes simultaneously, during vascular development, homeostasis or associated diseases. Understanding the genetic epistatic interactions that control the differentiation and behaviour of endothelial cells, in different contexts, and with high cellular definition, has the potential to unveil new mechanisms with high biological and therapeutic relevance.
Max ERC Funding
1 481 375 €
Duration
Start date: 2015-03-01, End date: 2020-02-29
Project acronym AP-1-FUN
Project AP-1 (Fos/Jun) Functions in Physiology and Disease
Researcher (PI) Erwin F. Wagner
Host Institution (HI) FUNDACION CENTRO NACIONAL DE INVESTIGACIONES ONCOLOGICAS CARLOS III
Call Details Advanced Grant (AdG), LS4, ERC-2008-AdG
Summary Our research interests lie in breaking new ground in studying mechanism-based functions of AP-1 (Fos/Jun) in vivo with the aim of obtaining a more global perspective on AP-1 in human physiology and disease/cancer. The unresolved issues regarding the AP-1 subunit composition will be tackled biochemically and genetically in various cell types including bone, liver and skin, the primary organs affected by altered AP-1 activity. I plan to utilize the knowledge gained on AP-1 functions in the mouse and transfer it to human disease. The opportunities here lie in exploiting the knowledge of AP-1 target genes and utilizing this information to interfere with pathways involved in normal physiology and disease/cancer. The past investigations revealed that the functions of AP-1 are an essential node at the crossroads between life and death in different cellular systems. I plan to further exploit our findings and concentrate on utilising better mouse models to define these connections. The emphasis will be on identifying molecular signatures and potential treatments in models for cancer, inflammatory and fibrotic diseases. Exploring genetically modified stem cell-based therapies in murine and human cells is an ongoing challenge I would like to meet in the forthcoming years at the CNIO. In addition, the mouse models will be used for mechanism-driven therapeutic strategies and these studies will be undertaken in collaboration with the Experimental Therapeutics Division and the service units such as the tumor bank. The project proposal is divided into 6 Goals (see also Figure 1): Some are a logical continuation based on previous work with completely new aspects (Goal 1-2), some focussing on in depth molecular analyses of disease models with innovative and unconventional concepts, such as for inflammation and cancer, psoriasis and fibrosis (Goal 3-5). A final section is devoted to mouse and human ES cells and their impact for regenerative medicine in bone diseases and cancer.
Summary
Our research interests lie in breaking new ground in studying mechanism-based functions of AP-1 (Fos/Jun) in vivo with the aim of obtaining a more global perspective on AP-1 in human physiology and disease/cancer. The unresolved issues regarding the AP-1 subunit composition will be tackled biochemically and genetically in various cell types including bone, liver and skin, the primary organs affected by altered AP-1 activity. I plan to utilize the knowledge gained on AP-1 functions in the mouse and transfer it to human disease. The opportunities here lie in exploiting the knowledge of AP-1 target genes and utilizing this information to interfere with pathways involved in normal physiology and disease/cancer. The past investigations revealed that the functions of AP-1 are an essential node at the crossroads between life and death in different cellular systems. I plan to further exploit our findings and concentrate on utilising better mouse models to define these connections. The emphasis will be on identifying molecular signatures and potential treatments in models for cancer, inflammatory and fibrotic diseases. Exploring genetically modified stem cell-based therapies in murine and human cells is an ongoing challenge I would like to meet in the forthcoming years at the CNIO. In addition, the mouse models will be used for mechanism-driven therapeutic strategies and these studies will be undertaken in collaboration with the Experimental Therapeutics Division and the service units such as the tumor bank. The project proposal is divided into 6 Goals (see also Figure 1): Some are a logical continuation based on previous work with completely new aspects (Goal 1-2), some focussing on in depth molecular analyses of disease models with innovative and unconventional concepts, such as for inflammation and cancer, psoriasis and fibrosis (Goal 3-5). A final section is devoted to mouse and human ES cells and their impact for regenerative medicine in bone diseases and cancer.
Max ERC Funding
2 500 000 €
Duration
Start date: 2009-11-01, End date: 2015-10-31
Project acronym ARISYS
Project Engineering an artificial immune system with functional components assembled from prokaryotic parts and modules
Researcher (PI) Víctor De Lorenzo Prieto
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Advanced Grant (AdG), LS9, ERC-2012-ADG_20120314
Summary The objective of this project is to overcome current limitations for antibody production that are inherent to the extant immune system of vertebrates. This will be done by creating an all-in-one artificial/synthetic counterpart based exclusively on prokaryotic parts, devices and modules. To this end, ARISYS will exploit design concepts, construction hierarchies and standardization notions that stem from contemporary Synthetic Biology for the assembly and validation of (what we believe is) the most complex artificial biological system ventured thus far. This all-bacterial immune-like system will not only simplify and make affordable the manipulations necessary for antibody generation, but will also permit the application of such binders by themselves or displayed on bacterial cells to biotechnological challenges well beyond therapeutic and health-related uses. The work plan involves the assembly and validation of autonomous functional modules for [i] displaying antibody/affibody (AB) scaffolds attached to the surface of bacterial cells, [ii] conditional diversification of target-binding sequences of the ABs, [iii] contact-dependent activation of gene expression, [iv] reversible bi-stable switches, and [v] clonal selection and amplification of improved binders. These modules composed of stand-alone parts and bearing well defined input/output functions, will be assembled in the genomic chassis of streamlined Escherichia coli and Pseudomonas putida strains. The resulting molecular network will make the ABs expressed and displayed on the cell surface to proceed spontaneously (or at the user's decision) through subsequent cycles of affinity and specificity maturation towards antigens or other targets presented to the bacterial population. In this way, a single, easy-to-handle (albeit heavily engineered) strain will govern all operations that are typically scattered in a multitude of separate methods and apparatuses for AB production.
Summary
The objective of this project is to overcome current limitations for antibody production that are inherent to the extant immune system of vertebrates. This will be done by creating an all-in-one artificial/synthetic counterpart based exclusively on prokaryotic parts, devices and modules. To this end, ARISYS will exploit design concepts, construction hierarchies and standardization notions that stem from contemporary Synthetic Biology for the assembly and validation of (what we believe is) the most complex artificial biological system ventured thus far. This all-bacterial immune-like system will not only simplify and make affordable the manipulations necessary for antibody generation, but will also permit the application of such binders by themselves or displayed on bacterial cells to biotechnological challenges well beyond therapeutic and health-related uses. The work plan involves the assembly and validation of autonomous functional modules for [i] displaying antibody/affibody (AB) scaffolds attached to the surface of bacterial cells, [ii] conditional diversification of target-binding sequences of the ABs, [iii] contact-dependent activation of gene expression, [iv] reversible bi-stable switches, and [v] clonal selection and amplification of improved binders. These modules composed of stand-alone parts and bearing well defined input/output functions, will be assembled in the genomic chassis of streamlined Escherichia coli and Pseudomonas putida strains. The resulting molecular network will make the ABs expressed and displayed on the cell surface to proceed spontaneously (or at the user's decision) through subsequent cycles of affinity and specificity maturation towards antigens or other targets presented to the bacterial population. In this way, a single, easy-to-handle (albeit heavily engineered) strain will govern all operations that are typically scattered in a multitude of separate methods and apparatuses for AB production.
Max ERC Funding
2 422 271 €
Duration
Start date: 2013-05-01, End date: 2019-04-30
Project acronym BacBio
Project Mechanistic and functional studies of Bacillus biofilms assembly on plants, and their impact in sustainable agriculture and food safety
Researcher (PI) Diego Francisco Romero Hinojosa
Host Institution (HI) UNIVERSIDAD DE MALAGA
Call Details Starting Grant (StG), LS9, ERC-2014-STG
Summary Sustainable agriculture is an ambitious concept conceived to improve productivity but minimizing side effects. Why the efficiency of a biocontrol agent is so variable? How can different therapies be efficiently exploited in a combined way to combat microbial diseases? These are questions that need investigation to convey with criteria of sustainability. What I present is an integral proposal aim to study the microbial ecology and specifically bacterial biofilms as a central axis of two differential but likely interconnected scenarios in plant health: i) the beneficial interaction of the biocontrol agent (BCA) Bacillus subtilis, and ii) the non-conventional interaction of the food-borne pathogen Bacillus cereus.
I will start working with B. subtilis, and reasons are: 1) Different isolates are promising BCAs and are commercialized for such purpose, 2) There exist vast information of the genetics circuitries that govern important aspects of B. subtilis physiology as antibiotic production, cell differentiation, and biofilm formation. In parallel I propose to study the way B. cereus, a food-borne pathogenic bacterium interacts with vegetables. I am planning to set up a multidisciplinary approach that will combine genetics, biochemistry, proteomics, cell biology and molecular biology to visualize how these bacterial population interacts, communicates with plants and other microorganisms, or how all these factors trigger or inhibit the developmental program ending in biofilm formation. I am also interested on knowing if structural components of the bacterial extracellular matrix (exopolysaccharides or amyloid proteins) are important for bacterial fitness. If this were the case, I will also investigate which external factors affect their expression and assembly in functional biofilms. The insights get on these studies are committed to impulse our knowledge on microbial ecology and their biotechnological applicability to sustainable agriculture and food safety.
Summary
Sustainable agriculture is an ambitious concept conceived to improve productivity but minimizing side effects. Why the efficiency of a biocontrol agent is so variable? How can different therapies be efficiently exploited in a combined way to combat microbial diseases? These are questions that need investigation to convey with criteria of sustainability. What I present is an integral proposal aim to study the microbial ecology and specifically bacterial biofilms as a central axis of two differential but likely interconnected scenarios in plant health: i) the beneficial interaction of the biocontrol agent (BCA) Bacillus subtilis, and ii) the non-conventional interaction of the food-borne pathogen Bacillus cereus.
I will start working with B. subtilis, and reasons are: 1) Different isolates are promising BCAs and are commercialized for such purpose, 2) There exist vast information of the genetics circuitries that govern important aspects of B. subtilis physiology as antibiotic production, cell differentiation, and biofilm formation. In parallel I propose to study the way B. cereus, a food-borne pathogenic bacterium interacts with vegetables. I am planning to set up a multidisciplinary approach that will combine genetics, biochemistry, proteomics, cell biology and molecular biology to visualize how these bacterial population interacts, communicates with plants and other microorganisms, or how all these factors trigger or inhibit the developmental program ending in biofilm formation. I am also interested on knowing if structural components of the bacterial extracellular matrix (exopolysaccharides or amyloid proteins) are important for bacterial fitness. If this were the case, I will also investigate which external factors affect their expression and assembly in functional biofilms. The insights get on these studies are committed to impulse our knowledge on microbial ecology and their biotechnological applicability to sustainable agriculture and food safety.
Max ERC Funding
1 453 563 €
Duration
Start date: 2015-03-01, End date: 2021-02-28
Project acronym BACCO
Project Bias and Clustering Calculations Optimised: Maximising discovery with galaxy surveys
Researcher (PI) Raúl Esteban ANGULO de la Fuente
Host Institution (HI) FUNDACION CENTRO DE ESTUDIOS DE FISICA DEL COSMOS DE ARAGON
Call Details Starting Grant (StG), PE9, ERC-2016-STG
Summary A new generation of galaxy surveys will soon start measuring the spatial distribution of millions of galaxies over a broad range of redshifts, offering an imminent opportunity to discover new physics. A detailed comparison of these measurements with theoretical models of galaxy clustering may reveal a new fundamental particle, a breakdown of General Relativity, or a hint on the nature of cosmic acceleration. Despite a large progress in the analytic treatment of structure formation in recent years, traditional clustering models still suffer from large uncertainties. This limits cosmological analyses to a very restricted range of scales and statistics, which will be one of the main obstacles to reach a comprehensive exploitation of future surveys.
Here I propose to develop a novel simulation--based approach to predict galaxy clustering. Combining recent advances in computational cosmology, from cosmological N--body calculations to physically-motivated galaxy formation models, I will develop a unified framework to directly predict the position and velocity of individual dark matter structures and galaxies as function of cosmological and astrophysical parameters. In this formulation, galaxy clustering will be a prediction of a set of physical assumptions in a given cosmological setting. The new theoretical framework will be flexible, accurate and fast: it will provide predictions for any clustering statistic, down to scales 100 times smaller than in state-of-the-art perturbation--theory--based models, and in less than 1 minute of CPU time. These advances will enable major improvements in future cosmological constraints, which will significantly increase the overall power of future surveys maximising our potential to discover new physics.
Summary
A new generation of galaxy surveys will soon start measuring the spatial distribution of millions of galaxies over a broad range of redshifts, offering an imminent opportunity to discover new physics. A detailed comparison of these measurements with theoretical models of galaxy clustering may reveal a new fundamental particle, a breakdown of General Relativity, or a hint on the nature of cosmic acceleration. Despite a large progress in the analytic treatment of structure formation in recent years, traditional clustering models still suffer from large uncertainties. This limits cosmological analyses to a very restricted range of scales and statistics, which will be one of the main obstacles to reach a comprehensive exploitation of future surveys.
Here I propose to develop a novel simulation--based approach to predict galaxy clustering. Combining recent advances in computational cosmology, from cosmological N--body calculations to physically-motivated galaxy formation models, I will develop a unified framework to directly predict the position and velocity of individual dark matter structures and galaxies as function of cosmological and astrophysical parameters. In this formulation, galaxy clustering will be a prediction of a set of physical assumptions in a given cosmological setting. The new theoretical framework will be flexible, accurate and fast: it will provide predictions for any clustering statistic, down to scales 100 times smaller than in state-of-the-art perturbation--theory--based models, and in less than 1 minute of CPU time. These advances will enable major improvements in future cosmological constraints, which will significantly increase the overall power of future surveys maximising our potential to discover new physics.
Max ERC Funding
1 484 240 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym BePreSysE
Project Beyond Precision Cosmology: dealing with Systematic Errors
Researcher (PI) Licia VERDE
Host Institution (HI) UNIVERSITAT DE BARCELONA
Call Details Consolidator Grant (CoG), PE9, ERC-2016-COG
Summary Over the past 20 years cosmology has made the transition to a precision science: the standard cosmological model has been established and its parameters are now measured with unprecedented precision. But precision is not enough: accuracy is also crucial. Accuracy accounts for systematic errors which can be both on the observational and on the theory/modelling side (and everywhere in between). While there is a well-defined and developed framework for treating statistical errors, there is no established approach for systematic errors. The next decade will see the era of large surveys; a large coordinated effort of the scientific community in the field is on-going to map the cosmos producing an exponentially growing amount of data. This will shrink the statistical errors, making mitigation and control of systematics of the utmost importance. While there are isolated and targeted efforts to quantify systematic errors and propagate them through all the way to the final results, there is no well-established, self-consistent methodology. To go beyond precision cosmology and reap the benefits of the forthcoming observational program, a systematic approach to systematics is needed. Systematics should be interpreted in the most general sense as shifts between the recovered measured values and true values of physical quantities. I propose to develop a comprehensive approach to tackle systematic errors with the goal to uncover and quantify otherwise unknown differences between the interpretation of a measurement and reality. This will require to fully develop, combine and systematize all approaches proposed so far (many pioneered by the PI), develop new ones to fill the gaps, study and explore their interplay and finally test and validate the procedure. Beyond Precision Cosmology: Dealing with Systematic Errors (BePreSysE) will develop a framework to deal with systematics in forthcoming Cosmological surveys which, could, in principle, be applied beyond Cosmology.
Summary
Over the past 20 years cosmology has made the transition to a precision science: the standard cosmological model has been established and its parameters are now measured with unprecedented precision. But precision is not enough: accuracy is also crucial. Accuracy accounts for systematic errors which can be both on the observational and on the theory/modelling side (and everywhere in between). While there is a well-defined and developed framework for treating statistical errors, there is no established approach for systematic errors. The next decade will see the era of large surveys; a large coordinated effort of the scientific community in the field is on-going to map the cosmos producing an exponentially growing amount of data. This will shrink the statistical errors, making mitigation and control of systematics of the utmost importance. While there are isolated and targeted efforts to quantify systematic errors and propagate them through all the way to the final results, there is no well-established, self-consistent methodology. To go beyond precision cosmology and reap the benefits of the forthcoming observational program, a systematic approach to systematics is needed. Systematics should be interpreted in the most general sense as shifts between the recovered measured values and true values of physical quantities. I propose to develop a comprehensive approach to tackle systematic errors with the goal to uncover and quantify otherwise unknown differences between the interpretation of a measurement and reality. This will require to fully develop, combine and systematize all approaches proposed so far (many pioneered by the PI), develop new ones to fill the gaps, study and explore their interplay and finally test and validate the procedure. Beyond Precision Cosmology: Dealing with Systematic Errors (BePreSysE) will develop a framework to deal with systematics in forthcoming Cosmological surveys which, could, in principle, be applied beyond Cosmology.
Max ERC Funding
1 835 220 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym BIOFORCE
Project Simultaneous multi-pathway engineering in crop plants through combinatorial genetic transformation: Creating nutritionally biofortified cereal grains for food security
Researcher (PI) Paul Christou
Host Institution (HI) UNIVERSIDAD DE LLEIDA
Call Details Advanced Grant (AdG), LS9, ERC-2008-AdG
Summary BIOFORCE has a highly ambitious applied objective: to create transgenic cereal plants that will provide a near-complete micronutrient complement (vitamins A, C, E, folate and essential minerals Ca, Fe, Se and Zn) for malnourished people in the developing world, as well as built-in resistance to insects and parasitic weeds. This in itself represents a striking advance over current efforts to address food insecurity using applied biotechnology in the developing world. We will also address fundamental mechanistic aspects of multi-gene/pathway engineering through transcriptome and metabolome profiling. Fundamental science and applied objectives will be achieved through the application of an exciting novel technology (combinatorial genetic transformation) developed and patented by my research group. This allows the simultaneous transfer of an unlimited number of transgenes into plants followed by library-based selection of plants with appropriate genotypes and phenotypes. All transgenes integrate into one locus ensuring expression stability over multiple generations. This proposal represents a new line of research in my laboratory, founded on incremental advances in the elucidation of transgene integration mechanisms in plants over the past two and a half decades. In addition to scientific issues, BIOFORCE address challenges such as intellectual property, regulatory and biosafety issues and crucially how the fruits of our work will be taken up through philanthropic initiatives in the developing world while creating exploitable opportunities elsewhere. BIOFORCE is comprehensive and it provides a complete package that stands to make an unprecedented contribution to food security in the developing world, while at the same time generating new knowledge to streamline and simplify multiplex gene transfer and the simultaneous modification of multiple complex plant metabolic pathways
Summary
BIOFORCE has a highly ambitious applied objective: to create transgenic cereal plants that will provide a near-complete micronutrient complement (vitamins A, C, E, folate and essential minerals Ca, Fe, Se and Zn) for malnourished people in the developing world, as well as built-in resistance to insects and parasitic weeds. This in itself represents a striking advance over current efforts to address food insecurity using applied biotechnology in the developing world. We will also address fundamental mechanistic aspects of multi-gene/pathway engineering through transcriptome and metabolome profiling. Fundamental science and applied objectives will be achieved through the application of an exciting novel technology (combinatorial genetic transformation) developed and patented by my research group. This allows the simultaneous transfer of an unlimited number of transgenes into plants followed by library-based selection of plants with appropriate genotypes and phenotypes. All transgenes integrate into one locus ensuring expression stability over multiple generations. This proposal represents a new line of research in my laboratory, founded on incremental advances in the elucidation of transgene integration mechanisms in plants over the past two and a half decades. In addition to scientific issues, BIOFORCE address challenges such as intellectual property, regulatory and biosafety issues and crucially how the fruits of our work will be taken up through philanthropic initiatives in the developing world while creating exploitable opportunities elsewhere. BIOFORCE is comprehensive and it provides a complete package that stands to make an unprecedented contribution to food security in the developing world, while at the same time generating new knowledge to streamline and simplify multiplex gene transfer and the simultaneous modification of multiple complex plant metabolic pathways
Max ERC Funding
2 290 046 €
Duration
Start date: 2009-04-01, End date: 2014-03-31
Project acronym BLOODCELLSCROSSTALK
Project The Crosstalk Between Red And White Blood Cells: The Case Of Fish
Researcher (PI) Maria del Mar Ortega-Villaizan Romo
Host Institution (HI) UNIVERSIDAD MIGUEL HERNANDEZ DE ELCHE
Call Details Starting Grant (StG), LS9, ERC-2014-STG
Summary Fish are the phylogenetically oldest vertebrate group with an immune system with clear similarities to the immune system of mammals. However, it is an actual matter of fact that the current knowledge of the fish immune system seems to lack the key piece to complete the puzzle.
In 1953 Nelson described a new role of human red blood cells (RBCs) which would go beyond the simple transport of O2 to the tissues. This new role, involved in the defence against microbes, described the antibody and complement-dependent binding of microbial immune complexes to RBCs. Regardless of the importance of this finding in the field of microbial infection, this phenomenon has been poorly evaluated. Just recently, a set of biological processes relevant to immunity have been described in the RBCs of a diverse group of organisms, which include: pathogen recognition, pathogen binding and clearance and cytokines production. Furthermore, it has been demonstrated that nucleated erythrocytes from fish and avian species develop specific responses to different pathogen associated molecular patterns and produce soluble factors that modulate leukocyte activity.
In the light of these pieces of evidences, and in an attempt to improve the knowledge of the immune mechanism(s) responsible for fish protection against viral infections, we raised the question: could nucleated fish erythrocytes be the key mediators of the antiviral responses? To answer this question we decided to focus our project on the evaluation of the crosstalk between red and white blood cells in the scenario of fish viral infections and prophylaxis. For that a working model composed of the rainbow trout and the viral haemorrhagic septicaemia virus (VHSV) was chosen, being the objectives of the project to evaluate: i) the implication trout RBCs (tRBCs) in the clearance of VHSV, and ii) the involvement of tRBCs in the blood transportation of the glycoprotein G of VHSV (GVHSV), the antigen encoded by the DNA vaccine.
Summary
Fish are the phylogenetically oldest vertebrate group with an immune system with clear similarities to the immune system of mammals. However, it is an actual matter of fact that the current knowledge of the fish immune system seems to lack the key piece to complete the puzzle.
In 1953 Nelson described a new role of human red blood cells (RBCs) which would go beyond the simple transport of O2 to the tissues. This new role, involved in the defence against microbes, described the antibody and complement-dependent binding of microbial immune complexes to RBCs. Regardless of the importance of this finding in the field of microbial infection, this phenomenon has been poorly evaluated. Just recently, a set of biological processes relevant to immunity have been described in the RBCs of a diverse group of organisms, which include: pathogen recognition, pathogen binding and clearance and cytokines production. Furthermore, it has been demonstrated that nucleated erythrocytes from fish and avian species develop specific responses to different pathogen associated molecular patterns and produce soluble factors that modulate leukocyte activity.
In the light of these pieces of evidences, and in an attempt to improve the knowledge of the immune mechanism(s) responsible for fish protection against viral infections, we raised the question: could nucleated fish erythrocytes be the key mediators of the antiviral responses? To answer this question we decided to focus our project on the evaluation of the crosstalk between red and white blood cells in the scenario of fish viral infections and prophylaxis. For that a working model composed of the rainbow trout and the viral haemorrhagic septicaemia virus (VHSV) was chosen, being the objectives of the project to evaluate: i) the implication trout RBCs (tRBCs) in the clearance of VHSV, and ii) the involvement of tRBCs in the blood transportation of the glycoprotein G of VHSV (GVHSV), the antigen encoded by the DNA vaccine.
Max ERC Funding
1 823 250 €
Duration
Start date: 2015-04-01, End date: 2020-03-31
Project acronym CAMAP
Project CAMAP: Computer Aided Modeling for Astrophysical Plasmas
Researcher (PI) Miguel-Ángel Aloy-Torás
Host Institution (HI) UNIVERSITAT DE VALENCIA
Call Details Starting Grant (StG), PE9, ERC-2010-StG_20091028
Summary This project will be aimed at obtaining a deeper insight into the physical processes taking place in astrophysical magnetized plasmas. To study these scenarios I will employ different numerical codes as virtual tools that enable me to experiment on computers (virtual labs) with distinct initial and boundary conditions. Among the kind of sources I am interested to consider, I outline the following: Gamma-Ray Bursts (GRBs), extragalactic jets from Active Galactic Nuclei (AGN), magnetars and collapsing stellar cores. A number of important questions are still open regarding the fundamental properties of these astrophysical sources (e.g., collimation, acceleration mechanism, composition, high-energy emission, gravitational wave signature). Additionally, there are analytical issues on the formalism in relativistic dynamics not resolved yet, e.g., the covariant extension of resistive magnetohydrodynamics (MHD). All these problems are so complex that only a computational approach is feasible. I plan to study them by means of relativistic and Newtonian MHD numerical simulations. A principal focus of the project will be to assess the relevance of magnetic fields in the generation, collimation and ulterior propagation of relativistic jets from the GRB progenitors and from AGNs. More generally, I will pursue the goal of understanding the process of amplification of seed magnetic fields until they become dynamically relevant, e.g., using semi-global and local simulations of representative boxes of collapsed stellar cores. A big emphasis will be put on including all the relevant microphysics (e.g. neutrino physics), non-ideal effects (particularly, reconnection physics) and energy transport due to neutrinos and photons to account for the relevant processes in the former systems. A milestone of this project will be to end up with a numerical tool that enables us to deal with General Relativistic Radiation Magnetohydrodynamics problems in Astrophysics.
Summary
This project will be aimed at obtaining a deeper insight into the physical processes taking place in astrophysical magnetized plasmas. To study these scenarios I will employ different numerical codes as virtual tools that enable me to experiment on computers (virtual labs) with distinct initial and boundary conditions. Among the kind of sources I am interested to consider, I outline the following: Gamma-Ray Bursts (GRBs), extragalactic jets from Active Galactic Nuclei (AGN), magnetars and collapsing stellar cores. A number of important questions are still open regarding the fundamental properties of these astrophysical sources (e.g., collimation, acceleration mechanism, composition, high-energy emission, gravitational wave signature). Additionally, there are analytical issues on the formalism in relativistic dynamics not resolved yet, e.g., the covariant extension of resistive magnetohydrodynamics (MHD). All these problems are so complex that only a computational approach is feasible. I plan to study them by means of relativistic and Newtonian MHD numerical simulations. A principal focus of the project will be to assess the relevance of magnetic fields in the generation, collimation and ulterior propagation of relativistic jets from the GRB progenitors and from AGNs. More generally, I will pursue the goal of understanding the process of amplification of seed magnetic fields until they become dynamically relevant, e.g., using semi-global and local simulations of representative boxes of collapsed stellar cores. A big emphasis will be put on including all the relevant microphysics (e.g. neutrino physics), non-ideal effects (particularly, reconnection physics) and energy transport due to neutrinos and photons to account for the relevant processes in the former systems. A milestone of this project will be to end up with a numerical tool that enables us to deal with General Relativistic Radiation Magnetohydrodynamics problems in Astrophysics.
Max ERC Funding
1 497 000 €
Duration
Start date: 2011-03-01, End date: 2017-02-28
Project acronym CancerADAPT
Project Targeting the adaptive capacity of prostate cancer through the manipulation of transcriptional and metabolic traits
Researcher (PI) Arkaitz CARRACEDO PEREZ
Host Institution (HI) ASOCIACION CENTRO DE INVESTIGACION COOPERATIVA EN BIOCIENCIAS
Call Details Consolidator Grant (CoG), LS4, ERC-2018-COG
Summary The composition and molecular features of tumours vary during the course of the disease, and the selection pressure imposed by the environment is a central component in this process. Evolutionary principles have been exploited to explain the genomic aberrations in cancer. However, the phenotypic changes underlying disease progression remain poorly understood. In the past years, I have contributed to identify and characterise the therapeutic implications underlying metabolic alterations that are intrinsic to primary tumours or metastasis. In CancerADAPT I postulate that cancer cells rely on adaptive transcriptional & metabolic mechanisms [converging on a Metabolic Phenotype] in order to rapidly succeed in their establishment in new microenvironments along disease progression. I aim to predict the molecular cues that govern the adaptive properties in prostate cancer (PCa), one of the most commonly diagnosed cancers in men and an important source of cancer-related deaths. I will exploit single cell RNASeq, spatial transcriptomics and multiregional OMICs in order to identify the transcriptional and metabolic diversity within tumours and along disease progression. I will complement experimental strategies with computational analyses that identify and classify the predicted adaptation strategies of PCa cells in response to variations in the tumour microenvironment. Metabolic phenotypes postulated to sustain PCa adaptability will be functionally and mechanistically deconstructed. We will identify therapeutic strategies emanating from these results through in silico methodologies and small molecule high-throughput screening, and evaluate their potential to hamper the adaptability of tumour cells in vitro and in vivo, in two specific aspects: metastasis and therapy response. CancerADAPT will generate fundamental understanding on how cancer cells adapt in our organism, in turn leading to therapeutic strategies that increase the efficacy of current treatments.
Summary
The composition and molecular features of tumours vary during the course of the disease, and the selection pressure imposed by the environment is a central component in this process. Evolutionary principles have been exploited to explain the genomic aberrations in cancer. However, the phenotypic changes underlying disease progression remain poorly understood. In the past years, I have contributed to identify and characterise the therapeutic implications underlying metabolic alterations that are intrinsic to primary tumours or metastasis. In CancerADAPT I postulate that cancer cells rely on adaptive transcriptional & metabolic mechanisms [converging on a Metabolic Phenotype] in order to rapidly succeed in their establishment in new microenvironments along disease progression. I aim to predict the molecular cues that govern the adaptive properties in prostate cancer (PCa), one of the most commonly diagnosed cancers in men and an important source of cancer-related deaths. I will exploit single cell RNASeq, spatial transcriptomics and multiregional OMICs in order to identify the transcriptional and metabolic diversity within tumours and along disease progression. I will complement experimental strategies with computational analyses that identify and classify the predicted adaptation strategies of PCa cells in response to variations in the tumour microenvironment. Metabolic phenotypes postulated to sustain PCa adaptability will be functionally and mechanistically deconstructed. We will identify therapeutic strategies emanating from these results through in silico methodologies and small molecule high-throughput screening, and evaluate their potential to hamper the adaptability of tumour cells in vitro and in vivo, in two specific aspects: metastasis and therapy response. CancerADAPT will generate fundamental understanding on how cancer cells adapt in our organism, in turn leading to therapeutic strategies that increase the efficacy of current treatments.
Max ERC Funding
1 999 882 €
Duration
Start date: 2019-11-01, End date: 2024-10-31
Project acronym CANCERMETAB
Project Metabolic requirements for prostate cancer cell fitness
Researcher (PI) Arkaitz Carracedo Perez
Host Institution (HI) ASOCIACION CENTRO DE INVESTIGACION COOPERATIVA EN BIOCIENCIAS
Call Details Starting Grant (StG), LS4, ERC-2013-StG
Summary The actual view of cellular transformation and cancer progression supports the notion that cancer cells must undergo metabolic reprogramming in order to survive in a hostile environment. This field has experienced a renaissance in recent years, with the discovery of cancer genes regulating metabolic homeostasis, in turn being accepted as an emergent hallmark of cancer. Prostate cancer presents one of the highest incidences in men mostly in developed societies and exhibits a significant association with lifestyle environmental factors. Prostate cancer recurrence is thought to rely on a subpopulation of cancer cells with low-androgen requirements, high self-renewal potential and multidrug resistance, defined as cancer-initiating cells. However, whether this cancer cell fraction presents genuine metabolic properties that can be therapeutically relevant remains undefined. In CancerMetab, we aim to understand the potential benefit of monitoring and manipulating metabolism for prostate cancer prevention, detection and therapy. My group will carry out a multidisciplinary strategy, comprising cellular systems, genetic mouse models of prostate cancer, human epidemiological and clinical studies and bioinformatic analysis. The singularity of this proposal stems from the approach to the three key aspects that we propose to study. For prostate cancer prevention, we will use our faithful mouse model of prostate cancer to shed light on the contribution of obesity to prostate cancer. For prostate cancer detection, we will overcome the consistency issues of previously reported metabolic biomarkers by adding robustness to the human studies with mouse data integration. For prostate cancer therapy, we will focus on a cell population for which the metabolic requirements and the potential of targeting them for therapy have been overlooked to date, that is the prostate cancer-initiating cell compartment.
Summary
The actual view of cellular transformation and cancer progression supports the notion that cancer cells must undergo metabolic reprogramming in order to survive in a hostile environment. This field has experienced a renaissance in recent years, with the discovery of cancer genes regulating metabolic homeostasis, in turn being accepted as an emergent hallmark of cancer. Prostate cancer presents one of the highest incidences in men mostly in developed societies and exhibits a significant association with lifestyle environmental factors. Prostate cancer recurrence is thought to rely on a subpopulation of cancer cells with low-androgen requirements, high self-renewal potential and multidrug resistance, defined as cancer-initiating cells. However, whether this cancer cell fraction presents genuine metabolic properties that can be therapeutically relevant remains undefined. In CancerMetab, we aim to understand the potential benefit of monitoring and manipulating metabolism for prostate cancer prevention, detection and therapy. My group will carry out a multidisciplinary strategy, comprising cellular systems, genetic mouse models of prostate cancer, human epidemiological and clinical studies and bioinformatic analysis. The singularity of this proposal stems from the approach to the three key aspects that we propose to study. For prostate cancer prevention, we will use our faithful mouse model of prostate cancer to shed light on the contribution of obesity to prostate cancer. For prostate cancer detection, we will overcome the consistency issues of previously reported metabolic biomarkers by adding robustness to the human studies with mouse data integration. For prostate cancer therapy, we will focus on a cell population for which the metabolic requirements and the potential of targeting them for therapy have been overlooked to date, that is the prostate cancer-initiating cell compartment.
Max ERC Funding
1 498 686 €
Duration
Start date: 2013-11-01, End date: 2019-10-31
Project acronym CELLPLASTICITY
Project New Frontiers in Cellular Reprogramming: Exploiting Cellular Plasticity
Researcher (PI) Manuel SERRANO MARUGAN
Host Institution (HI) FUNDACIO INSTITUT DE RECERCA BIOMEDICA (IRB BARCELONA)
Call Details Advanced Grant (AdG), LS4, ERC-2014-ADG
Summary "Our research group has worked over the years at the interface between cancer and ageing, with a strong emphasis on mouse models. More recently, we became interested in cellular reprogramming because we hypothesized that understanding cellular plasticity could yield new insights into cancer and ageing. Indeed, during the previous ERC Advanced Grant, we made relevant contributions to the fields of cellular reprogramming (Nature 2013), cellular senescence (Cell 2013), cancer (Cancer Cell 2012), and ageing (Cell Metabolism 2012). Now, we take advantage of our diverse background and integrate the above processes. Our unifying hypothesis is that cellular plasticity lies at the basis of tissue regeneration (“adaptive cellular plasticity”), as well as at the origin of cancer (“maladaptive gain of cellular plasticity”) and ageing (“maladaptive loss of cellular plasticity”). A key experimental system will be our “reprogrammable mice” (with inducible expression of the four Yamanaka factors), which we regard as a tool to induce cellular plasticity in vivo. The project is divided as follows: Objective #1 – Cellular plasticity and cancer: role of tumour suppressors in in vivo de-differentiation and reprogramming / impact of transient de-differentiation on tumour initiation / lineage tracing of Oct4 to determine whether a transient pluripotent-state occurs during cancer. Objective #2 – Cellular plasticity in tissue regeneration and ageing: impact of transient de-differentiation on tissue regeneration / contribution of the damage-induced microenvironment to tissue regeneration / impact of transient de-differentiation on ageing. Objective #3: New frontiers in cellular plasticity: chemical manipulation of cellular plasticity in vivo / new states of pluripotency / characterization of in vivo induced pluripotency and its unique properties. We anticipate that the completion of this project will yield new fundamental insights into cancer, regeneration and ageing."
Summary
"Our research group has worked over the years at the interface between cancer and ageing, with a strong emphasis on mouse models. More recently, we became interested in cellular reprogramming because we hypothesized that understanding cellular plasticity could yield new insights into cancer and ageing. Indeed, during the previous ERC Advanced Grant, we made relevant contributions to the fields of cellular reprogramming (Nature 2013), cellular senescence (Cell 2013), cancer (Cancer Cell 2012), and ageing (Cell Metabolism 2012). Now, we take advantage of our diverse background and integrate the above processes. Our unifying hypothesis is that cellular plasticity lies at the basis of tissue regeneration (“adaptive cellular plasticity”), as well as at the origin of cancer (“maladaptive gain of cellular plasticity”) and ageing (“maladaptive loss of cellular plasticity”). A key experimental system will be our “reprogrammable mice” (with inducible expression of the four Yamanaka factors), which we regard as a tool to induce cellular plasticity in vivo. The project is divided as follows: Objective #1 – Cellular plasticity and cancer: role of tumour suppressors in in vivo de-differentiation and reprogramming / impact of transient de-differentiation on tumour initiation / lineage tracing of Oct4 to determine whether a transient pluripotent-state occurs during cancer. Objective #2 – Cellular plasticity in tissue regeneration and ageing: impact of transient de-differentiation on tissue regeneration / contribution of the damage-induced microenvironment to tissue regeneration / impact of transient de-differentiation on ageing. Objective #3: New frontiers in cellular plasticity: chemical manipulation of cellular plasticity in vivo / new states of pluripotency / characterization of in vivo induced pluripotency and its unique properties. We anticipate that the completion of this project will yield new fundamental insights into cancer, regeneration and ageing."
Max ERC Funding
2 488 850 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym DeAge
Project Deconstructing Ageing: from molecular mechanisms to intervention strategies
Researcher (PI) Carlos LOPEZ OTIN
Host Institution (HI) UNIVERSIDAD DE OVIEDO
Call Details Advanced Grant (AdG), LS4, ERC-2016-ADG
Summary Over many years, our research group has explored the complex relationship between cancer and ageing. As part of this work, we have generated mouse models of protease deficiency which are protected from cancer but exhibit accelerated ageing. Further studies with these mice have allowed us to unveil novel mechanisms of both normal and pathological ageing, to discover two new human progeroid syndromes, and to develop therapies for the Hutchinson-Gilford progeria syndrome, now in clinical trials. We have also integrated data from many laboratories to first define The hallmarks of ageing and the current possibilities for Metabolic control of longevity. Now, we propose to leverage our extensive experience in this field to further explore the relative relevance of cell-intrinsic and -extrinsic mechanisms of ageing. Our central hypothesis is that ageing derives from the combination of both systemic and cell-autonomous deficiencies which lead to the characteristic loss of fitness associated with this process. Accordingly, it is necessary to integrate multiple approaches to understand the mechanisms underlying ageing. This integrative and multidisciplinary project is organized around three major aims: 1) to characterize critical cell-intrinsic alterations which drive ageing; 2) to investigate ageing as a systemic process; and 3) to design intervention strategies aimed at expanding longevity. To fully address these objectives, we will use both hypothesis-driven and unbiased approaches, including next-generation sequencing, genome editing, and cell reprogramming. We will also perform in vivo experiments with mouse models of premature ageing, genomic and metagenomic studies with short- and long-lived organisms, and functional analyses with human samples from both progeria patients and centenarians. The information derived from this project will provide new insights into the molecular mechanisms of ageing and may lead to discover new opportunities to extend human healthspan.
Summary
Over many years, our research group has explored the complex relationship between cancer and ageing. As part of this work, we have generated mouse models of protease deficiency which are protected from cancer but exhibit accelerated ageing. Further studies with these mice have allowed us to unveil novel mechanisms of both normal and pathological ageing, to discover two new human progeroid syndromes, and to develop therapies for the Hutchinson-Gilford progeria syndrome, now in clinical trials. We have also integrated data from many laboratories to first define The hallmarks of ageing and the current possibilities for Metabolic control of longevity. Now, we propose to leverage our extensive experience in this field to further explore the relative relevance of cell-intrinsic and -extrinsic mechanisms of ageing. Our central hypothesis is that ageing derives from the combination of both systemic and cell-autonomous deficiencies which lead to the characteristic loss of fitness associated with this process. Accordingly, it is necessary to integrate multiple approaches to understand the mechanisms underlying ageing. This integrative and multidisciplinary project is organized around three major aims: 1) to characterize critical cell-intrinsic alterations which drive ageing; 2) to investigate ageing as a systemic process; and 3) to design intervention strategies aimed at expanding longevity. To fully address these objectives, we will use both hypothesis-driven and unbiased approaches, including next-generation sequencing, genome editing, and cell reprogramming. We will also perform in vivo experiments with mouse models of premature ageing, genomic and metagenomic studies with short- and long-lived organisms, and functional analyses with human samples from both progeria patients and centenarians. The information derived from this project will provide new insights into the molecular mechanisms of ageing and may lead to discover new opportunities to extend human healthspan.
Max ERC Funding
2 456 250 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym DecodeDiabetes
Project Expanding the genetic etiological and diagnostic spectrum of monogenic diabetes mellitus
Researcher (PI) Jorge FERRER
Host Institution (HI) FUNDACIO CENTRE DE REGULACIO GENOMICA
Call Details Advanced Grant (AdG), LS4, ERC-2017-ADG
Summary Whole genome sequencing is quickly becoming a routine clinical instrument. However, our ability to decipher DNA variants is still largely limited to protein-coding exons, which comprise 1% of the genome. Most known Mendelian mutations are in exons, yet genetic testing still fails to show causal coding mutations in more than 50% of well-characterized Mendelian disorders. This defines a pressing need to interpret noncoding genome sequences, and to establish the role of noncoding mutations in Mendelian disease.
A recent case study harnessed whole genome sequencing, epigenomics, and functional genomics to show that mutations in an enhancer cause most cases of neonatal diabetes due to pancreas agenesis. This example raises major questions: (i) what is the overall impact of penetrant regulatory mutations in human diabetes? (ii) do regulatory mutations cause distinct forms of diabetes? (iii) more generally, can we develop a strategy to systematically tackle regulatory variation in Mendelian disease?
The current project will address these questions with unique resources. First, we have created epigenomic and functional perturbation resources to interpret the regulatory genome in embryonic pancreas and adult pancreatic islets. Second, we have collected an unprecedented international cohort of patients with a phenotype consistent with monogenic diabetes, yet lacking mutations in known gene culprits after genetic testing, and therefore with increased likelihood of harboring noncoding mutations. Third, we have developed a prototype platform to sequence regulatory mutations in a large number of patients.
These resources will be combined with innovative strategies to uncover causal enhancer mutations underlying Mendelian diabetes. If successful, this project will expand the diagnostic spectrum of diabetes, it will discover new genetic regulators of diabetes-relevant networks, and will provide a framework to understand regulatory variation in Mendelian disease.
Summary
Whole genome sequencing is quickly becoming a routine clinical instrument. However, our ability to decipher DNA variants is still largely limited to protein-coding exons, which comprise 1% of the genome. Most known Mendelian mutations are in exons, yet genetic testing still fails to show causal coding mutations in more than 50% of well-characterized Mendelian disorders. This defines a pressing need to interpret noncoding genome sequences, and to establish the role of noncoding mutations in Mendelian disease.
A recent case study harnessed whole genome sequencing, epigenomics, and functional genomics to show that mutations in an enhancer cause most cases of neonatal diabetes due to pancreas agenesis. This example raises major questions: (i) what is the overall impact of penetrant regulatory mutations in human diabetes? (ii) do regulatory mutations cause distinct forms of diabetes? (iii) more generally, can we develop a strategy to systematically tackle regulatory variation in Mendelian disease?
The current project will address these questions with unique resources. First, we have created epigenomic and functional perturbation resources to interpret the regulatory genome in embryonic pancreas and adult pancreatic islets. Second, we have collected an unprecedented international cohort of patients with a phenotype consistent with monogenic diabetes, yet lacking mutations in known gene culprits after genetic testing, and therefore with increased likelihood of harboring noncoding mutations. Third, we have developed a prototype platform to sequence regulatory mutations in a large number of patients.
These resources will be combined with innovative strategies to uncover causal enhancer mutations underlying Mendelian diabetes. If successful, this project will expand the diagnostic spectrum of diabetes, it will discover new genetic regulators of diabetes-relevant networks, and will provide a framework to understand regulatory variation in Mendelian disease.
Max ERC Funding
2 243 746 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym editCRC
Project A genome editing-based approach to study the stem cell hierarchy of human colorectal cancers
Researcher (PI) Eduardo Batlle Gómez
Host Institution (HI) FUNDACIO INSTITUT DE RECERCA BIOMEDICA (IRB BARCELONA)
Call Details Advanced Grant (AdG), LS4, ERC-2013-ADG
Summary A hallmark of cancer is tumor cell heterogeneity, which results from combinations of multiple genetic and epigenetic alterations within an individual tumor. In contrast, we have recently discovered that most human colorectal cancers (CRCs) are composed of mixtures of phenotypically distinct tumor cells organized into a stem cell hierarchy that displays a striking resemblance to the healthy colonic epithelium. We showed that long-term regeneration potential of tumor cells is largely influenced by the position that they occupy within the tumor's hierarchy. To analyze the organization of CRCs without the constraints imposed by tumor cell transplantation experiments, we have developed a method that allows for the first time tracking and manipulating the fate of specific cell populations in whole human tumors. This technology is based on editing the genomes of primary human CRCs cultured in the form of tumor organoids using Zinc-Finger Nucleases to knock-in either lineage tracing or cell ablation alleles in genes that define colorectal cancer stem cells (CRC-SCs) or differentiated-like tumor cells. Edited tumor organoids generate CRCs in mice that reproduce the tumor of origin while carrying the desired genetic modifications. This technological advance opens the gate to perform classical genetic and developmental analysis in human tumors. We will exploit this advantage to address fundamental questions about the cell heterogeneity and organization of human CRCs that cannot be tackled through currently existing experimental approaches such as: Are CRC-SCs the only tumor cell population with long term regenerating potential? Can we cure CRC with anti-CRC-SC specific therapies? Will tumor cell plasticity contribute to the regeneration of the CRC-SC pool after therapy? Do quiescent-SCs regenerate CRC tumors after standard chemotherapy? Can we identify these cells? How do common genetic alterations in CRC influence the CRC hierarchy? Do they affect the stem cell phenotype?
Summary
A hallmark of cancer is tumor cell heterogeneity, which results from combinations of multiple genetic and epigenetic alterations within an individual tumor. In contrast, we have recently discovered that most human colorectal cancers (CRCs) are composed of mixtures of phenotypically distinct tumor cells organized into a stem cell hierarchy that displays a striking resemblance to the healthy colonic epithelium. We showed that long-term regeneration potential of tumor cells is largely influenced by the position that they occupy within the tumor's hierarchy. To analyze the organization of CRCs without the constraints imposed by tumor cell transplantation experiments, we have developed a method that allows for the first time tracking and manipulating the fate of specific cell populations in whole human tumors. This technology is based on editing the genomes of primary human CRCs cultured in the form of tumor organoids using Zinc-Finger Nucleases to knock-in either lineage tracing or cell ablation alleles in genes that define colorectal cancer stem cells (CRC-SCs) or differentiated-like tumor cells. Edited tumor organoids generate CRCs in mice that reproduce the tumor of origin while carrying the desired genetic modifications. This technological advance opens the gate to perform classical genetic and developmental analysis in human tumors. We will exploit this advantage to address fundamental questions about the cell heterogeneity and organization of human CRCs that cannot be tackled through currently existing experimental approaches such as: Are CRC-SCs the only tumor cell population with long term regenerating potential? Can we cure CRC with anti-CRC-SC specific therapies? Will tumor cell plasticity contribute to the regeneration of the CRC-SC pool after therapy? Do quiescent-SCs regenerate CRC tumors after standard chemotherapy? Can we identify these cells? How do common genetic alterations in CRC influence the CRC hierarchy? Do they affect the stem cell phenotype?
Max ERC Funding
2 499 405 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym ELONGAN
Project Gene editing and in vitro approaches to understand conceptus elongation in ungulates
Researcher (PI) Pablo BERMEJO-ÁLVAREZ
Host Institution (HI) INSTITUTO NACIONAL DE INVESTIGACION Y TECNOLOGIA AGRARIA Y ALIMENTARIA OA MP
Call Details Starting Grant (StG), LS9, ERC-2017-STG
Summary In contrast to human or rodent embryos, ungulate embryos do not implant into the uterus right after blastocyst hatching. Before implantation, the hatched ungulate blastocyst must undergo dramatic morphological changes characterized by cell differentiation, proliferation and migration processes leading to the development of extra-embryonic membranes, the appearance of a flat embryonic disc and gastrulation. This prolonged preimplantation development is termed conceptus elongation and deficiencies on this process constitute the most frequent cause of reproductive failures in ungulates, including the 4 most relevant mammalian livestock species in Europe. The purpose of this project is to elucidate the factors involved in conceptus elongation by gene editing and in vitro culture approaches. A first objective will be to identify key genes involved in differentiation processes by RNA-seq analysis of different embryo derivatives from bovine conceptuses at different developmental stages. Subsequently, the function of some of the genes identified as well as others known to play a crucial role in mouse development or putatively involved in embryo-maternal interactions will be assessed. For this aim, bovine embryos in which a candidate gene has been ablated (KO) will be generated by CRISPR and transferred to recipient females to assess in vivo the function of such particular gene on conceptus development. A second set of experiments pursue the development of an in vitro system for conceptus elongation that would bypass the requirement for in vivo experiments. For this aim we will perform metabolomics and proteomics analyses of bovine uterine fluid at different stages and will use these data to rationally develop a culture system able to sustain conceptus development. The knowledge generated by this project will serve to develop strategies to enhance farming profitability by reducing embryonic loss and to understand Developmental Biology questions unanswered by the mouse model.
Summary
In contrast to human or rodent embryos, ungulate embryos do not implant into the uterus right after blastocyst hatching. Before implantation, the hatched ungulate blastocyst must undergo dramatic morphological changes characterized by cell differentiation, proliferation and migration processes leading to the development of extra-embryonic membranes, the appearance of a flat embryonic disc and gastrulation. This prolonged preimplantation development is termed conceptus elongation and deficiencies on this process constitute the most frequent cause of reproductive failures in ungulates, including the 4 most relevant mammalian livestock species in Europe. The purpose of this project is to elucidate the factors involved in conceptus elongation by gene editing and in vitro culture approaches. A first objective will be to identify key genes involved in differentiation processes by RNA-seq analysis of different embryo derivatives from bovine conceptuses at different developmental stages. Subsequently, the function of some of the genes identified as well as others known to play a crucial role in mouse development or putatively involved in embryo-maternal interactions will be assessed. For this aim, bovine embryos in which a candidate gene has been ablated (KO) will be generated by CRISPR and transferred to recipient females to assess in vivo the function of such particular gene on conceptus development. A second set of experiments pursue the development of an in vitro system for conceptus elongation that would bypass the requirement for in vivo experiments. For this aim we will perform metabolomics and proteomics analyses of bovine uterine fluid at different stages and will use these data to rationally develop a culture system able to sustain conceptus development. The knowledge generated by this project will serve to develop strategies to enhance farming profitability by reducing embryonic loss and to understand Developmental Biology questions unanswered by the mouse model.
Max ERC Funding
1 480 880 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym EndoMitTalk
Project Endolysosomal-mitochondria crosstalk in cell and organism homeostasis
Researcher (PI) María MITTELBRUM
Host Institution (HI) UNIVERSIDAD AUTONOMA DE MADRID
Call Details Starting Grant (StG), LS4, ERC-2016-STG
Summary For many years, mitochondria were viewed as semiautonomous organelles, required only for cellular energetics. This view has been displaced by the concept that mitochondria are fully integrated into the life of the cell and that mitochondrial function and stress response rapidly affect other organelles, and even other tissues. A recent discovery from my lab demonstrated that mitochondrial metabolism regulates lysosomal degradation (Cell Metabolism, 2015), thus opening the way to investigate the mechanism behind communication between these organelles and its consequences for homeostasis. With this proposal, we want to assess how mitochondrial crosstalk with endolysosomal compartment controls cellular homeostasis and how mitochondrial dysfunction in certain tissues may account for systemic effects on the rest of the organism. EndoMitTalk will deliver significant breakthroughs on (1) the molecular mediators of endolysosomal-mitochondria communication, and how deregulation of this crosstalk alters cellular (2), and organism homeostasis (3). Our central goals are: 1a,b. To identify metabolic and physical connections mediating endolysosomal-mitochondria crosstalk; 2a. To decode the consequences of altered interorganelle communication in cellular homeostasis 2b. To study the therapeutic potential of improving lysosomal function in respiration-deficient cells; 3a. To assess how unresolved organelle dysfunction and metabolic stresses exclusively in immune cells affects organism homeostasis and lifespan. 3b. To decipher the molecular mediators by which organelle dysfunction in T cells contributes to age-associated diseases, with special focus in cardiorenal and metabolic syndromes. In sum, EndoMitTalk puts forward an ambitious and multidisciplinary but feasible program with the wide purpose of understanding and improving clinical interventions in mitochondrial diseases and age-related pathologies.
Summary
For many years, mitochondria were viewed as semiautonomous organelles, required only for cellular energetics. This view has been displaced by the concept that mitochondria are fully integrated into the life of the cell and that mitochondrial function and stress response rapidly affect other organelles, and even other tissues. A recent discovery from my lab demonstrated that mitochondrial metabolism regulates lysosomal degradation (Cell Metabolism, 2015), thus opening the way to investigate the mechanism behind communication between these organelles and its consequences for homeostasis. With this proposal, we want to assess how mitochondrial crosstalk with endolysosomal compartment controls cellular homeostasis and how mitochondrial dysfunction in certain tissues may account for systemic effects on the rest of the organism. EndoMitTalk will deliver significant breakthroughs on (1) the molecular mediators of endolysosomal-mitochondria communication, and how deregulation of this crosstalk alters cellular (2), and organism homeostasis (3). Our central goals are: 1a,b. To identify metabolic and physical connections mediating endolysosomal-mitochondria crosstalk; 2a. To decode the consequences of altered interorganelle communication in cellular homeostasis 2b. To study the therapeutic potential of improving lysosomal function in respiration-deficient cells; 3a. To assess how unresolved organelle dysfunction and metabolic stresses exclusively in immune cells affects organism homeostasis and lifespan. 3b. To decipher the molecular mediators by which organelle dysfunction in T cells contributes to age-associated diseases, with special focus in cardiorenal and metabolic syndromes. In sum, EndoMitTalk puts forward an ambitious and multidisciplinary but feasible program with the wide purpose of understanding and improving clinical interventions in mitochondrial diseases and age-related pathologies.
Max ERC Funding
1 498 625 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym EPINORC
Project Epigenetic Disruption of Non-Coding RNAs in Human Cancer
Researcher (PI) Manel Esteller Badosa
Host Institution (HI) FUNDACIO INSTITUT D'INVESTIGACIO BIOMEDICA DE BELLVITGE
Call Details Advanced Grant (AdG), LS4, ERC-2010-AdG_20100317
Summary In recent years, my laboratory, as well as others, have established the observation that epigenetic disruption, particularly in the DNA methylation and histone modification patterns, contributes to the initiation and progression of human tumors (Esteller, Nat Rev Genet 2007; Esteller, N Engl J Med 2008; Esteller, Nat Rev Biotech, In Press, 2010). Even more recently, it has been recognized that microRNAs, small non-coding RNAs that are thought to regulate gene expression by sequence-specific base pairing in mRNA targets, also play a key role in the biology of the cell, and that they can also have an impact in the development of many diseases, including cancer (le Sage and Agami, 2006; Blenkiron and Miska, 2007). However, there is little understanding about epigenetic modifications that might regulate the activity of microRNAs and other non-coding RNAs (ncRNAs), such as long non-coding RNAs (lncRNAs), Piwi-interacting RNAs (piRNAs), small-interfering RNAs (siRNAs), transcribed ultraconserved regions (T-UCRs), small nuclear RNAs (snRNAs), small nucleolar RNAs (snoRNAs), long interspersed ncRNAs (lincRNAs), promoter-associated RNAs (PASRs and PALRs) and terminator-associated sRNAs (TASRs) (Calin et al., 2007; Mercer, et al., 2009; Ghildiyal & Zamore, 2009; Jacquier, 2009). Our ignorance in this respect is even more significant if we consider these questions in the domain of cancer. Making best use of our expertise in several of these fields, my group will tackle the study of the epigenetic modifications that regulate ncRNA expression and how the DNA methylation and histone modifications profiles of these loci might become distorted in human cancer. These findings could have profound consequences not only in the understading of tumor biology, but in the design of better molecular staging, diagnosis and treatments of human malignancies.
Summary
In recent years, my laboratory, as well as others, have established the observation that epigenetic disruption, particularly in the DNA methylation and histone modification patterns, contributes to the initiation and progression of human tumors (Esteller, Nat Rev Genet 2007; Esteller, N Engl J Med 2008; Esteller, Nat Rev Biotech, In Press, 2010). Even more recently, it has been recognized that microRNAs, small non-coding RNAs that are thought to regulate gene expression by sequence-specific base pairing in mRNA targets, also play a key role in the biology of the cell, and that they can also have an impact in the development of many diseases, including cancer (le Sage and Agami, 2006; Blenkiron and Miska, 2007). However, there is little understanding about epigenetic modifications that might regulate the activity of microRNAs and other non-coding RNAs (ncRNAs), such as long non-coding RNAs (lncRNAs), Piwi-interacting RNAs (piRNAs), small-interfering RNAs (siRNAs), transcribed ultraconserved regions (T-UCRs), small nuclear RNAs (snRNAs), small nucleolar RNAs (snoRNAs), long interspersed ncRNAs (lincRNAs), promoter-associated RNAs (PASRs and PALRs) and terminator-associated sRNAs (TASRs) (Calin et al., 2007; Mercer, et al., 2009; Ghildiyal & Zamore, 2009; Jacquier, 2009). Our ignorance in this respect is even more significant if we consider these questions in the domain of cancer. Making best use of our expertise in several of these fields, my group will tackle the study of the epigenetic modifications that regulate ncRNA expression and how the DNA methylation and histone modifications profiles of these loci might become distorted in human cancer. These findings could have profound consequences not only in the understading of tumor biology, but in the design of better molecular staging, diagnosis and treatments of human malignancies.
Max ERC Funding
2 497 240 €
Duration
Start date: 2011-04-01, End date: 2016-03-31
Project acronym GALACTICNUCLEUS
Project The Fingerprint of a Galactic Nucleus: A Multi-Wavelength, High-Angular Resolution, Near-Infrared Study of the Centre of the Milky Way
Researcher (PI) Rainer Schödel
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Consolidator Grant (CoG), PE9, ERC-2013-CoG
Summary Galactic stellar nuclei are very common in all types of galaxies and are marked by the presence of nuclear star clusters, the densest and most massive star clusters in the present-day Universe. Their formation is still an unresolved puzzle. The centre of the Milky Way contains a massive black hole and a stellar nucleus and is orders of magnitude closer than any comparable target. It is the only galactic nucleus and the most extreme astrophysical environment that we can examine on scales of milli-parsecs. It is therefore a crucial laboratory for studying galactic nuclei and their role in the context of galaxy evolution. Yet, suitable data that would allow us to examine the stellar component of the Galactic Centre exist for less than 1% of its projected area. Moreover, the well-explored regions are extraordinary, like the central parsec around the massive black hole, and therefore probably not representative for the overall environment. Fundamental questions on the stellar population, structure and assembly history of the Galactic Centre remain therefore unanswered. This project aims at addressing the open questions by obtaining accurate, high-angular resolution, multi-wavelength near-infrared photometry for an area of several 100 pc^2, a more than ten-fold increase compared to the current state of affairs. The Galactic Centre presents unique observational challenges because of a combination of high extinction and extreme stellar crowding. It is therefore not adequately covered by existing or upcoming imaging surveys. I present a project that is specifically tailored to overcome these observational challenges. In particular, I have developed a key technique to obtain the necessary sensitive, high-angular resolution images with a stable point spread function over large, crowded fields. It works with a range of existing ground-based instruments and will serve to complement existing data to provide a global and detailed picture of the stellar nucleus of the Milky Way.
Summary
Galactic stellar nuclei are very common in all types of galaxies and are marked by the presence of nuclear star clusters, the densest and most massive star clusters in the present-day Universe. Their formation is still an unresolved puzzle. The centre of the Milky Way contains a massive black hole and a stellar nucleus and is orders of magnitude closer than any comparable target. It is the only galactic nucleus and the most extreme astrophysical environment that we can examine on scales of milli-parsecs. It is therefore a crucial laboratory for studying galactic nuclei and their role in the context of galaxy evolution. Yet, suitable data that would allow us to examine the stellar component of the Galactic Centre exist for less than 1% of its projected area. Moreover, the well-explored regions are extraordinary, like the central parsec around the massive black hole, and therefore probably not representative for the overall environment. Fundamental questions on the stellar population, structure and assembly history of the Galactic Centre remain therefore unanswered. This project aims at addressing the open questions by obtaining accurate, high-angular resolution, multi-wavelength near-infrared photometry for an area of several 100 pc^2, a more than ten-fold increase compared to the current state of affairs. The Galactic Centre presents unique observational challenges because of a combination of high extinction and extreme stellar crowding. It is therefore not adequately covered by existing or upcoming imaging surveys. I present a project that is specifically tailored to overcome these observational challenges. In particular, I have developed a key technique to obtain the necessary sensitive, high-angular resolution images with a stable point spread function over large, crowded fields. It works with a range of existing ground-based instruments and will serve to complement existing data to provide a global and detailed picture of the stellar nucleus of the Milky Way.
Max ERC Funding
1 547 657 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym icyMARS
Project Cold and wet early Mars: Proposing and testing a new theory to understand the early Martian environments
Researcher (PI) Alberto Gonzalez Fairen
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Starting Grant (StG), PE9, ERC-2012-StG_20111012
Summary Geologic evidence indicative of flowing and ponding liquid water on the surface of ancient Mars appears abundantly across most of the Martian landscape, indicating that liquid water was present in variable amounts and for long periods of time on and/or near the surface at different moments of Mars’ early history, the Noachian era. Early Mars appears to have been “wet”. However, the presence of liquid water on the surface of early Mars is difficult to reconcile with the reduced solar luminosity at 3.8 Ga. and before, which would have imposed mean temperatures below freezing all over the planet. Atmospheric greenhouse gases and carbon dioxide ice clouds in the upper troposphere are suggested to provide over freezing temperatures, explaining some of this discrepancy, but these solutions have been probed to face numerous problems. So, it is difficult to explain the early Martian hydrology invoking global “warm” conditions. Here I propose to conduct interdisciplinary investigations in order to define and test a new hypothesis to understand the early environmental traits on Mars: that the young Martian surface was characterized by global mean freezing conditions, as predicted by climate models, and at the same time a vigorous hydrogeological cycle was active during hundreds of millions of years, as confirmed by geomorphological and mineralogical analyses. The aim of this investigation is to comprehensively analyze the triggers, traits and consequences of a cold aqueous environment dominating the Noachian, studying the geomorphological, mineralogical and geochemical evidences that such a hydrological cycle would have left behind, and also proposing new paths for the astrobiological exploration of Mars on the basis of geochemical and geomicrobiological studies in cold aqueous environments. Mission-derived datasets will be used to test hypotheses through paleogeomorphological reconstructions, theoretical modeling and experiments in the laboratory.
Summary
Geologic evidence indicative of flowing and ponding liquid water on the surface of ancient Mars appears abundantly across most of the Martian landscape, indicating that liquid water was present in variable amounts and for long periods of time on and/or near the surface at different moments of Mars’ early history, the Noachian era. Early Mars appears to have been “wet”. However, the presence of liquid water on the surface of early Mars is difficult to reconcile with the reduced solar luminosity at 3.8 Ga. and before, which would have imposed mean temperatures below freezing all over the planet. Atmospheric greenhouse gases and carbon dioxide ice clouds in the upper troposphere are suggested to provide over freezing temperatures, explaining some of this discrepancy, but these solutions have been probed to face numerous problems. So, it is difficult to explain the early Martian hydrology invoking global “warm” conditions. Here I propose to conduct interdisciplinary investigations in order to define and test a new hypothesis to understand the early environmental traits on Mars: that the young Martian surface was characterized by global mean freezing conditions, as predicted by climate models, and at the same time a vigorous hydrogeological cycle was active during hundreds of millions of years, as confirmed by geomorphological and mineralogical analyses. The aim of this investigation is to comprehensively analyze the triggers, traits and consequences of a cold aqueous environment dominating the Noachian, studying the geomorphological, mineralogical and geochemical evidences that such a hydrological cycle would have left behind, and also proposing new paths for the astrobiological exploration of Mars on the basis of geochemical and geomicrobiological studies in cold aqueous environments. Mission-derived datasets will be used to test hypotheses through paleogeomorphological reconstructions, theoretical modeling and experiments in the laboratory.
Max ERC Funding
1 411 200 €
Duration
Start date: 2014-01-01, End date: 2018-12-31
Project acronym IDRICA
Project Improving Drought Resistance in Crops and Arabidopsis
Researcher (PI) Ana Isabel Caño Delgado
Host Institution (HI) CENTRE DE RECERCA EN AGRIGENOMICA CSIC-IRTA-UAB-UB
Call Details Consolidator Grant (CoG), LS9, ERC-2015-CoG
Summary Drought is the first cause of agricultural losses globally, and represents a major threat to food security. Currently, plant biotechnology stands as the most promising strategy to produce crops capable of producing high yields in fed rain conditions. From the study of whole-plants, the main underlying mechanism for responses to drought stress has been uncovered, and multiple drought resistance genes have been engineered into crops. So far, plants with enhanced drought resistance displayed reduced crop yield, which imposes the search of novel approaches to uncouple drought resistance from plant growth. Our laboratory has recently shown, for the first time, that the receptors of Brassinosteroid hormones use cell-specific pathways to allocate different developmental responses during root growth. In particular, we have found that cell-specific components of the stem cell niche have the ability to control cellular responses to stress to promote stem renewal to ensure root growth. Additionally, we have also found that BR mutants are resistant to drought, together opening an exceptional opportunity to investigate the mechanisms that confer drought resistance with cellular specificity in plants. In this project, we will use Brassinosteroid signaling in the Arabidopsis root to investigate the mechanism for drought stress resistance in plant and to design novel molecules able to confer resistance to the drought stress. Finally, we will translate our research results and tools into Sorghum bicolor (Sorghum), a crop cereal of paramount importance in fed rain regions of the planet. Our research will impact in science, providing new avenues for the study of hormone signaling in plants, and in society, by providing sustainable solutions for enhance crop production in limiting water environments.
Summary
Drought is the first cause of agricultural losses globally, and represents a major threat to food security. Currently, plant biotechnology stands as the most promising strategy to produce crops capable of producing high yields in fed rain conditions. From the study of whole-plants, the main underlying mechanism for responses to drought stress has been uncovered, and multiple drought resistance genes have been engineered into crops. So far, plants with enhanced drought resistance displayed reduced crop yield, which imposes the search of novel approaches to uncouple drought resistance from plant growth. Our laboratory has recently shown, for the first time, that the receptors of Brassinosteroid hormones use cell-specific pathways to allocate different developmental responses during root growth. In particular, we have found that cell-specific components of the stem cell niche have the ability to control cellular responses to stress to promote stem renewal to ensure root growth. Additionally, we have also found that BR mutants are resistant to drought, together opening an exceptional opportunity to investigate the mechanisms that confer drought resistance with cellular specificity in plants. In this project, we will use Brassinosteroid signaling in the Arabidopsis root to investigate the mechanism for drought stress resistance in plant and to design novel molecules able to confer resistance to the drought stress. Finally, we will translate our research results and tools into Sorghum bicolor (Sorghum), a crop cereal of paramount importance in fed rain regions of the planet. Our research will impact in science, providing new avenues for the study of hormone signaling in plants, and in society, by providing sustainable solutions for enhance crop production in limiting water environments.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-11-01, End date: 2021-10-31
Project acronym INFANTLEUKEMIA
Project GENOMIC, CELLULAR AND DEVELOPMENTAL RECONSTRUCTION OFINFANT MLL-AF4+ ACUTE LYMPHOBLASTIC LEUKEMIA
Researcher (PI) Pablo Menendez Buján
Host Institution (HI) FUNDACIO INSTITUT DE RECERCA CONTRA LA LEUCEMIA JOSEP CARRERAS
Call Details Consolidator Grant (CoG), LS4, ERC-2014-CoG
Summary Infant cancer is very distinct to adult cancer and it is progressively seen as a developmental disease. An intriguing infant cancer is the t(4;11) acute lymphoblastic leukemia (ALL) characterized by the hallmark rearrangement MLL-AF4 (MA4), and associated with dismal prognosis. The 100% concordance in twins and its prenatal onset suggest an extremely rapid disease progression. Many key issues remain elusive:
Is MA4 leukemogenic?
Which are other relevant oncogenic drivers?
Which is the nature of the cell transformed by MA4?
Which is the leukemia-initiating cell (LIC)?
Does this ALL follow a hierarchical or stochastic cancer model?
How to explain therapy resistance and CNS involvement?
To what extent do genetics vs epigenetics contribute this ALL?
These questions remain a challenge due to: 1) the absence of prospective studies on diagnostic/remission-matched samples, 2) the lack of models which faithfully reproduce the disease and 3) a surprising genomic stability of this ALL.
I hypothesize that a Multilayer-Omics to function approach in patient blasts and early human hematopoietic stem/progenitor cells (HSPC) is required to fully scrutinize the biology underlying this life-threatening leukemia. I will perform genome-wide studies on the mutational landscape, DNA and H3K79 methylation profiles, and transcriptome on a uniquely available, large cohort of diagnostic/remission-matched samples. Omics data integration will provide unprecedented information about oncogenic drivers which must be analyzed in ground-breaking functional assays using patient blasts and early HSPCs carrying a CRISPR/Cas9-mediated locus/allele-specific t(4;11). Serial xenografts combined with exome-seq in paired diagnostic samples and xenografts will identify the LIC and determine whether variegated genetics may underlie clonal functional heterogeneity. This project will provide a precise understanding and a disease model for MA4+ ALL, offering a platform for new treatment strategies.
Summary
Infant cancer is very distinct to adult cancer and it is progressively seen as a developmental disease. An intriguing infant cancer is the t(4;11) acute lymphoblastic leukemia (ALL) characterized by the hallmark rearrangement MLL-AF4 (MA4), and associated with dismal prognosis. The 100% concordance in twins and its prenatal onset suggest an extremely rapid disease progression. Many key issues remain elusive:
Is MA4 leukemogenic?
Which are other relevant oncogenic drivers?
Which is the nature of the cell transformed by MA4?
Which is the leukemia-initiating cell (LIC)?
Does this ALL follow a hierarchical or stochastic cancer model?
How to explain therapy resistance and CNS involvement?
To what extent do genetics vs epigenetics contribute this ALL?
These questions remain a challenge due to: 1) the absence of prospective studies on diagnostic/remission-matched samples, 2) the lack of models which faithfully reproduce the disease and 3) a surprising genomic stability of this ALL.
I hypothesize that a Multilayer-Omics to function approach in patient blasts and early human hematopoietic stem/progenitor cells (HSPC) is required to fully scrutinize the biology underlying this life-threatening leukemia. I will perform genome-wide studies on the mutational landscape, DNA and H3K79 methylation profiles, and transcriptome on a uniquely available, large cohort of diagnostic/remission-matched samples. Omics data integration will provide unprecedented information about oncogenic drivers which must be analyzed in ground-breaking functional assays using patient blasts and early HSPCs carrying a CRISPR/Cas9-mediated locus/allele-specific t(4;11). Serial xenografts combined with exome-seq in paired diagnostic samples and xenografts will identify the LIC and determine whether variegated genetics may underlie clonal functional heterogeneity. This project will provide a precise understanding and a disease model for MA4+ ALL, offering a platform for new treatment strategies.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym INTRAHETEROSEQ
Project Molecular characterization of the role of intra-tumor heterogeneity in cancer progression and metastasis
Researcher (PI) Ignacio VARELA EGOCHEAGA
Host Institution (HI) UNIVERSIDAD DE CANTABRIA
Call Details Starting Grant (StG), LS4, ERC-2014-STG
Summary Cancer is caused by somatically acquired changes in the DNA. Some of these changes fall in “cancer genes”, conferring clonal selective advantage to the cells that carry the mutant alleles. Identifying these genes/pathways is of vital importance for a correct understanding of cancer biology as well as for the diagnosis and treatment of human malignancies. In this respect, the use of genetically modified mice has been extremely useful in the past for characterizing the molecular pathways involved in cancer progression. The remarkable progress made during the last two decades on the genetic modification of mouse genomes offers unique opportunities to investigate different aspects of tumor molecular behavior, impossible to study on human samples.
Recently, the advent of next-generation sequencing technologies has provided new strategies for the systematic genome-wide identification of somatic changes in cancer cell genomes. Using these technologies, we and others have characterized the high intra-tumor heterogeneity observed in some human tumors. Although the exact significance of this heterogeneity is uncertain, it seems to be responsible for key aspects in the management of cancer patients such as metastasis predisposition and tissue specificity or treatment resistance.
Taking advantage of next-generation sequencing, we propose to finely characterize the intra-tumor heterogeneity evolution during the progression of tumors induced in a mouse model of pancreatic cancer, as well as, for the first time, to purify the different cell populations these primary tumors are composed of. A complete genomic and transcriptomic characterization of these populations followed by posterior functional assays will help us to identify the genes/pathways involved in tumor progression as well as metastatic potential and its tissue specificity. This new knowledge could finally contribute to a better understanding of cancer and to the design of more efficient anti-tumor therapies
Summary
Cancer is caused by somatically acquired changes in the DNA. Some of these changes fall in “cancer genes”, conferring clonal selective advantage to the cells that carry the mutant alleles. Identifying these genes/pathways is of vital importance for a correct understanding of cancer biology as well as for the diagnosis and treatment of human malignancies. In this respect, the use of genetically modified mice has been extremely useful in the past for characterizing the molecular pathways involved in cancer progression. The remarkable progress made during the last two decades on the genetic modification of mouse genomes offers unique opportunities to investigate different aspects of tumor molecular behavior, impossible to study on human samples.
Recently, the advent of next-generation sequencing technologies has provided new strategies for the systematic genome-wide identification of somatic changes in cancer cell genomes. Using these technologies, we and others have characterized the high intra-tumor heterogeneity observed in some human tumors. Although the exact significance of this heterogeneity is uncertain, it seems to be responsible for key aspects in the management of cancer patients such as metastasis predisposition and tissue specificity or treatment resistance.
Taking advantage of next-generation sequencing, we propose to finely characterize the intra-tumor heterogeneity evolution during the progression of tumors induced in a mouse model of pancreatic cancer, as well as, for the first time, to purify the different cell populations these primary tumors are composed of. A complete genomic and transcriptomic characterization of these populations followed by posterior functional assays will help us to identify the genes/pathways involved in tumor progression as well as metastatic potential and its tissue specificity. This new knowledge could finally contribute to a better understanding of cancer and to the design of more efficient anti-tumor therapies
Max ERC Funding
1 498 850 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym LIPOMET
Project Dietary Influences on Metastasis: How, When, and Why
Researcher (PI) Salvador Aznar Benitah
Host Institution (HI) FUNDACIO INSTITUT DE RECERCA BIOMEDICA (IRB BARCELONA)
Call Details Advanced Grant (AdG), LS4, ERC-2017-ADG
Summary We have recently identified metastasis-initiating cells (MICs) in several types of tumors (Nature, 2017)1.
Intriguingly, MICs: (i) are exclusive in their ability to generate metastases when transplanted; (ii) express the
fatty acid channel CD36 and have a unique lipid metabolic signature; (iii) are exquisitely sensitive to the
levels of fat in circulation, thus providing a link between the predisposition of metastasis and dietary fat; (iv)
are highly sensitive to CD36 inhibition, which almost completely abolishes their metastatic potential.
We still do not know how MICs promote metastasis or how MICs are influenced by dietary fat. In
particular: (A) where are MICs located within the tumor, and does this location influence their behavior?
How and where do they attach and expand at metastatic sites? (B) Why are MICs so sensitive to specific
dietary lipids, and how do these lipids promote metastasis at the molecular and cellular levels? (C) Is the
prolonged consumption of a high-fat diet a risk factor for developing metastatic tumors? If so, what are the
underlying genetic and epigenetic causes for this effect? Can we revert these causes?
To answer these questions, we will combine state-of-the-art in vivo functional models of metastasis, with
quantitative metabolomics and proteomics, epigenetic and geographical position (3D) single-cell
transcriptomic studies, as well as integrative computational analyses, using preclinical models and patientderived
carcinomas of melanoma, oral cancer and breast cancer.
We expect our project to provide fundamental insights into the mechanisms of metastasis, and how they are
influenced by diet. This is highly relevant as 1) large quantities of fatty acids are typically consumed in
Western diets; and 2) metastasis is the leading cause of cancer-related deaths. We also tackle a timely
medical unmet need by exploring the therapeutic anti-metastatic potential of targeting fatty acid metabolism
in cancer patients.
Summary
We have recently identified metastasis-initiating cells (MICs) in several types of tumors (Nature, 2017)1.
Intriguingly, MICs: (i) are exclusive in their ability to generate metastases when transplanted; (ii) express the
fatty acid channel CD36 and have a unique lipid metabolic signature; (iii) are exquisitely sensitive to the
levels of fat in circulation, thus providing a link between the predisposition of metastasis and dietary fat; (iv)
are highly sensitive to CD36 inhibition, which almost completely abolishes their metastatic potential.
We still do not know how MICs promote metastasis or how MICs are influenced by dietary fat. In
particular: (A) where are MICs located within the tumor, and does this location influence their behavior?
How and where do they attach and expand at metastatic sites? (B) Why are MICs so sensitive to specific
dietary lipids, and how do these lipids promote metastasis at the molecular and cellular levels? (C) Is the
prolonged consumption of a high-fat diet a risk factor for developing metastatic tumors? If so, what are the
underlying genetic and epigenetic causes for this effect? Can we revert these causes?
To answer these questions, we will combine state-of-the-art in vivo functional models of metastasis, with
quantitative metabolomics and proteomics, epigenetic and geographical position (3D) single-cell
transcriptomic studies, as well as integrative computational analyses, using preclinical models and patientderived
carcinomas of melanoma, oral cancer and breast cancer.
We expect our project to provide fundamental insights into the mechanisms of metastasis, and how they are
influenced by diet. This is highly relevant as 1) large quantities of fatty acids are typically consumed in
Western diets; and 2) metastasis is the leading cause of cancer-related deaths. We also tackle a timely
medical unmet need by exploring the therapeutic anti-metastatic potential of targeting fatty acid metabolism
in cancer patients.
Max ERC Funding
2 370 625 €
Duration
Start date: 2018-08-01, End date: 2023-07-31
Project acronym LT-NRBS
Project Lab-in-a-tube and Nanorobotic biosensors
Researcher (PI) Samuel Sánchez Ordóñez
Host Institution (HI) FUNDACIO INSTITUT DE BIOENGINYERIA DE CATALUNYA
Call Details Starting Grant (StG), LS9, ERC-2012-StG_20111109
Summary The goal of this project is to develop new types of biosensors based on two different approaches: (i) a new bioanalytic microsystem platform for cell growth, manipulation and analysis using on-chip integrated microtubes and (ii) the use of synthetic self-propelled nanomotors for bioanalytical and biosensing applications. Based on the novel “Lab-in-a-tube” concept, we will design a multifunctional device for the capturing, growth and sensing of single cell behaviours inside “glass” microtubes to be employed for diverse biological applications. We will decorate the walls of the microtubes with proteins from the extracellular matrix enabling the long-term study of cellular changes such as mitosis time, spindle reorientation, DNA damage and cellular differentiation. These microtubes are fabricated by the well-established rolled-up nanotechnology developed in the host institution. Moreover, the multifunctionality of the “Lab-in-a-tube” platform will be extended by integrating different modules into a single microtubular unit, bringing up several applications such as optofluidics(bio)sensors, electrodes for electrochemical control and sensing, and magnetic biodetection.
At the IIN institute in IFW Dresden, we are pioneers on the fabrication of catalytic microjet engines (microbots) and their use for transporting different kinds of objects in vitro into a fluid. The remote controlled motion of these autonomous microbots and the transport of microobjects and cells to specific targets within lab-on-a-chip systems is possible. Their walls can be biofunctionalized with enzymes, antibodies or DNA, the catalytic microbots representing a novel and unique tool for biosensing, environmental and biomedical applications. Our next step is to use biocompatible fuels to propel these microbots with the final aim of transporting and delivering drugs in vivo.The separation of cancer cells, bacteria and other biomaterials to build up new tissues or to replace disease cells are also aimed.
Summary
The goal of this project is to develop new types of biosensors based on two different approaches: (i) a new bioanalytic microsystem platform for cell growth, manipulation and analysis using on-chip integrated microtubes and (ii) the use of synthetic self-propelled nanomotors for bioanalytical and biosensing applications. Based on the novel “Lab-in-a-tube” concept, we will design a multifunctional device for the capturing, growth and sensing of single cell behaviours inside “glass” microtubes to be employed for diverse biological applications. We will decorate the walls of the microtubes with proteins from the extracellular matrix enabling the long-term study of cellular changes such as mitosis time, spindle reorientation, DNA damage and cellular differentiation. These microtubes are fabricated by the well-established rolled-up nanotechnology developed in the host institution. Moreover, the multifunctionality of the “Lab-in-a-tube” platform will be extended by integrating different modules into a single microtubular unit, bringing up several applications such as optofluidics(bio)sensors, electrodes for electrochemical control and sensing, and magnetic biodetection.
At the IIN institute in IFW Dresden, we are pioneers on the fabrication of catalytic microjet engines (microbots) and their use for transporting different kinds of objects in vitro into a fluid. The remote controlled motion of these autonomous microbots and the transport of microobjects and cells to specific targets within lab-on-a-chip systems is possible. Their walls can be biofunctionalized with enzymes, antibodies or DNA, the catalytic microbots representing a novel and unique tool for biosensing, environmental and biomedical applications. Our next step is to use biocompatible fuels to propel these microbots with the final aim of transporting and delivering drugs in vivo.The separation of cancer cells, bacteria and other biomaterials to build up new tissues or to replace disease cells are also aimed.
Max ERC Funding
1 499 880 €
Duration
Start date: 2013-01-01, End date: 2017-12-31
Project acronym MAGNESIA
Project The impact of highly magnetic neutron stars in the explosive and transient Universe
Researcher (PI) Nanda Rea
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Consolidator Grant (CoG), PE9, ERC-2018-COG
Summary The gravitational wave window is now open. It is then imperative to build quantitative models of neutron stars that use all the available tracers to constrain fundamental physics at the highest densities and magnetic fields. The most magnetic neutron stars, the magnetars, have been recently suggested to be powering a large variety of explosive and transient events. The enormous rotational power at birth, and the magnetic energy they can release via large flares, put the magnetars in the (yet) hand-wavy interpretations of gamma-ray bursts, the early phases of double neutron star mergers, super-luminous supernovae, hypernovae, fast radio bursts, and ultra-luminous X-ray sources. However, despite knowing about 30 magnetars, we are lacking a census of how many we expect within the pulsar population, nor we have robust constraints on their flaring rates. The recent discovery of transient magnetars, of magnetar-like flares from sources with measured low dipolar magnetic fields and from typical radio pulsars, clearly showed that the magnetar census in our Galaxy is largely under-estimated. This hampers our understanding not only of the pulsar and magnetar populations, but also of them as possibly related to many of Universe’s explosive events. MAGNESIA will infer a sound Magnetar Census via an innovative approach that will build the first Pulsar Population Synthesis model able to cope with constraints/limits from multi-band observations, and taking into account 3D magnetic field evolution models and flaring rates for neutron stars. Combining expertise in multi-band observations, numerical modeling, nuclear physics, and computation, MAGNESIA will solve the physics, the observational systematic errors, and the computational challenges that inhibited previous works, to finally constrain the spin period and magnetic field distribution at birth of the neutron star population.
Summary
The gravitational wave window is now open. It is then imperative to build quantitative models of neutron stars that use all the available tracers to constrain fundamental physics at the highest densities and magnetic fields. The most magnetic neutron stars, the magnetars, have been recently suggested to be powering a large variety of explosive and transient events. The enormous rotational power at birth, and the magnetic energy they can release via large flares, put the magnetars in the (yet) hand-wavy interpretations of gamma-ray bursts, the early phases of double neutron star mergers, super-luminous supernovae, hypernovae, fast radio bursts, and ultra-luminous X-ray sources. However, despite knowing about 30 magnetars, we are lacking a census of how many we expect within the pulsar population, nor we have robust constraints on their flaring rates. The recent discovery of transient magnetars, of magnetar-like flares from sources with measured low dipolar magnetic fields and from typical radio pulsars, clearly showed that the magnetar census in our Galaxy is largely under-estimated. This hampers our understanding not only of the pulsar and magnetar populations, but also of them as possibly related to many of Universe’s explosive events. MAGNESIA will infer a sound Magnetar Census via an innovative approach that will build the first Pulsar Population Synthesis model able to cope with constraints/limits from multi-band observations, and taking into account 3D magnetic field evolution models and flaring rates for neutron stars. Combining expertise in multi-band observations, numerical modeling, nuclear physics, and computation, MAGNESIA will solve the physics, the observational systematic errors, and the computational challenges that inhibited previous works, to finally constrain the spin period and magnetic field distribution at birth of the neutron star population.
Max ERC Funding
2 263 148 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym MAMI
Project The Power of Maternal Microbes on Infant Health
Researcher (PI) MARIA CARMEN Collado Amores
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Starting Grant (StG), LS9, ERC-2014-STG
Summary Recent reports suggest that early microbial colonization has an important role for in promoting health. This may contribute to reduce the risk of chronic diseases such as obesity, allergies and inflammatory conditions. Advances in understanding host-microbe interactions imply that maternal microbiota plays a crucial role on health programming. This process begins in utero and it is modulated by mode of delivery and diet. My research has shown that i) specific shifts in milk microbial composition are associated with lactation time and mode of delivery, ii) milk microbes drive the infant microbiota composition; iii) maternal microbiota dysbiosis may be transferred to the infant. However, factors defining maternal microbiota and its biological role upon infant’s health are not yet fully understood. Hence, this project aims to characterize maternal microbes to be transferred to neonates and determine their function in infant health programming. The specific aims are:(1) understanding how the maternal microbiome is influenced by host and environmental factors;(2) characterizing the microbial core and bioactive compounds transmitted to the offspring mainly via breastfeeding and their key roles in the microbial modulation and host response;(3) understanding the interactions among breast milk bioactive compounds and their role in infant health;(4) shedding light on how maternal microbes influence the infant immune system & (5)development of new dietary strategies and therapies based on microbial replacement and modulation. To achieve these objectives, a systems biology approach by means of state-of-the-art techniques and new methodologies based on subpopulation enrichment by flow cytometer-sorter to study host–microbe interactions will be used. Results obtained will demonstrate the interaction between infant nutrition, microbes and host response in early life and its key role in health programming, enabling new applications in the field of personalized nutrition & medicine.
Summary
Recent reports suggest that early microbial colonization has an important role for in promoting health. This may contribute to reduce the risk of chronic diseases such as obesity, allergies and inflammatory conditions. Advances in understanding host-microbe interactions imply that maternal microbiota plays a crucial role on health programming. This process begins in utero and it is modulated by mode of delivery and diet. My research has shown that i) specific shifts in milk microbial composition are associated with lactation time and mode of delivery, ii) milk microbes drive the infant microbiota composition; iii) maternal microbiota dysbiosis may be transferred to the infant. However, factors defining maternal microbiota and its biological role upon infant’s health are not yet fully understood. Hence, this project aims to characterize maternal microbes to be transferred to neonates and determine their function in infant health programming. The specific aims are:(1) understanding how the maternal microbiome is influenced by host and environmental factors;(2) characterizing the microbial core and bioactive compounds transmitted to the offspring mainly via breastfeeding and their key roles in the microbial modulation and host response;(3) understanding the interactions among breast milk bioactive compounds and their role in infant health;(4) shedding light on how maternal microbes influence the infant immune system & (5)development of new dietary strategies and therapies based on microbial replacement and modulation. To achieve these objectives, a systems biology approach by means of state-of-the-art techniques and new methodologies based on subpopulation enrichment by flow cytometer-sorter to study host–microbe interactions will be used. Results obtained will demonstrate the interaction between infant nutrition, microbes and host response in early life and its key role in health programming, enabling new applications in the field of personalized nutrition & medicine.
Max ERC Funding
1 499 979 €
Duration
Start date: 2015-06-01, End date: 2020-12-31
Project acronym MarsFirstWater
Project The physicochemical nature of water on early Mars
Researcher (PI) Alberto Gonzalez Fairen
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Consolidator Grant (CoG), PE9, ERC-2018-COG
Summary Concepts of large bodies of glacial ice and liquid standing water, a robust hydrological cycle, and a rich Martian history of climate change are part of the current consensus model for early Mars. However, questions still poorly constrained include: a precise understanding of the inventory of water during the first billion years of Mars history and its early evolution on both global and local scales; whether liquid or solid H2O dominated, for what duration of time and where the water resided; what were the host-rock weathering rates and patterns and the physicochemical parameters defining such interactions; what specific landforms and mineralogies were generated during those periods; and what implications all these processes had on the possible inception of life on Mars. These fundamental questions represent large uncertainties and knowledge gaps. Therefore, a quantitative understanding of the basic characteristics of water on early Mars is very much needed and is the focus of this proposal.
This application outlines a plan for my research in the next five years, and explains how I propose to fully characterize the aqueous environments of early Mars through a quantitative and truly interdisciplinary investigation. Spacecraft mission-derived datasets will be consistently used to test hypotheses through paleogeomorphological reconstructions, geochemical modeling, mineralogical studies, and astrobiological investigations. The derived results will produce hard constraints on the physical evolution, chemical alteration and habitability of surface and near-surface aqueous environments on early Mars. The planned investigations will benefit from the combination of working with first-hand data from ongoing Mars missions and with the state-of-the-art laboratory tools at the host institution. The final expected result will be a complete understanding of the physicochemical nature of water on early Mars, also opening new paths for the astrobiological exploration of the planet.
Summary
Concepts of large bodies of glacial ice and liquid standing water, a robust hydrological cycle, and a rich Martian history of climate change are part of the current consensus model for early Mars. However, questions still poorly constrained include: a precise understanding of the inventory of water during the first billion years of Mars history and its early evolution on both global and local scales; whether liquid or solid H2O dominated, for what duration of time and where the water resided; what were the host-rock weathering rates and patterns and the physicochemical parameters defining such interactions; what specific landforms and mineralogies were generated during those periods; and what implications all these processes had on the possible inception of life on Mars. These fundamental questions represent large uncertainties and knowledge gaps. Therefore, a quantitative understanding of the basic characteristics of water on early Mars is very much needed and is the focus of this proposal.
This application outlines a plan for my research in the next five years, and explains how I propose to fully characterize the aqueous environments of early Mars through a quantitative and truly interdisciplinary investigation. Spacecraft mission-derived datasets will be consistently used to test hypotheses through paleogeomorphological reconstructions, geochemical modeling, mineralogical studies, and astrobiological investigations. The derived results will produce hard constraints on the physical evolution, chemical alteration and habitability of surface and near-surface aqueous environments on early Mars. The planned investigations will benefit from the combination of working with first-hand data from ongoing Mars missions and with the state-of-the-art laboratory tools at the host institution. The final expected result will be a complete understanding of the physicochemical nature of water on early Mars, also opening new paths for the astrobiological exploration of the planet.
Max ERC Funding
1 998 368 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym METALSYM
Project Metal transport in the tripartite symbiosis arbuscular mycorrhizal fungi-legume-rhizobia
Researcher (PI) Manuel Gonzalez Guerrero
Host Institution (HI) UNIVERSIDAD POLITECNICA DE MADRID
Call Details Starting Grant (StG), LS9, ERC-2013-StG
Summary Plant nutrition is essential to understand any physiological process in plant biology, as well as to improve crops, and agricultural practices. The root microbiome plays an important role in plant nutrition. The best characterized microbiome elements are two plant endosymbionts: arbuscular mycorrhizal fungi (AMF) and rhizobia. AMF are responsible for delivering most of the mineral nutrients required by the host plant. Similarly, rhizobia in legume nodules provide the vast majority of the nitrogen requirements. Given their importance for plant nutrition a significant effort in understanding macronutrient exchange among the symbionts has been made. However, very little is known about metal micronutrient exchange.
This is in contrast to the role of metals as essential nutrients for life (30-50 % of the proteins are metalloproteins) and to the yield-limiting effect that low soil metal bioavailability has worldwide. AMF are a source of metals, transferring the incorporated metals to the host,. Nitrogen-fixing rhizobia in mature nodules act as metal sinks, since the main enzymes required are highly expressed metalloproteins. We hypothesize that by changing the expression levels of the metal transporters involved, we will increase nitrogen fixation rates and increase plant metal uptake, resulting in higher crop production and fruit metal biofortification. Towards this goal, we will answer: i) How are metals incorporated from the AMF into the plant?, ii) How are metals delivered to the nodule?, iii) How are metals recovered from senescent nodules?, and iv) How does the natural variation of symbiotic-specific metal transporters affect yields and metal content of the fruit? In this project, we will use a multidisciplinary approach that involves metallotranscriptomics, plant physiology and molecular biology, and state-of-the art synchrotron based X-ray fluorescence to study metal distributions.
Summary
Plant nutrition is essential to understand any physiological process in plant biology, as well as to improve crops, and agricultural practices. The root microbiome plays an important role in plant nutrition. The best characterized microbiome elements are two plant endosymbionts: arbuscular mycorrhizal fungi (AMF) and rhizobia. AMF are responsible for delivering most of the mineral nutrients required by the host plant. Similarly, rhizobia in legume nodules provide the vast majority of the nitrogen requirements. Given their importance for plant nutrition a significant effort in understanding macronutrient exchange among the symbionts has been made. However, very little is known about metal micronutrient exchange.
This is in contrast to the role of metals as essential nutrients for life (30-50 % of the proteins are metalloproteins) and to the yield-limiting effect that low soil metal bioavailability has worldwide. AMF are a source of metals, transferring the incorporated metals to the host,. Nitrogen-fixing rhizobia in mature nodules act as metal sinks, since the main enzymes required are highly expressed metalloproteins. We hypothesize that by changing the expression levels of the metal transporters involved, we will increase nitrogen fixation rates and increase plant metal uptake, resulting in higher crop production and fruit metal biofortification. Towards this goal, we will answer: i) How are metals incorporated from the AMF into the plant?, ii) How are metals delivered to the nodule?, iii) How are metals recovered from senescent nodules?, and iv) How does the natural variation of symbiotic-specific metal transporters affect yields and metal content of the fruit? In this project, we will use a multidisciplinary approach that involves metallotranscriptomics, plant physiology and molecular biology, and state-of-the art synchrotron based X-ray fluorescence to study metal distributions.
Max ERC Funding
1 499 405 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym METARNAFLAMMATION
Project The RNA bridge between IRE-1 and PKR leading to metaflammation: discovery and intervention in atherosclerosis
Researcher (PI) Ebru Erbay
Host Institution (HI) BILKENT UNIVERSITESI VAKIF
Call Details Starting Grant (StG), LS4, ERC-2013-StG
Summary A close functional and molecular integration between metabolic and immune systems is crucial for systemic homeostasis and its’ deregulation is causally linked to obesity and associated diseases including insulin resistance, diabetes and atherosclerosis and known as cardiometabolic syndrome (CMS). Metabolic overload initiates a chronic inflammatory and stress response known as metaflammation and promotes the complications of CMS. The precise molecular mechanisms linking metabolic stress to immune activation and stress responses, however, remain elusive.
Earlier studies demonstrated metabolic overload stresses the endoplasmic reticulum (ER) and activates the unfolded protein response (UPR). ER is a critical intracellular metabolic hub orchestrating protein, lipid and calcium metabolism. These vital functions of ER are maintained by a conserved, adaptive stress response or UPR that emanates from its membranes. ER stress has emerged as a central paradigm in the pathogenesis of CMS and its reduction prevents atherosclerosis and promotes insulin sensitivity. However, a clear understanding of how metabolic stress is sensed and communicated by the ER is fundamental in designing specific and targeted therapy to ER stress in CMS. This application will investigate the ER stress response that can sense excess lipids and couple to inflammatory and stress responses, and whether its unique operation under metabolic stress can be suitable for therapeutic exploitation in CMS. This proposal tackles the unique modes of operation of two important players in the ER stress response that are coupled by metabolic stress, inositol-requiring enzyme-1 (IRE-1) and double-stranded RNA-activated kinase (PKR), by taking advantage of chemical-genetics to specifically modify their activities. When completed the proposed studies will have shed light on a little explored but central question in the field of immunometabolism regarding how nutrients engage inflammatory and stress pathways.
Summary
A close functional and molecular integration between metabolic and immune systems is crucial for systemic homeostasis and its’ deregulation is causally linked to obesity and associated diseases including insulin resistance, diabetes and atherosclerosis and known as cardiometabolic syndrome (CMS). Metabolic overload initiates a chronic inflammatory and stress response known as metaflammation and promotes the complications of CMS. The precise molecular mechanisms linking metabolic stress to immune activation and stress responses, however, remain elusive.
Earlier studies demonstrated metabolic overload stresses the endoplasmic reticulum (ER) and activates the unfolded protein response (UPR). ER is a critical intracellular metabolic hub orchestrating protein, lipid and calcium metabolism. These vital functions of ER are maintained by a conserved, adaptive stress response or UPR that emanates from its membranes. ER stress has emerged as a central paradigm in the pathogenesis of CMS and its reduction prevents atherosclerosis and promotes insulin sensitivity. However, a clear understanding of how metabolic stress is sensed and communicated by the ER is fundamental in designing specific and targeted therapy to ER stress in CMS. This application will investigate the ER stress response that can sense excess lipids and couple to inflammatory and stress responses, and whether its unique operation under metabolic stress can be suitable for therapeutic exploitation in CMS. This proposal tackles the unique modes of operation of two important players in the ER stress response that are coupled by metabolic stress, inositol-requiring enzyme-1 (IRE-1) and double-stranded RNA-activated kinase (PKR), by taking advantage of chemical-genetics to specifically modify their activities. When completed the proposed studies will have shed light on a little explored but central question in the field of immunometabolism regarding how nutrients engage inflammatory and stress pathways.
Max ERC Funding
1 362 921 €
Duration
Start date: 2014-01-01, End date: 2018-06-30
Project acronym MITOSENSING
Project Decoding mitochondrial nutrient-sensing programs in POMC neurons as key determinants of metabolic health
Researcher (PI) Marc CLARET CARLES
Host Institution (HI) CONSORCI INSTITUT D'INVESTIGACIONS BIOMEDIQUES AUGUST PI I SUNYER
Call Details Consolidator Grant (CoG), LS4, ERC-2016-COG
Summary Nutrient-sensing by POMC neurons is a critical process to monitor the metabolic status of the organism and to coordinate adaptive neuroendocrine, behavioural and metabolic effectors of energy balance. Mitochondria, as central commanders of cellular energy production and primary sources of bioenergetic signals, are logical candidates to play a key role in metabolic control. However, a comprehensive understanding of the mitochondria as nutrient-sensors and modulators of systemic energy homeostasis is lacking. MITOSENSING hypothesizes that dedicated mitochondrial networks in POMC neurons are able to sense, integrate and respond to alterations in the nutritional milieu and engage physiological actions to maintain energy balance. Thus, defects in these mitochondrial nutrient-sensing programs in this subset of neurons underlie the development of metabolic conditions such as obesity and type-2 diabetes (T2D). To test it, we will pursue three aims: 1) to identify transcriptionally-modulated mitochondrial nutrient-sensing programs in POMC neurons; 2) to investigate whether disruption of specific nutrient-sensing programs in POMC neurons cause metabolic disorders; 3) to investigate whether the development of lifestyle-associated metabolic disorders are caused by defective mitochondrial nutrient-sensing programs in POMC neurons. By providing neuron-specific, integrative, functional and mechanistic in vivo strategies, MITOSENSING will represent a major step forward into the understanding of mitochondria as a nutrient-sensing entity, the gene programs involved and their physiological regulatory functions in the context of energy balance control. Adequate coordination of neuronal nutrient-sensing with energy balance control is critical to sustain life, and thus understanding the molecular mechanisms governing these physiological programs will have an enormous scientific impact and also potential therapeutical implications for obesity and T2D.
Summary
Nutrient-sensing by POMC neurons is a critical process to monitor the metabolic status of the organism and to coordinate adaptive neuroendocrine, behavioural and metabolic effectors of energy balance. Mitochondria, as central commanders of cellular energy production and primary sources of bioenergetic signals, are logical candidates to play a key role in metabolic control. However, a comprehensive understanding of the mitochondria as nutrient-sensors and modulators of systemic energy homeostasis is lacking. MITOSENSING hypothesizes that dedicated mitochondrial networks in POMC neurons are able to sense, integrate and respond to alterations in the nutritional milieu and engage physiological actions to maintain energy balance. Thus, defects in these mitochondrial nutrient-sensing programs in this subset of neurons underlie the development of metabolic conditions such as obesity and type-2 diabetes (T2D). To test it, we will pursue three aims: 1) to identify transcriptionally-modulated mitochondrial nutrient-sensing programs in POMC neurons; 2) to investigate whether disruption of specific nutrient-sensing programs in POMC neurons cause metabolic disorders; 3) to investigate whether the development of lifestyle-associated metabolic disorders are caused by defective mitochondrial nutrient-sensing programs in POMC neurons. By providing neuron-specific, integrative, functional and mechanistic in vivo strategies, MITOSENSING will represent a major step forward into the understanding of mitochondria as a nutrient-sensing entity, the gene programs involved and their physiological regulatory functions in the context of energy balance control. Adequate coordination of neuronal nutrient-sensing with energy balance control is critical to sustain life, and thus understanding the molecular mechanisms governing these physiological programs will have an enormous scientific impact and also potential therapeutical implications for obesity and T2D.
Max ERC Funding
1 999 573 €
Duration
Start date: 2017-04-01, End date: 2022-03-31
Project acronym MITOSIGAGE
Project Mitochondrial crosstalk signalling in the regulation of ageing
Researcher (PI) Marta Artal Sanz
Host Institution (HI) UNIVERSIDAD PABLO DE OLAVIDE
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary Mitochondrial defects are associated with aging and age-related pathology, but the molecular mechanisms regulating mitochondrial function during ageing are poorly understood. The most relevant genetic pathway regulating ageing is the insulin/IGF-1 signalling (IIS) pathway. The mitochondrial prohibitin (PHB) complex influences cellular metabolism and mitochondrial biogenesis, affecting ageing in opposite ways in wild-type animals and IIS-defective C. elegans mutants. The aim of the proposed research programme is to shed light on the intricate communication between mitochondria and cell-signalling networks in the regulation of ageing. Our specific objectives are: 1-Elucidate the cellular signalling pathways involved in the metabolic responses to mitochondrial dysfunction upon PHB depletion in wild type animals and IIS-defective mutants, using genome-wide RNAi screens, 2-Conduct a comprehensive metabolic profiling of wild type and IIS mutants in the presence and absence of prohibitins and 3-Identify genetic suppressors of prohibitins by performing forward genetic suppressor screens. As an ultimate goal, genes discovered in C. elegans will be tested in vertebrate assays for a conserved role in ageing. We will implement an interdisciplinary approach that combines the genetic power of C. elegans with state-of-the-art metabolomic approaches as well as automated sorting and optical imaging technologies to monitor fat content and mitochondrial biogenesis, in a genome-wide scale, in vivo. The fine-tuning of cellular metabolism, by integration of diverse signalling inputs is the molecular basis of longevity. This project represents a truly integrative and innovative approach to identify cellular signalling pathways involved in mediating lifespan-extending metabolism adjustments, and what these metabolic adjustments entail. These studies will provide fundamental insights to understand the ageing process and to combat ageing-related diseases.
Summary
Mitochondrial defects are associated with aging and age-related pathology, but the molecular mechanisms regulating mitochondrial function during ageing are poorly understood. The most relevant genetic pathway regulating ageing is the insulin/IGF-1 signalling (IIS) pathway. The mitochondrial prohibitin (PHB) complex influences cellular metabolism and mitochondrial biogenesis, affecting ageing in opposite ways in wild-type animals and IIS-defective C. elegans mutants. The aim of the proposed research programme is to shed light on the intricate communication between mitochondria and cell-signalling networks in the regulation of ageing. Our specific objectives are: 1-Elucidate the cellular signalling pathways involved in the metabolic responses to mitochondrial dysfunction upon PHB depletion in wild type animals and IIS-defective mutants, using genome-wide RNAi screens, 2-Conduct a comprehensive metabolic profiling of wild type and IIS mutants in the presence and absence of prohibitins and 3-Identify genetic suppressors of prohibitins by performing forward genetic suppressor screens. As an ultimate goal, genes discovered in C. elegans will be tested in vertebrate assays for a conserved role in ageing. We will implement an interdisciplinary approach that combines the genetic power of C. elegans with state-of-the-art metabolomic approaches as well as automated sorting and optical imaging technologies to monitor fat content and mitochondrial biogenesis, in a genome-wide scale, in vivo. The fine-tuning of cellular metabolism, by integration of diverse signalling inputs is the molecular basis of longevity. This project represents a truly integrative and innovative approach to identify cellular signalling pathways involved in mediating lifespan-extending metabolism adjustments, and what these metabolic adjustments entail. These studies will provide fundamental insights to understand the ageing process and to combat ageing-related diseases.
Max ERC Funding
1 424 640 €
Duration
Start date: 2012-06-01, End date: 2018-05-31
Project acronym MYCOCHASSIS
Project Engineering of a minimal bacterial therapeutic chassis
Researcher (PI) Luis-Felipe Serrano Púbul
Host Institution (HI) FUNDACIO CENTRE DE REGULACIO GENOMICA
Call Details Advanced Grant (AdG), LS9, ERC-2014-ADG
Summary Engineering bacteria to deliver therapeutic agents or to present antigens for vaccination is an emerging area of research with great clinical potential. The most challenging issue in this field is the selection of the right bacteria to engineer, commonly known as “chassis”. The best chassis depends on the application but there is a common drawback in bacteria used nowadays: their complexity and the lack of quantitative information for many reactions which limits genome engineering to classical trial and error approaches. In this project, we want to engineer the genome-reduced bacterium M. pneumoniae using a whole-cell model that will drive the rational to create a chassis for human and animal therapy. Its small size (816 Kbases), the lack of cell wall, and the vast amount of comprehensive quantitative –omics datasets makes this bacterium one of the best candidates for chassis design. By combining bioinformatics, -omics, and biochemistry approaches with genome engineering tools, systems biology analyses, and computational whole-cell models, MYCOCHASSIS aims to: i) develop a whole cell-model based on organism-specific experimental data that will be validated experimentally and that can predict the impact of genome modifications; ii) implement genome engineering tools to delete non-essential pathogenic and virulent elements predicted by the whole-cell model to engineer a therapeutical chassis; iii) using the whole-cell model design and engineer genes and circuits to improve growth rate in a defined medium. iv) as a proof of concept introduce orthogonal gene circuits to secrete peptides and enzymes capable of dissolving in vitro biofilms made by the lung pathogens Pseudomonas aeruginosa and Staphylococus aureus. This project will validate the usefulness of whole-cell models for synthetic biology by modelling multiple genomic modifications orientated to facilitate engineering of biological systems.
Summary
Engineering bacteria to deliver therapeutic agents or to present antigens for vaccination is an emerging area of research with great clinical potential. The most challenging issue in this field is the selection of the right bacteria to engineer, commonly known as “chassis”. The best chassis depends on the application but there is a common drawback in bacteria used nowadays: their complexity and the lack of quantitative information for many reactions which limits genome engineering to classical trial and error approaches. In this project, we want to engineer the genome-reduced bacterium M. pneumoniae using a whole-cell model that will drive the rational to create a chassis for human and animal therapy. Its small size (816 Kbases), the lack of cell wall, and the vast amount of comprehensive quantitative –omics datasets makes this bacterium one of the best candidates for chassis design. By combining bioinformatics, -omics, and biochemistry approaches with genome engineering tools, systems biology analyses, and computational whole-cell models, MYCOCHASSIS aims to: i) develop a whole cell-model based on organism-specific experimental data that will be validated experimentally and that can predict the impact of genome modifications; ii) implement genome engineering tools to delete non-essential pathogenic and virulent elements predicted by the whole-cell model to engineer a therapeutical chassis; iii) using the whole-cell model design and engineer genes and circuits to improve growth rate in a defined medium. iv) as a proof of concept introduce orthogonal gene circuits to secrete peptides and enzymes capable of dissolving in vitro biofilms made by the lung pathogens Pseudomonas aeruginosa and Staphylococus aureus. This project will validate the usefulness of whole-cell models for synthetic biology by modelling multiple genomic modifications orientated to facilitate engineering of biological systems.
Max ERC Funding
2 454 522 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym NutrientSensingVivo
Project The Physiology of Nutrient Sensing by mTOR
Researcher (PI) Alejo Efeyan
Host Institution (HI) FUNDACION CENTRO NACIONAL DE INVESTIGACIONES ONCOLOGICAS CARLOS III
Call Details Starting Grant (StG), LS4, ERC-2014-STG
Summary A major role of metabolic alterations in the development of several human diseases, as diabetes, cancer and in the onset of ageing is becoming increasingly evident. This has a deep impact for human health due to the alarming increase in nutrient intake and obesity in the last decades. Fundamental aspects of how aberrant nutrient fluctuations trigger deregulated hormone levels and endocrine signals have been elucidated, being a prime example the phenomenon of insulin resistance. In contrast, how changes in nutrient levels elicit direct cell-autonomous signal transduction cascades and the consequences of these responses in physiology are less clear.
The signalling circuitry of direct nutrient sensing converges with that of hormones in the regulation of the mechanistic target of rapamycin (mTOR) kinase, a driver of anabolism, cell growth and proliferation. Deregulation of mTORC1 activity underlies the pathogenesis of cancer and diabetes, and its inhibitor rapamycin is approved as an anti-cancer agent and delays ageing from yeast to mammals. In spite of its importance for human disease, our understanding of the nutrient sensing signalling pathway and its impact in physiology is largely incomplete, as only a few years ago the direct molecular link between nutrients and mTORC1 activation, the Rag GTPases, were identified.
The present proposal aims to determine how the nutrient sensing signalling pathway affects mammalian physiology and metabolism, and whether its deregulation contributes to cancer, insulin resistance and aging. In particular, the objectives are: 1) To identify novel regulators of the Rag GTPases with unbiased and candidate-based approaches. 2) To establish the consequences of deregulated nutrient-dependent activation of mTORC1 in physiology, by means of genetically engineered mice. 3) To determine the impact of the nutrient sensing pathway in the effects of dietary restriction and nutrient limitation in glucose homeostasis and cancer.
Summary
A major role of metabolic alterations in the development of several human diseases, as diabetes, cancer and in the onset of ageing is becoming increasingly evident. This has a deep impact for human health due to the alarming increase in nutrient intake and obesity in the last decades. Fundamental aspects of how aberrant nutrient fluctuations trigger deregulated hormone levels and endocrine signals have been elucidated, being a prime example the phenomenon of insulin resistance. In contrast, how changes in nutrient levels elicit direct cell-autonomous signal transduction cascades and the consequences of these responses in physiology are less clear.
The signalling circuitry of direct nutrient sensing converges with that of hormones in the regulation of the mechanistic target of rapamycin (mTOR) kinase, a driver of anabolism, cell growth and proliferation. Deregulation of mTORC1 activity underlies the pathogenesis of cancer and diabetes, and its inhibitor rapamycin is approved as an anti-cancer agent and delays ageing from yeast to mammals. In spite of its importance for human disease, our understanding of the nutrient sensing signalling pathway and its impact in physiology is largely incomplete, as only a few years ago the direct molecular link between nutrients and mTORC1 activation, the Rag GTPases, were identified.
The present proposal aims to determine how the nutrient sensing signalling pathway affects mammalian physiology and metabolism, and whether its deregulation contributes to cancer, insulin resistance and aging. In particular, the objectives are: 1) To identify novel regulators of the Rag GTPases with unbiased and candidate-based approaches. 2) To establish the consequences of deregulated nutrient-dependent activation of mTORC1 in physiology, by means of genetically engineered mice. 3) To determine the impact of the nutrient sensing pathway in the effects of dietary restriction and nutrient limitation in glucose homeostasis and cancer.
Max ERC Funding
1 846 494 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym OBECAN
Project Role of obesity in the development of hepatocellular carcinoma
Researcher (PI) Guadalupe Sabio Buzo
Host Institution (HI) CENTRO NACIONAL DE INVESTIGACIONESCARDIOVASCULARES CARLOS III (F.S.P.)
Call Details Starting Grant (StG), LS4, ERC-2010-StG_20091118
Summary Obesity is associated with increased risk for epithelial tumors such as hepatocellular carcinoma (HCC). It is not known, however, whether obesity increases the risk for HCC simply because it promotes cirrhosis, a general risk factor for HCC, or through other mechanisms that operate independently of cirrhosis. Among these, obesity is associated with a chronic inflammatory state, with the release of cytokines such as IL-6 and TNFalpha, well-known HCC mediators. Obesity is normally linked to diabetes and in consequence, to hyperinsulinemia. This increase in circulating insulin levels is suggested to be a factor that contributes to cancer. Moreover, the increase in free fatty acids (FFA) in blood among obese patients promotes a compensatory response from liver that activates the transcription of genes required for beta-oxidation, leading to a reduction in non-physiological stores of lipids in the liver. This increase in beta-oxidation could result in oxidative stress, inflammation and the production of lipid peroxidation bioproducts, which are known mutagens. The precise mechanisms whereby FFA and cytosolic triglycerides exert their effects, resulting in the diabetic phenotype, remain poorly understood. Emerging evidence nonetheless links microRNA (miRNA) with lipid metabolism, suggesting that these small RNAs mediate this increase in beta-oxidation.
Our goal is to study how the components of the obesity state (inflammation, steatosis hyperinsulinemia and microRNA control of gene regulation) affect HCC development. We will use several mouse models in which one or more of these factors are reduced following induction of metabolic disease. We will also determine whether specific miRNAs that are down- or upregulated in the liver of mice on a high fat diet are implicated in HCC development.
Summary
Obesity is associated with increased risk for epithelial tumors such as hepatocellular carcinoma (HCC). It is not known, however, whether obesity increases the risk for HCC simply because it promotes cirrhosis, a general risk factor for HCC, or through other mechanisms that operate independently of cirrhosis. Among these, obesity is associated with a chronic inflammatory state, with the release of cytokines such as IL-6 and TNFalpha, well-known HCC mediators. Obesity is normally linked to diabetes and in consequence, to hyperinsulinemia. This increase in circulating insulin levels is suggested to be a factor that contributes to cancer. Moreover, the increase in free fatty acids (FFA) in blood among obese patients promotes a compensatory response from liver that activates the transcription of genes required for beta-oxidation, leading to a reduction in non-physiological stores of lipids in the liver. This increase in beta-oxidation could result in oxidative stress, inflammation and the production of lipid peroxidation bioproducts, which are known mutagens. The precise mechanisms whereby FFA and cytosolic triglycerides exert their effects, resulting in the diabetic phenotype, remain poorly understood. Emerging evidence nonetheless links microRNA (miRNA) with lipid metabolism, suggesting that these small RNAs mediate this increase in beta-oxidation.
Our goal is to study how the components of the obesity state (inflammation, steatosis hyperinsulinemia and microRNA control of gene regulation) affect HCC development. We will use several mouse models in which one or more of these factors are reduced following induction of metabolic disease. We will also determine whether specific miRNAs that are down- or upregulated in the liver of mice on a high fat diet are implicated in HCC development.
Max ERC Funding
1 498 043 €
Duration
Start date: 2010-12-01, End date: 2016-11-30
Project acronym OBERSTRESS
Project Hypothalamic Lipotoxicity and Endoplasmic Reticulum Stress: a New Pathophysiological Mechanism of Obesity
Researcher (PI) Miguel Antonio López Pérez
Host Institution (HI) UNIVERSIDAD DE SANTIAGO DE COMPOSTELA
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary It is well established that metabolically relevant organs such as adipose tissue, pancreatic beta cells, liver and skeletal muscle develop endoplasmic reticulum (ER) stress under conditions of obesity induced lipotoxicity. Here, the applicant will investigate if/how the hypothalamus is affected by lipotoxicity and ER stress in the context of obesity
* Scientific Hypotheses:
1. Obesity is associated with lipotoxicity and ER stress in the hypothalamus.
2. Hypothalamic ER stress may contribute to the development of obesity through dysregulation of the mechanisms controlling energy balance.
3. Based on our preliminary data, we hypothesize that CHOP, a mediator of ER stress, could be a key modulator of the association between obesity and ER dysfunction in the hypothalamus.
* General Objective: to determine the relevance of hypothalamic lipotoxicity and ER stress for the development of obesity and whether targeting ER stress mechanisms is a successful therapeutic strategy to prevent or revert obesity and its metabolic complications.
* Specific Objectives:
1. To determine whether obesity and the metabolic syndrome are associated with hypothalamic lipotoxicity, ER stress and whether these effects are hypothalamic nuclei specific
2. To determine whether primary hypothalamic ER stress is a cause of altered energy balance leading to obesity and metabolic complications
3. To determine whether in the context of obesity inhibition of ER stress in hypothalamus affects energy balance and obesity associated metabolic complications
4. To determine the role of CHOP on energy balance and obesity in specific hypothalamic neuronal populations
This project is central to the applicant’s goal of understanding how the hypothalamus regulates energy balance under physiological and pathophysiological conditions, as an essential step towards identifying and developing novel molecular drug targets to tackle the problem of obesity and their metabolic complications.
Summary
It is well established that metabolically relevant organs such as adipose tissue, pancreatic beta cells, liver and skeletal muscle develop endoplasmic reticulum (ER) stress under conditions of obesity induced lipotoxicity. Here, the applicant will investigate if/how the hypothalamus is affected by lipotoxicity and ER stress in the context of obesity
* Scientific Hypotheses:
1. Obesity is associated with lipotoxicity and ER stress in the hypothalamus.
2. Hypothalamic ER stress may contribute to the development of obesity through dysregulation of the mechanisms controlling energy balance.
3. Based on our preliminary data, we hypothesize that CHOP, a mediator of ER stress, could be a key modulator of the association between obesity and ER dysfunction in the hypothalamus.
* General Objective: to determine the relevance of hypothalamic lipotoxicity and ER stress for the development of obesity and whether targeting ER stress mechanisms is a successful therapeutic strategy to prevent or revert obesity and its metabolic complications.
* Specific Objectives:
1. To determine whether obesity and the metabolic syndrome are associated with hypothalamic lipotoxicity, ER stress and whether these effects are hypothalamic nuclei specific
2. To determine whether primary hypothalamic ER stress is a cause of altered energy balance leading to obesity and metabolic complications
3. To determine whether in the context of obesity inhibition of ER stress in hypothalamus affects energy balance and obesity associated metabolic complications
4. To determine the role of CHOP on energy balance and obesity in specific hypothalamic neuronal populations
This project is central to the applicant’s goal of understanding how the hypothalamus regulates energy balance under physiological and pathophysiological conditions, as an essential step towards identifying and developing novel molecular drug targets to tackle the problem of obesity and their metabolic complications.
Max ERC Funding
1 484 000 €
Duration
Start date: 2011-12-01, End date: 2017-05-31
Project acronym OBESITY53
Project p53 as a New Mediator of Energy Balance in the Brain
Researcher (PI) Ruben Nogueiras Pozo
Host Institution (HI) UNIVERSIDAD DE SANTIAGO DE COMPOSTELA
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary p53 is a transcriptional factor modulating numerous biological actions. Although it is best known for its role in cancer development, it is now evident that it is implicated in metabolism. More specifically, p53 modulates energy metabolism and homeostasis through their effects on adipocyte development and function. However, nothing is known about the potential metabolic function of p53 in the central nervous system.
Neuronal networks within the central nervous system play a crucial role in the regulation of food intake, body weight, and glucose homeostasis, so the main objective of this project will be to evaluate the potential of brain p53 as anti-obesity and/or anti-diabetic drug candidate. Our project will dissect precisely which specific components of energy balance are altered after central disruption or rescue of p53 signalling in selective neuronal populations, as well as the molecular pathways mediating these actions.
More precisely, we will disrupt the central p53 signalling specifically in hypothalamic POMC and AgRP neurons, which are crucial for energy and glucose homeostasis. We will also generate and characterize mice lacking p53 in dopamine neurons that are essential for mechanisms related with the reward of food. Once we know which specific areas are crucial for the central actions of p53, we will complete the experiments rescuing p53 expression in selective neuronal populations (POMC, AgRP or dopamine neurons) of p53 null mice. We will also investigate the interaction between p53 with leptin and ghrelin, likely the two more important hormones in the regulation of energy balance, which act through homeostatic and hedonic mechanisms. Understanding the precise role and mechanisms regulated by central p53 on energy balance may open new avenues for the identification of potential anti-obesity drug targets directed towards specific molecular pathways.
Summary
p53 is a transcriptional factor modulating numerous biological actions. Although it is best known for its role in cancer development, it is now evident that it is implicated in metabolism. More specifically, p53 modulates energy metabolism and homeostasis through their effects on adipocyte development and function. However, nothing is known about the potential metabolic function of p53 in the central nervous system.
Neuronal networks within the central nervous system play a crucial role in the regulation of food intake, body weight, and glucose homeostasis, so the main objective of this project will be to evaluate the potential of brain p53 as anti-obesity and/or anti-diabetic drug candidate. Our project will dissect precisely which specific components of energy balance are altered after central disruption or rescue of p53 signalling in selective neuronal populations, as well as the molecular pathways mediating these actions.
More precisely, we will disrupt the central p53 signalling specifically in hypothalamic POMC and AgRP neurons, which are crucial for energy and glucose homeostasis. We will also generate and characterize mice lacking p53 in dopamine neurons that are essential for mechanisms related with the reward of food. Once we know which specific areas are crucial for the central actions of p53, we will complete the experiments rescuing p53 expression in selective neuronal populations (POMC, AgRP or dopamine neurons) of p53 null mice. We will also investigate the interaction between p53 with leptin and ghrelin, likely the two more important hormones in the regulation of energy balance, which act through homeostatic and hedonic mechanisms. Understanding the precise role and mechanisms regulated by central p53 on energy balance may open new avenues for the identification of potential anti-obesity drug targets directed towards specific molecular pathways.
Max ERC Funding
1 477 680 €
Duration
Start date: 2011-12-01, End date: 2017-05-31
Project acronym OVER-HER2
Project OVErcoming Resistance to anti-HER2 therapy
Researcher (PI) Jose Manuel Baselga Torres
Host Institution (HI) FUNDACIO PRIVADA INSTITUT D'INVESTIGACIO ONCOLOGICA DE VALL-HEBRON
Call Details Advanced Grant (AdG), LS4, ERC-2009-AdG
Summary HER2 is a membrane receptor tyrosine kinase overexpressed in 30% of breast tumors and results in an aggressive clinical course. Anti-HER2 therapies including monoclonal antibodies (trastuzumab) and small-molecule tyrosine kinase inhibitors (lapatinib) are active and have improved survival of patients with HER2 overexpressing breast cancer. However, the emergence of primary or acquired resistance to these agents limits their efficacy. We have previously identified mechanisms of resistance to anti-HER2 therapies such as the co-expression of a truncated form of HER2 that correlates with trastuzumab resistance or the presence of downstream oncogenic mutations of PI3K or PTEN loss that result in resistance to lapatinib . Not surprisingly, PI3K/mTOR inhibitors overcome lapatinib resistance in the later example. Building on our results to date, this proposal is aimed at identifying novel mechanisms of resistance to anti-HER2 agents and to devise therapeutic strategies to revert it. To uncover such mechanisms, we have generated cancer cells with acquired resistance to lapatinib or trastuzumab by continuous exposure to increasing concentrations of these agents. We will perform genome wide screens, including shRNA libraries, gene expression and SNPs arrays, to discover candidate genes responsible for decreased sensitivity to anti-HER2 agents. To overcome anti-HER2 therapy resistance we will study several therapeutic strategies, such as combinations of different anti-HER2 compounds and the use of alternative agents targeting downstream/parallel pathways. Among the novel targeted therapies, we plan to study the use of PI3K, Akt, CDK2 and Hsp90 inhibitors, for which we will also start resistance-screens. It is anticipated that any promising preclinical leads will stimulate trial design and conduct for subsequent evaluation and confirmation in the clinic.
Summary
HER2 is a membrane receptor tyrosine kinase overexpressed in 30% of breast tumors and results in an aggressive clinical course. Anti-HER2 therapies including monoclonal antibodies (trastuzumab) and small-molecule tyrosine kinase inhibitors (lapatinib) are active and have improved survival of patients with HER2 overexpressing breast cancer. However, the emergence of primary or acquired resistance to these agents limits their efficacy. We have previously identified mechanisms of resistance to anti-HER2 therapies such as the co-expression of a truncated form of HER2 that correlates with trastuzumab resistance or the presence of downstream oncogenic mutations of PI3K or PTEN loss that result in resistance to lapatinib . Not surprisingly, PI3K/mTOR inhibitors overcome lapatinib resistance in the later example. Building on our results to date, this proposal is aimed at identifying novel mechanisms of resistance to anti-HER2 agents and to devise therapeutic strategies to revert it. To uncover such mechanisms, we have generated cancer cells with acquired resistance to lapatinib or trastuzumab by continuous exposure to increasing concentrations of these agents. We will perform genome wide screens, including shRNA libraries, gene expression and SNPs arrays, to discover candidate genes responsible for decreased sensitivity to anti-HER2 agents. To overcome anti-HER2 therapy resistance we will study several therapeutic strategies, such as combinations of different anti-HER2 compounds and the use of alternative agents targeting downstream/parallel pathways. Among the novel targeted therapies, we plan to study the use of PI3K, Akt, CDK2 and Hsp90 inhibitors, for which we will also start resistance-screens. It is anticipated that any promising preclinical leads will stimulate trial design and conduct for subsequent evaluation and confirmation in the clinic.
Max ERC Funding
1 666 700 €
Duration
Start date: 2011-01-01, End date: 2014-12-31
Project acronym OxygenSensing
Project Molecular mechanisms of acute oxygen sensing.
Researcher (PI) Jose Lopez Barneo
Host Institution (HI) UNIVERSIDAD DE SEVILLA
Call Details Advanced Grant (AdG), LS4, ERC-2014-ADG
Summary Oxygen (O2) is essential for life on Earth. This proposal deals with the study of the molecular mechanisms underlying acute O2 sensing by cells, a long-standing issue that is yet to be elucidated. In recent years, the discovery of hypoxia inducible transcription factors and their regulation by the O2-dependent hydroxylases has provided a solid framework for understanding genetic responses to sustained (chronic) hypoxia. However the mechanisms of acute O2 sensing, necessary for the activation of rapid, life-saving, compensatory respiratory and cardiovascular reflexes (e.g. hyperventilation and sympathetic activation), are unknown. While the primary goal of the project is to characterize the molecular mechanisms underlying acute O2 sensing by arterial chemoreceptors (carotid body –CB- and adrenal medulla –AM-), we will also extend our study to other organs (e.g. pulmonary and systemic arteries) of the homeostatic acute O2-sensing system. We will investigate the role of mitochondria, in particular complex I (MCI), in acute O2 sensing. Previous data from our group demonstrated that rotenone, a MCI blocker, selectively occludes responsiveness to hypoxia in CB cells. In addition, our unpublished data indicate that sensitivity to hypoxia (but not to other stimuli) is lost in mice with genetic disruption of MCI genes in CB and AM cells. We have shown that the adult CB is a plastic organ that contains a population of multipotent neural stem cells. Hence, another objective of the project is to study the role of these stem cells in CB modulation (over- or infra-activation), which may participate in the pathogenesis of diseases. In the past, our group has made seminal contributions to unveiling the cellular bases of arterial chemoreception. The discovery of stem cells in the CB and the generation of new genetically modified mouse models, put us in a leading position to elucidate the molecular bases of acute O2 sensing and their biomedical implications.
Summary
Oxygen (O2) is essential for life on Earth. This proposal deals with the study of the molecular mechanisms underlying acute O2 sensing by cells, a long-standing issue that is yet to be elucidated. In recent years, the discovery of hypoxia inducible transcription factors and their regulation by the O2-dependent hydroxylases has provided a solid framework for understanding genetic responses to sustained (chronic) hypoxia. However the mechanisms of acute O2 sensing, necessary for the activation of rapid, life-saving, compensatory respiratory and cardiovascular reflexes (e.g. hyperventilation and sympathetic activation), are unknown. While the primary goal of the project is to characterize the molecular mechanisms underlying acute O2 sensing by arterial chemoreceptors (carotid body –CB- and adrenal medulla –AM-), we will also extend our study to other organs (e.g. pulmonary and systemic arteries) of the homeostatic acute O2-sensing system. We will investigate the role of mitochondria, in particular complex I (MCI), in acute O2 sensing. Previous data from our group demonstrated that rotenone, a MCI blocker, selectively occludes responsiveness to hypoxia in CB cells. In addition, our unpublished data indicate that sensitivity to hypoxia (but not to other stimuli) is lost in mice with genetic disruption of MCI genes in CB and AM cells. We have shown that the adult CB is a plastic organ that contains a population of multipotent neural stem cells. Hence, another objective of the project is to study the role of these stem cells in CB modulation (over- or infra-activation), which may participate in the pathogenesis of diseases. In the past, our group has made seminal contributions to unveiling the cellular bases of arterial chemoreception. The discovery of stem cells in the CB and the generation of new genetically modified mouse models, put us in a leading position to elucidate the molecular bases of acute O2 sensing and their biomedical implications.
Max ERC Funding
2 843 750 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym P38CANCER
Project Signal integration and rewiring during tumor development
Researcher (PI) Manuel Angel Rodriguez Nebreda
Host Institution (HI) FUNDACIO INSTITUT DE RECERCA BIOMEDICA (IRB BARCELONA)
Call Details Advanced Grant (AdG), LS4, ERC-2011-ADG_20110310
Summary Cell fate decisions rely on signaling pathways that integrate external signals to coordinate specific intracellular programs. One of these pathways leads to the activation of p38α MAPK, which plays key roles in cell responses to many types of stresses as well as chemotherapeutic agents and oncogenes. Importantly, p38α acts in a cell context-specific and cell type-specific manner to integrate signals that affect cell proliferation, differentiation and survival. Evidence from mouse models and human cell lines indicates that p38α can negatively regulate tumor initiation at different levels. Intriguingly, recent results suggest that p38α activation may also sometimes have pro-tumorigenic functions. The molecular basis for the different functions of p38α are not well understood but it is likely that the network of substrates phosphorylated by p38α plays a major role. This project proposes to investigate molecular mechanisms of p38 MAPK signaling during tumorigenesis including the systematic identification of substrates and how they contribute to the different functions of this pathway. An important part of the studies will focus on the mechanisms underlying the rewiring of p38α signaling to serve pro-tumorigenic functions, including in-depth characterization of how p38α regulates the survival, proliferation and spreading of cancer cells, as well as its role in the interplay between cancer cells and stromal cells. We also plan to investigate the implication of p38α in tumor progression in vivo, using both xenografts and genetically modified mice that we have generated to either inactivate or hyperactivate the p38 MAPK pathway. These models will allow us to analyze the role of this signaling pathway in the regulation of tumor initiation, growth and spreading in different tissues. Taken together, this project will address important questions on cellular signaling and tumor development, which might be also useful for more rational anti-tumoral treatments.
Summary
Cell fate decisions rely on signaling pathways that integrate external signals to coordinate specific intracellular programs. One of these pathways leads to the activation of p38α MAPK, which plays key roles in cell responses to many types of stresses as well as chemotherapeutic agents and oncogenes. Importantly, p38α acts in a cell context-specific and cell type-specific manner to integrate signals that affect cell proliferation, differentiation and survival. Evidence from mouse models and human cell lines indicates that p38α can negatively regulate tumor initiation at different levels. Intriguingly, recent results suggest that p38α activation may also sometimes have pro-tumorigenic functions. The molecular basis for the different functions of p38α are not well understood but it is likely that the network of substrates phosphorylated by p38α plays a major role. This project proposes to investigate molecular mechanisms of p38 MAPK signaling during tumorigenesis including the systematic identification of substrates and how they contribute to the different functions of this pathway. An important part of the studies will focus on the mechanisms underlying the rewiring of p38α signaling to serve pro-tumorigenic functions, including in-depth characterization of how p38α regulates the survival, proliferation and spreading of cancer cells, as well as its role in the interplay between cancer cells and stromal cells. We also plan to investigate the implication of p38α in tumor progression in vivo, using both xenografts and genetically modified mice that we have generated to either inactivate or hyperactivate the p38 MAPK pathway. These models will allow us to analyze the role of this signaling pathway in the regulation of tumor initiation, growth and spreading in different tissues. Taken together, this project will address important questions on cellular signaling and tumor development, which might be also useful for more rational anti-tumoral treatments.
Max ERC Funding
2 497 800 €
Duration
Start date: 2012-07-01, End date: 2017-12-31
Project acronym PHYS.LSS
Project Cosmological Physics with future large-scale structure surveys
Researcher (PI) Licia Verde
Host Institution (HI) UNIVERSITAT DE BARCELONA
Call Details Starting Grant (StG), PE9, ERC-2009-StG
Summary Future, large galaxy surveys (such as BOSS, DES, LSST, EUCLID, ADEPT etc.) will cover of the order of 10000 square degrees on the sky, with the primary science goal to unravel the nature of the physics responsible for the current accelerated expansion of the universe. This acceleration likely involves new physics which could imply ether a modification of our understanding of particles and fields (if the acceleration is caused by a new negative pressure-component) or a change in our understanding of space and time (by modifying Einstein's General Relativity laws). The unprecedented and exquisite data provided by these surveys will make possible also other interesting science with implications for fundamental physics (e.g., inflation, neutrino properties) and astrophysics (e.g., biasing, galaxy formation). The success of future large-scale galaxy surveys evidently requires a correct interpretation of their data. The current proposal, which benefits from the interaction of Cosmology, astrophysics and particle physics, aims at building up a set of robust tools to maximize the physics extracted from large-scale structure data. Such an interplay is mandatory to ensure a suitable modeling of the observables and a meaningful comparison with the theoretical predictions. The PI is involved with surveys such as BOSS, ADEPT and LSST and for the past year has been leading a working group with the goal of bringing together particle physicists and cosmology to better understand dark energy. The methods developed in the proposal presented here are expected to be used by the international community involved in future surveys. This would imply a big step for Spanish groups joining or even leading future Cosmology or Astro-particle physics projects.
Summary
Future, large galaxy surveys (such as BOSS, DES, LSST, EUCLID, ADEPT etc.) will cover of the order of 10000 square degrees on the sky, with the primary science goal to unravel the nature of the physics responsible for the current accelerated expansion of the universe. This acceleration likely involves new physics which could imply ether a modification of our understanding of particles and fields (if the acceleration is caused by a new negative pressure-component) or a change in our understanding of space and time (by modifying Einstein's General Relativity laws). The unprecedented and exquisite data provided by these surveys will make possible also other interesting science with implications for fundamental physics (e.g., inflation, neutrino properties) and astrophysics (e.g., biasing, galaxy formation). The success of future large-scale galaxy surveys evidently requires a correct interpretation of their data. The current proposal, which benefits from the interaction of Cosmology, astrophysics and particle physics, aims at building up a set of robust tools to maximize the physics extracted from large-scale structure data. Such an interplay is mandatory to ensure a suitable modeling of the observables and a meaningful comparison with the theoretical predictions. The PI is involved with surveys such as BOSS, ADEPT and LSST and for the past year has been leading a working group with the goal of bringing together particle physicists and cosmology to better understand dark energy. The methods developed in the proposal presented here are expected to be used by the international community involved in future surveys. This would imply a big step for Spanish groups joining or even leading future Cosmology or Astro-particle physics projects.
Max ERC Funding
1 395 000 €
Duration
Start date: 2009-11-01, End date: 2015-10-31
Project acronym PI2FA
Project Partial Ionisation: Two-Fluid Approach
Researcher (PI) Olena KHOMENKO
Host Institution (HI) INSTITUTO DE ASTROFISICA DE CANARIAS
Call Details Consolidator Grant (CoG), PE9, ERC-2017-COG
Summary PI2FA proposal’s overarching aim is to make a major breakthrough in our understanding of the magnetised solar chromosphere under a novel frame of a multi-fluid plasma theory. Future large-aperture solar telescopes, EST and DKIST, will have among their primary focus observations of chromospheric magnetic fields. The correct interpretation of solar data requires sophisticated theories. The solar atmosphere is made of strongly stratified, weakly ionised and not completely collisionally coupled plasma. In the previous PI’s ERC SPIA project we opened a new research line and performed systematic investigations of non-ideal effects due to neutrals in the solar plasma. To build the complexity step by step, we advanced a single-fluid formalism, best valid for a strongly collisionally coupled case. Nevertheless, a multi-fluid treatment is essential for the weakly coupled chromosphere because the processes of the energy transport and conversion happen at nearly collisional scales. Now it is the right moment to take advantage and consolidate the experience gained in the SPIA project and to bring our research to a new level of challenge. The ambition of the PI2FA proposal is to create and apply tools for multi-dimensional modelling of the solar chromosphere under a precise two-fluid multi-species approach. In the recent few years it has been repeatedly demonstrated that processes related to non-ideal plasma behaviour due to neutrals may be the key to solve the problem of chromospheric heating and dynamics. PI2FA project will make progress in the following questions: determination of chromospheric heating mechanisms; understanding destabilization mechanisms of prominences related to neutrals, and creation of multi-dimensional two-fluid models of the solar chromosphere. These models will include altogether complex interactions down to smallest scales and allow direct comparison to observations, as a way to prepare our community for the coming large-aperture telescopes.
Summary
PI2FA proposal’s overarching aim is to make a major breakthrough in our understanding of the magnetised solar chromosphere under a novel frame of a multi-fluid plasma theory. Future large-aperture solar telescopes, EST and DKIST, will have among their primary focus observations of chromospheric magnetic fields. The correct interpretation of solar data requires sophisticated theories. The solar atmosphere is made of strongly stratified, weakly ionised and not completely collisionally coupled plasma. In the previous PI’s ERC SPIA project we opened a new research line and performed systematic investigations of non-ideal effects due to neutrals in the solar plasma. To build the complexity step by step, we advanced a single-fluid formalism, best valid for a strongly collisionally coupled case. Nevertheless, a multi-fluid treatment is essential for the weakly coupled chromosphere because the processes of the energy transport and conversion happen at nearly collisional scales. Now it is the right moment to take advantage and consolidate the experience gained in the SPIA project and to bring our research to a new level of challenge. The ambition of the PI2FA proposal is to create and apply tools for multi-dimensional modelling of the solar chromosphere under a precise two-fluid multi-species approach. In the recent few years it has been repeatedly demonstrated that processes related to non-ideal plasma behaviour due to neutrals may be the key to solve the problem of chromospheric heating and dynamics. PI2FA project will make progress in the following questions: determination of chromospheric heating mechanisms; understanding destabilization mechanisms of prominences related to neutrals, and creation of multi-dimensional two-fluid models of the solar chromosphere. These models will include altogether complex interactions down to smallest scales and allow direct comparison to observations, as a way to prepare our community for the coming large-aperture telescopes.
Max ERC Funding
1 968 750 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym PLANT CIRES BIOTECH
Project Functional characterization of plant cellular IRES in response to abiotic stress and their use as biotechnological tools
Researcher (PI) María Del Mar Castellano
Host Institution (HI) INSTITUTO NACIONAL DE INVESTIGACION Y TECNOLOGIA AGRARIA Y ALIMENTARIA OA MP
Call Details Starting Grant (StG), LS9, ERC-2010-StG_20091118
Summary To cope with abiotic stresses plants require an extensive molecular regulation of gene expression. In plants, translation is a key step in the control of gene expression under abiotic stress conditions. This translational regulation involves (1) a global inhibition of protein synthesis and (2) an efficient and selective translation of certain mRNAs, generally codifying proteins involved in the abiotic stress response. Although in plants the mechanisms involved in the onset of this dual regulation are currently unknown, some evidences point out that cap independent translation, via recognition of internal ribosome entry sites (IRES) within the mRNAs efficiently translated, could be the clue for the selective protein synthesis observed under such conditions.
In this proposal we aim to further characterize the cellular IRESs operating under abiotic stress conditions in plants and to exploit the identified cellular IRESs as biotechnological tools to allow the efficient and selective translation of mRNAs of interest under abiotic stress conditions. In plants, no IRES trans-acting factors (ITAFs) and only two cellular IRESs have been identified so far. Therefore, the systematic identification of new cellular IRESs, the identification for the first time of ITAFs and the study of how they can control IRES activity-specificity under abiotic stress conditions are important steps forward in the knowledge of how plants adapt to environmental stresses. In addition, the pioneering use of the identified cellular IRESs as a tool to tightly and specifically control the expression of proteins of interest under abiotic stress conditions will open up a new perspective for the study of abiotic stress in plants and for the generation of plants with increased tolerance to such conditions.
Summary
To cope with abiotic stresses plants require an extensive molecular regulation of gene expression. In plants, translation is a key step in the control of gene expression under abiotic stress conditions. This translational regulation involves (1) a global inhibition of protein synthesis and (2) an efficient and selective translation of certain mRNAs, generally codifying proteins involved in the abiotic stress response. Although in plants the mechanisms involved in the onset of this dual regulation are currently unknown, some evidences point out that cap independent translation, via recognition of internal ribosome entry sites (IRES) within the mRNAs efficiently translated, could be the clue for the selective protein synthesis observed under such conditions.
In this proposal we aim to further characterize the cellular IRESs operating under abiotic stress conditions in plants and to exploit the identified cellular IRESs as biotechnological tools to allow the efficient and selective translation of mRNAs of interest under abiotic stress conditions. In plants, no IRES trans-acting factors (ITAFs) and only two cellular IRESs have been identified so far. Therefore, the systematic identification of new cellular IRESs, the identification for the first time of ITAFs and the study of how they can control IRES activity-specificity under abiotic stress conditions are important steps forward in the knowledge of how plants adapt to environmental stresses. In addition, the pioneering use of the identified cellular IRESs as a tool to tightly and specifically control the expression of proteins of interest under abiotic stress conditions will open up a new perspective for the study of abiotic stress in plants and for the generation of plants with increased tolerance to such conditions.
Max ERC Funding
1 237 500 €
Duration
Start date: 2010-12-01, End date: 2017-05-31
Project acronym PLANTGROWTH
Project Exploiting genome replication to design improved plant growth strategies
Researcher (PI) Crisanto GUTIERREZ
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Advanced Grant (AdG), LS9, ERC-2018-ADG
Summary This project will identify the principles governing genome replication in relation to the chromatin landscape and how they impact on plant organ growth. The results will provide the basis to design novel strategies to improve plant growth performance.
The large plant genomes, as in all eukaryotes, must be faithfully duplicated every cell cycle, a process regulated at the level of DNA replication origins (ORIs). Our understanding of how ORIs are determined is still very limited. Most of our knowledge comes from cultured cells, precluding the identification of regulatory layers operating at the organism level. Importantly, genome replication can offer unexplored possibilities to modulate plant architecture and growth and, consequently, plant performance.
Results generated so far unable us to address a fundamental question: what are the regulatory mechanisms of DNA and genome replication and how they can be exploited to design improved plant growth strategies. This innovative perspective will reveal how genome replication is regulated by DNA sequence context, replication factors and chromatin landscape. Integration of molecular, cellular, genomic and genetic approaches in a whole organism will serve to evaluate the phenotypic effects of modulating genome replication on organ growth. We will also learn how DNA replication control is exerted during endoreplication and in coordination with transcriptional programs, both crucial for plant organogenesis, growth and response to environmental stresses.
This program goes beyond incremental research, is timely, innovative, ambitious but realistic, and high risk/high gain, combining different approaches to address a fundamental process. Given the conservation of proteins and pathways, and the availability of well-annotated genomic information for many plant species, PLANTGROWTH will pave the way to translate the technological and conceptual know-how derived from this program to crop species to improve yield.
Summary
This project will identify the principles governing genome replication in relation to the chromatin landscape and how they impact on plant organ growth. The results will provide the basis to design novel strategies to improve plant growth performance.
The large plant genomes, as in all eukaryotes, must be faithfully duplicated every cell cycle, a process regulated at the level of DNA replication origins (ORIs). Our understanding of how ORIs are determined is still very limited. Most of our knowledge comes from cultured cells, precluding the identification of regulatory layers operating at the organism level. Importantly, genome replication can offer unexplored possibilities to modulate plant architecture and growth and, consequently, plant performance.
Results generated so far unable us to address a fundamental question: what are the regulatory mechanisms of DNA and genome replication and how they can be exploited to design improved plant growth strategies. This innovative perspective will reveal how genome replication is regulated by DNA sequence context, replication factors and chromatin landscape. Integration of molecular, cellular, genomic and genetic approaches in a whole organism will serve to evaluate the phenotypic effects of modulating genome replication on organ growth. We will also learn how DNA replication control is exerted during endoreplication and in coordination with transcriptional programs, both crucial for plant organogenesis, growth and response to environmental stresses.
This program goes beyond incremental research, is timely, innovative, ambitious but realistic, and high risk/high gain, combining different approaches to address a fundamental process. Given the conservation of proteins and pathways, and the availability of well-annotated genomic information for many plant species, PLANTGROWTH will pave the way to translate the technological and conceptual know-how derived from this program to crop species to improve yield.
Max ERC Funding
2 497 800 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym PLEIO-RANK
Project Pleiotropic treatment of cancer: RANK inhibitors targeting cancer stem cells and immunity
Researcher (PI) Eva Gonzalez suarez
Host Institution (HI) FUNDACIO INSTITUT D'INVESTIGACIO BIOMEDICA DE BELLVITGE
Call Details Consolidator Grant (CoG), LS4, ERC-2015-CoG
Summary "Thousands of cancer patients worldwide are taking RANKL inhibitors for the management of bone metastasis, based on the key role of RANKL and its receptor, RANK, in osteoclasts. RANK signaling has multiple divergent effects in immunity and inflammation, both in the generation of active immune responses, as well as in the induction of tolerance. We showed that RANK overexpression induces stemness and interferes with differentiation in non transformed mammary epithelial cells and promotes mammary tumorigenesis, acting as a paracrine mediator of progesterone.
However, the therapeutic potential of inhibiting RANK signaling once tumors develop and its effects on tumor immunity remain unexplored. Our proposal tackles novel concepts: Is RANK a better therapeutic target than RANKL? Does RANK induce ""stemness"" in other epithelia and solid tumors and how? Does RANK regulate the tumor-immune cell crosstalk? Would inhibition of RANK signaling in tumor and immune cells result in synergistic or opposing effects on tumor outcome?
We hypotesize that RANK activation in solid tumors expands the cancer stem cells pool and induces an immnunosuppressive environment leading to tumor recurrence and metastasis.
In PLEIO-RANK we aim to:
1. Define the contribution of RANK to the epithelial hierarchy in mammary, skin and colon, during homeostasis and tumorigenesis, undertaking lineage tracing approaches.
2. Dissect the impact of RANK loss in the epithelial or the immune compartment in tumor outcome, exploiting tissue inducible models, in breast cancer and solid tumors driven by chronic inflammation.
3. Validate the clinical implications of our findings using patient derived xenografts and human tumor samples.
Based on the results of our proposal RANK inhibition could become a unique targeted therapy able to reduce metastasis and mortality in solid tumors for its pleiotropic antitumor effects in cancer stem cells, immune cells and their crosstalk.
"
Summary
"Thousands of cancer patients worldwide are taking RANKL inhibitors for the management of bone metastasis, based on the key role of RANKL and its receptor, RANK, in osteoclasts. RANK signaling has multiple divergent effects in immunity and inflammation, both in the generation of active immune responses, as well as in the induction of tolerance. We showed that RANK overexpression induces stemness and interferes with differentiation in non transformed mammary epithelial cells and promotes mammary tumorigenesis, acting as a paracrine mediator of progesterone.
However, the therapeutic potential of inhibiting RANK signaling once tumors develop and its effects on tumor immunity remain unexplored. Our proposal tackles novel concepts: Is RANK a better therapeutic target than RANKL? Does RANK induce ""stemness"" in other epithelia and solid tumors and how? Does RANK regulate the tumor-immune cell crosstalk? Would inhibition of RANK signaling in tumor and immune cells result in synergistic or opposing effects on tumor outcome?
We hypotesize that RANK activation in solid tumors expands the cancer stem cells pool and induces an immnunosuppressive environment leading to tumor recurrence and metastasis.
In PLEIO-RANK we aim to:
1. Define the contribution of RANK to the epithelial hierarchy in mammary, skin and colon, during homeostasis and tumorigenesis, undertaking lineage tracing approaches.
2. Dissect the impact of RANK loss in the epithelial or the immune compartment in tumor outcome, exploiting tissue inducible models, in breast cancer and solid tumors driven by chronic inflammation.
3. Validate the clinical implications of our findings using patient derived xenografts and human tumor samples.
Based on the results of our proposal RANK inhibition could become a unique targeted therapy able to reduce metastasis and mortality in solid tumors for its pleiotropic antitumor effects in cancer stem cells, immune cells and their crosstalk.
"
Max ERC Funding
1 999 960 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym POLMAG
Project Polarized Radiation Diagnostics for Exploring the Magnetism of the Outer Solar Atmosphere
Researcher (PI) Javier Trujillo Bueno
Host Institution (HI) INSTITUTO DE ASTROFISICA DE CANARIAS
Call Details Advanced Grant (AdG), PE9, ERC-2016-ADG
Summary POLMAG aims at a true breakthrough in the development and application of polarized radiation diagnostic methods for exploring the magnetic fields of the chromosphere, transition region and corona of the Sun via the interpretation of the Stokes profiles produced by optically polarized atoms and the Hanle and Zeeman effects in ultraviolet (UV), visible and near-infrared spectral lines. To this end, POLMAG will combine and expand expertise on atomic physics, on the quantum theory of radiation, on high-precision spectropolarimetry, on advanced methods in numerical radiative transfer, and on the confrontation of spectropolarimetric observations with spectral synthesis in increasingly realistic three-dimensional (3D) numerical models of the solar atmosphere.
POLMAG targets the following very challenging issues:
- Which are the optimum spectral lines for probing the magnetism of the outer solar atmosphere?
- How to compute efficiently the Stokes profiles taking into account partial frequency redistribution, J-state quantum interference and the Hanle and Zeeman effects?
- How to determine the magnetic, thermal and dynamic structure of the outer solar atmosphere through confrontations with spectropolarimetric observations?
POLMAG will go well beyond the current state of the art as follows:
- Applying and extending the quantum theory of light polarization
- Developing and applying efficient radiative transfer codes
- Modeling the Ly-alpha and Mg II h & k observations of our CLASP suborbital rocket experiments
- Developing novel coronal magnetometry methods by complementing for the first time the information provided by forbidden and permitted lines
- Developing the plasma diagnostic techniques needed for the scientific exploitation of spectropolarimetric observations with the new generation of solar telescopes and putting them at the disposal of the astrophysical community
POLMAG will open up a new diagnostic window in astrophysics.
Summary
POLMAG aims at a true breakthrough in the development and application of polarized radiation diagnostic methods for exploring the magnetic fields of the chromosphere, transition region and corona of the Sun via the interpretation of the Stokes profiles produced by optically polarized atoms and the Hanle and Zeeman effects in ultraviolet (UV), visible and near-infrared spectral lines. To this end, POLMAG will combine and expand expertise on atomic physics, on the quantum theory of radiation, on high-precision spectropolarimetry, on advanced methods in numerical radiative transfer, and on the confrontation of spectropolarimetric observations with spectral synthesis in increasingly realistic three-dimensional (3D) numerical models of the solar atmosphere.
POLMAG targets the following very challenging issues:
- Which are the optimum spectral lines for probing the magnetism of the outer solar atmosphere?
- How to compute efficiently the Stokes profiles taking into account partial frequency redistribution, J-state quantum interference and the Hanle and Zeeman effects?
- How to determine the magnetic, thermal and dynamic structure of the outer solar atmosphere through confrontations with spectropolarimetric observations?
POLMAG will go well beyond the current state of the art as follows:
- Applying and extending the quantum theory of light polarization
- Developing and applying efficient radiative transfer codes
- Modeling the Ly-alpha and Mg II h & k observations of our CLASP suborbital rocket experiments
- Developing novel coronal magnetometry methods by complementing for the first time the information provided by forbidden and permitted lines
- Developing the plasma diagnostic techniques needed for the scientific exploitation of spectropolarimetric observations with the new generation of solar telescopes and putting them at the disposal of the astrophysical community
POLMAG will open up a new diagnostic window in astrophysics.
Max ERC Funding
2 478 750 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym ProNANO
Project Protein-based functional nanostructures
Researcher (PI) Aitziber Lopez Cortajarena
Host Institution (HI) ASOCIACION CENTRO DE INVESTIGACION COOPERATIVA EN BIOMATERIALES- CIC biomaGUNE
Call Details Consolidator Grant (CoG), LS9, ERC-2014-CoG
Summary The precise synthesis of nano-devices with tailored complex structures and properties is a requisite for their use in nanotechnology and medicine. Nowadays, the technology for the generation of these devices lacks the precision to determine their properties, and is accomplished mostly by “trial and error” experimental approaches. Bottom-up self-assembly that relies on highly specific biomolecular interactions of small and simple components, is an attractive approach for nanostructure templating.
Here, we propose to overcome aforementioned challenges by using self-assembling protein building blocks as templates for nanofabrication. In nature, protein assemblies govern sophisticated structures and functions, which are inspiration to engineer novel assemblies by exploiting the same set of tools and interactions to create nanostructures with numerous potential applications in synthetic biology and nanotechnology.
We hypothesize that we can rationally assemble a variety functional nanostructures by the logical combination of simple protein building blocks with specified properties. We propose to use a designed repeat protein scaffold for which we acquired a deep understanding of its molecular structure, stability, function, and inherent assembly properties. Only few conserved residues define the structure of the building block, which allow us to mutate its sequence to modulate assembly properties and to introduce reactive functionalities without compromising the structure of the scaffolding molecule.
First, we will design a collection of protein-based nanostructures. Then, we will introduce reactive functionalities to create hybrid nanostructures with nanoparticles, metals and electro-active molecules. Finally, these conjugates will be used to build nano-devices such as nanocircuits, catalysts and electroactive materials.
The outcome of this project will be a modular versatile platform for the fabrication of multiple protein-based hybrid functional nanostructures.
Summary
The precise synthesis of nano-devices with tailored complex structures and properties is a requisite for their use in nanotechnology and medicine. Nowadays, the technology for the generation of these devices lacks the precision to determine their properties, and is accomplished mostly by “trial and error” experimental approaches. Bottom-up self-assembly that relies on highly specific biomolecular interactions of small and simple components, is an attractive approach for nanostructure templating.
Here, we propose to overcome aforementioned challenges by using self-assembling protein building blocks as templates for nanofabrication. In nature, protein assemblies govern sophisticated structures and functions, which are inspiration to engineer novel assemblies by exploiting the same set of tools and interactions to create nanostructures with numerous potential applications in synthetic biology and nanotechnology.
We hypothesize that we can rationally assemble a variety functional nanostructures by the logical combination of simple protein building blocks with specified properties. We propose to use a designed repeat protein scaffold for which we acquired a deep understanding of its molecular structure, stability, function, and inherent assembly properties. Only few conserved residues define the structure of the building block, which allow us to mutate its sequence to modulate assembly properties and to introduce reactive functionalities without compromising the structure of the scaffolding molecule.
First, we will design a collection of protein-based nanostructures. Then, we will introduce reactive functionalities to create hybrid nanostructures with nanoparticles, metals and electro-active molecules. Finally, these conjugates will be used to build nano-devices such as nanocircuits, catalysts and electroactive materials.
The outcome of this project will be a modular versatile platform for the fabrication of multiple protein-based hybrid functional nanostructures.
Max ERC Funding
1 718 850 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym RAS AHEAD
Project Ras Genes in Health and Disease
Researcher (PI) Mariano Barbacid Montalban
Host Institution (HI) FUNDACION CENTRO NACIONAL DE INVESTIGACIONES ONCOLOGICAS CARLOS III
Call Details Advanced Grant (AdG), LS4, ERC-2009-AdG
Summary Ras genes are some of the best-studied genes in biomedical research due to their central role in mitogenic signaling and their oncogenic activation in one third of all human tumors. More recently, Ras genes have also been implicated in developmental disorders including Costello and Noonan syndromes. In spite of this wealth of information, we still do not know the role of Ras proteins in adult homeostasis and, more importantly, how their misregulation affects human health. The latter is of paramount importance in order to develop efficacious therapies against tumors carrying Ras oncogenes - an achievement that could save thousands of lives worldwide. We propose to address these issues using genetic approaches in mouse models. We aim to systemically ablate all Ras genes in adult mice as well as in selective tissues to understand their role in normal homeostasis. We also propose to characterize mouse models for developmental disorders induced by hyperactive Ras proteins. These models should help us to better understand these human disorders as well as tools to test potential therapeutic strategies. Finally, we propose to use K-Ras driven mouse tumor models for human PDA and NSCLC to address key questions that may be directly translated to the clinic. In the case of PDA, we propose to study the contribution of the inflammatory response induced by pancreatitis to tumor development. In the case of NCSLC, we propose to isolate cancer initiating cells in an attempt to reveal the earliest events in tumor development. Moreover, we intend to use this tumor model to validate druggable Ras downstream effectors as therapeutic targets. The results derived from these studies should provide key information to design forthcoming clinical trials that will benefit cancer patients.
Summary
Ras genes are some of the best-studied genes in biomedical research due to their central role in mitogenic signaling and their oncogenic activation in one third of all human tumors. More recently, Ras genes have also been implicated in developmental disorders including Costello and Noonan syndromes. In spite of this wealth of information, we still do not know the role of Ras proteins in adult homeostasis and, more importantly, how their misregulation affects human health. The latter is of paramount importance in order to develop efficacious therapies against tumors carrying Ras oncogenes - an achievement that could save thousands of lives worldwide. We propose to address these issues using genetic approaches in mouse models. We aim to systemically ablate all Ras genes in adult mice as well as in selective tissues to understand their role in normal homeostasis. We also propose to characterize mouse models for developmental disorders induced by hyperactive Ras proteins. These models should help us to better understand these human disorders as well as tools to test potential therapeutic strategies. Finally, we propose to use K-Ras driven mouse tumor models for human PDA and NSCLC to address key questions that may be directly translated to the clinic. In the case of PDA, we propose to study the contribution of the inflammatory response induced by pancreatitis to tumor development. In the case of NCSLC, we propose to isolate cancer initiating cells in an attempt to reveal the earliest events in tumor development. Moreover, we intend to use this tumor model to validate druggable Ras downstream effectors as therapeutic targets. The results derived from these studies should provide key information to design forthcoming clinical trials that will benefit cancer patients.
Max ERC Funding
2 496 192 €
Duration
Start date: 2010-04-01, End date: 2015-03-31
Project acronym SENTIFLEX
Project Fluorescence-based photosynthesis estimates for vegetation productivity monitoring from space
Researcher (PI) Jochem VERRELST
Host Institution (HI) UNIVERSITAT DE VALENCIA
Call Details Starting Grant (StG), LS9, ERC-2017-STG
Summary Global food security will remain a worldwide concern for the next 50 years and beyond. Agricultural production undergoes an increasing pressure by global anthropogenic changes, including rising population, increased protein demands and climatic extremes. Because of the immediate and dynamic nature of these changes, productivity monitoring measures are urgently needed to ensure both the stability and continued increase of the global food supply. Europe has expressed ambitions to keep its fingers on the pulse of its agricultural lands. In response to that, this proposal - named SENTIFLEX - is dedicated to developing a European vegetation productivity monitoring facility based on the synergy of Sentinel-3 (S3) with FLEX satellite fluorescence data. ESA's 8th Earth Explorer FLEX is the first mission specifically designed to globally measure Sun-Induced chlorophyll Fluorescence (SIF) emission from terrestrial vegetation. These two European Earth observation missions offer immense possibilities to increase our knowledge of the basic functioning of the Earth’s vegetation, i.e., the photosynthetic activity of plants resulting in carbon fixation. Two complementary approaches are envisioned to realize quantification of photosynthesis through satellite SIF and S3. First, the work seeks to advance the science in establishing and consolidating relationships between canopy-leaving SIF and unbiased estimates of photosynthesis of the plants, thereby disentangling the role of dynamic vegetative and atmospheric variables. Second, consolidated relationships between SIF and photosynthesis will be used to build a FLEX-S3 data processing assimilation scheme through process-based vegetation models that will deliver spatiotemporally highly resolved information on Europe’s vegetation productivity. To streamline all these datasets into a prototype vegetation productivity monitoring facility, new data processing concepts will be introduced such as the emulation of radiative transfer models.
Summary
Global food security will remain a worldwide concern for the next 50 years and beyond. Agricultural production undergoes an increasing pressure by global anthropogenic changes, including rising population, increased protein demands and climatic extremes. Because of the immediate and dynamic nature of these changes, productivity monitoring measures are urgently needed to ensure both the stability and continued increase of the global food supply. Europe has expressed ambitions to keep its fingers on the pulse of its agricultural lands. In response to that, this proposal - named SENTIFLEX - is dedicated to developing a European vegetation productivity monitoring facility based on the synergy of Sentinel-3 (S3) with FLEX satellite fluorescence data. ESA's 8th Earth Explorer FLEX is the first mission specifically designed to globally measure Sun-Induced chlorophyll Fluorescence (SIF) emission from terrestrial vegetation. These two European Earth observation missions offer immense possibilities to increase our knowledge of the basic functioning of the Earth’s vegetation, i.e., the photosynthetic activity of plants resulting in carbon fixation. Two complementary approaches are envisioned to realize quantification of photosynthesis through satellite SIF and S3. First, the work seeks to advance the science in establishing and consolidating relationships between canopy-leaving SIF and unbiased estimates of photosynthesis of the plants, thereby disentangling the role of dynamic vegetative and atmospheric variables. Second, consolidated relationships between SIF and photosynthesis will be used to build a FLEX-S3 data processing assimilation scheme through process-based vegetation models that will deliver spatiotemporally highly resolved information on Europe’s vegetation productivity. To streamline all these datasets into a prototype vegetation productivity monitoring facility, new data processing concepts will be introduced such as the emulation of radiative transfer models.
Max ERC Funding
1 499 587 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym SPIA
Project Magnetic connectivity through the Solar Partially Ionized Atmosphere
Researcher (PI) Olena Khomenko
Host Institution (HI) INSTITUTO DE ASTROFISICA DE CANARIAS
Call Details Starting Grant (StG), PE9, ERC-2011-StG_20101014
Summary The broad scientific objective of the SPIA proposal is to understand the magnetism of the Sun and stars and to establish connections between the magnetic activity in sub-surface layers and its manifestation in the outer atmosphere. The complex interactions in magnetized stellar plasmas are best studied via numerical simulations, a new powerful method of research that appeared in astrophysics with the development of large supercomputer facilities. With a coming era of large aperture solar telescopes, ATST and EST, spectropolarimetric observations of the Sun will become available at extraordinary high spatial and temporal resolutions. New modelling tools are required to understand the plasma behaviour at these scales. I propose to consolidate a research group of bright scientists around the PI to explore a novel promising approach for the description solar atmospheric plasma under multi-fluid approximation. The degree of plasma ionization in the photosphere and chromosphere of the Sun is extremely low and significant deviations from the classical magneto-hydrodynamic description are expected. A major development of the SPIA proposal will be the implementation of a multi-fluid plasma description, appropriate for a partially ionized medium, relaxing approximations of classical magneto-hydrodynamics. With the inclusion of standard radiative transfer into the three-dimensional multi-fluid code to be developed by the project team, it will be possible to perform simulations of solar sub-photospheric and photospheric regions, up to the low chromosphere, with a realism not achieved before. The importance of the non-ideal plasma effect for the energy balance of the solar chromosphere will be evaluated, and three-dimensional time-dependent models of multi-fluid magneto-convection will be created. This effort will produce a significant step toward the solution of the long-standing question of the origin of solar chromosphere, one of the most poorly understood regions of the Sun.
Summary
The broad scientific objective of the SPIA proposal is to understand the magnetism of the Sun and stars and to establish connections between the magnetic activity in sub-surface layers and its manifestation in the outer atmosphere. The complex interactions in magnetized stellar plasmas are best studied via numerical simulations, a new powerful method of research that appeared in astrophysics with the development of large supercomputer facilities. With a coming era of large aperture solar telescopes, ATST and EST, spectropolarimetric observations of the Sun will become available at extraordinary high spatial and temporal resolutions. New modelling tools are required to understand the plasma behaviour at these scales. I propose to consolidate a research group of bright scientists around the PI to explore a novel promising approach for the description solar atmospheric plasma under multi-fluid approximation. The degree of plasma ionization in the photosphere and chromosphere of the Sun is extremely low and significant deviations from the classical magneto-hydrodynamic description are expected. A major development of the SPIA proposal will be the implementation of a multi-fluid plasma description, appropriate for a partially ionized medium, relaxing approximations of classical magneto-hydrodynamics. With the inclusion of standard radiative transfer into the three-dimensional multi-fluid code to be developed by the project team, it will be possible to perform simulations of solar sub-photospheric and photospheric regions, up to the low chromosphere, with a realism not achieved before. The importance of the non-ideal plasma effect for the energy balance of the solar chromosphere will be evaluated, and three-dimensional time-dependent models of multi-fluid magneto-convection will be created. This effort will produce a significant step toward the solution of the long-standing question of the origin of solar chromosphere, one of the most poorly understood regions of the Sun.
Max ERC Funding
969 600 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym STEM-AGING
Project Tissue regeneration and aging: the decisive quiescent stem-cell state
Researcher (PI) Purificación MUNOZ-CANOVES
Host Institution (HI) UNIVERSIDAD POMPEU FABRA
Call Details Advanced Grant (AdG), LS4, ERC-2016-ADG
Summary The basic mechanisms of stem cell malfunction during aging are poorly understood even though they underlie the regenerative decline of most organs and tissues as we age. Based on our recent contributions (Nature 2014, Nature 2016), the fields of tissue regeneration and aging converge on the key role of the quiescent state, the preferred state of stem cells in low turnover tissues such as skeletal muscle. Our unifying hypothesis is that stem-cell quiescence maintenance, which requires active proteostasis (protein homeostasis), lies at the basis of stemness, and that its substitution by a senescence state in aging impairs regeneration. How these variables connect to drive stem cell aging is not known. Crucial experimental systems in this proposal are sensitive reporter mice for proteostasis, senescence and quiescence/fate in aging muscle stem cells. The project is divided as follows: Objective 1. Proteostasis and stem cell quiescence maintenance: tracing proteostasis in quiescent stem cells from autophagy and chaperone-mediated autophagy (CMA) reporter mice during aging / impact of autophagy/CMA loss on quiescence and regeneration / molecular regulators of proteostasis. Objective 2. Proteostasis and quiescent stem cell heterogeneity and fate: asymmetric segregation of proteotoxic waste as an instructor of stem cell heterogeneity and regenerative fate. Objective 3. The quiescence-to-senescence-switch in aging muscle stem cells: tracing and isolating senescent stem cells in senescence-cell reporter mice during aging / impact of senescent cell ablation on regenerating aged muscle. Objective 4. Circadian regulation in the quiescent stem-cell state: impact of aging on circadian rhythms and consequences for quiescence maintenance and regeneration. We expect that completion of these objectives will provide new fundamental knowledge on stem-cell biology, regeneration and aging.
Summary
The basic mechanisms of stem cell malfunction during aging are poorly understood even though they underlie the regenerative decline of most organs and tissues as we age. Based on our recent contributions (Nature 2014, Nature 2016), the fields of tissue regeneration and aging converge on the key role of the quiescent state, the preferred state of stem cells in low turnover tissues such as skeletal muscle. Our unifying hypothesis is that stem-cell quiescence maintenance, which requires active proteostasis (protein homeostasis), lies at the basis of stemness, and that its substitution by a senescence state in aging impairs regeneration. How these variables connect to drive stem cell aging is not known. Crucial experimental systems in this proposal are sensitive reporter mice for proteostasis, senescence and quiescence/fate in aging muscle stem cells. The project is divided as follows: Objective 1. Proteostasis and stem cell quiescence maintenance: tracing proteostasis in quiescent stem cells from autophagy and chaperone-mediated autophagy (CMA) reporter mice during aging / impact of autophagy/CMA loss on quiescence and regeneration / molecular regulators of proteostasis. Objective 2. Proteostasis and quiescent stem cell heterogeneity and fate: asymmetric segregation of proteotoxic waste as an instructor of stem cell heterogeneity and regenerative fate. Objective 3. The quiescence-to-senescence-switch in aging muscle stem cells: tracing and isolating senescent stem cells in senescence-cell reporter mice during aging / impact of senescent cell ablation on regenerating aged muscle. Objective 4. Circadian regulation in the quiescent stem-cell state: impact of aging on circadian rhythms and consequences for quiescence maintenance and regeneration. We expect that completion of these objectives will provide new fundamental knowledge on stem-cell biology, regeneration and aging.
Max ERC Funding
2 499 949 €
Duration
Start date: 2017-11-01, End date: 2022-10-31
Project acronym STEMCLOCK
Project Spatiotemporal regulation of epidermal stem cells by circadian rhythms: impact on homeostasis and aging
Researcher (PI) Salvador Aznar Benitah
Host Institution (HI) FUNDACIO INSTITUT DE RECERCA BIOMEDICA (IRB BARCELONA)
Call Details Starting Grant (StG), LS4, ERC-2012-StG_20111109
Summary "Most adult stem cells are compartmentalized in functionally deterministic niches where they self-renew and maintain homeostasis. From there, stem cells are instructed by combinations of signals and spatial tensile forces which they translate into a specific behavior. However how stem cells spatiotemporally coordinate their stem cell potential with niche- and systemic cues is poorly understood. These issues are essential since perturbations in stem cell function can cause tissue malfunction, such as tumorigenesis and aging.
We propose to perform a systematic analysis to identify the molecular causes that underlie epidermal stem cell aging. We will focus on the interplay between circadian rhythms and stem cell function. The circadian machinery anticipates and synchronizes the daily function of tissues according to the entrainment by natural changes in light and metabolism. We have shown that the molecular clock fine-tunes the behavior of epidermal stem cells by imposing oscillations in the expression of stem cell regulatory genes. These oscillations provide stem cells with a spatiotemporal axis for responding to dormancy, activating, and differentiation cues. Notably, the stem cell clock is naturally dampened upon aging, and forced circadian arrhythmia causes severe epidermal aging and predisposition to tumorigenesis.
We now propose to understand how the circadian clock coordinates the communication between stem cells with local and systemic cues, and how these are perturbed during aging. Specifically we aim: i) To study whether circadian rhythms coordinate the function of niche cells and epidermal stem cells; ii) To identify the molecular causes underlying the age-related dampening of the stem cell clock. We will combine large-scale genomic data, mouse models of circadian arrhythmia, and bioinformatic analysis. We hope to unveil some of the molecular causes underlying the loss of communication between epidermal stem cells and their environment resulting in aging."
Summary
"Most adult stem cells are compartmentalized in functionally deterministic niches where they self-renew and maintain homeostasis. From there, stem cells are instructed by combinations of signals and spatial tensile forces which they translate into a specific behavior. However how stem cells spatiotemporally coordinate their stem cell potential with niche- and systemic cues is poorly understood. These issues are essential since perturbations in stem cell function can cause tissue malfunction, such as tumorigenesis and aging.
We propose to perform a systematic analysis to identify the molecular causes that underlie epidermal stem cell aging. We will focus on the interplay between circadian rhythms and stem cell function. The circadian machinery anticipates and synchronizes the daily function of tissues according to the entrainment by natural changes in light and metabolism. We have shown that the molecular clock fine-tunes the behavior of epidermal stem cells by imposing oscillations in the expression of stem cell regulatory genes. These oscillations provide stem cells with a spatiotemporal axis for responding to dormancy, activating, and differentiation cues. Notably, the stem cell clock is naturally dampened upon aging, and forced circadian arrhythmia causes severe epidermal aging and predisposition to tumorigenesis.
We now propose to understand how the circadian clock coordinates the communication between stem cells with local and systemic cues, and how these are perturbed during aging. Specifically we aim: i) To study whether circadian rhythms coordinate the function of niche cells and epidermal stem cells; ii) To identify the molecular causes underlying the age-related dampening of the stem cell clock. We will combine large-scale genomic data, mouse models of circadian arrhythmia, and bioinformatic analysis. We hope to unveil some of the molecular causes underlying the loss of communication between epidermal stem cells and their environment resulting in aging."
Max ERC Funding
1 495 484 €
Duration
Start date: 2013-01-01, End date: 2017-12-31
Project acronym STROMALIGN
Project Mechanisms and Targeting Stromal Contribution to Tumour Invasion and Metastasis
Researcher (PI) Oriol Casanovas Casanovas
Host Institution (HI) INSTITUT CATALA D'ONCOLOGIA
Call Details Starting Grant (StG), LS4, ERC-2011-StG_20101109
Summary Angiogenesis inhibition has proven to be a successful anti-cancer therapeutic approach and anti-angiogenic therapies are currently approved as standard therapy in several types of cancer for their clinically validated beneficial extension of overall survival, progression-free survival and/or time-to-progression in these cancer patients. Nevertheless, in preclinical mouse models of cancer, although these therapies also show significant anti-tumour effects and overall survival benefit, these therapies are also triggering increased tumour local invasion, with more distant dissemination and emergence of metastasis (overall tumour malignization). The clinical relevance of this malignization effect in patients that are currently being treated with antiangiogenic drugs still remains elusive, but the frequent tumour relapses and acquired resistance to therapy in some patients strongly suggests this insidious event should be exhaustively evaluated.
In this sense, many laboratories are rapidly advancing in the study of the molecular players and signalling pathways implicated in this event, although an important limitation of all these approaches is that they are all based on Tumour-centered studies of the causes of malignization and are not taking into account the Tumour Stroma contribution to tumour invasion and metastasis. Thus, we postulate that the tumour stroma indeed plays a critical role in malignization after anti-angiogenic therapies and its specific mechanisms should be determined in order to target these stromal components to impede tumour malignization after antiangiogenic therapies.
STROMALIGN is a groundbreaking project designed to overcome these current limitations from a novel perspective: instead of focusing on the genetically instable and highly adaptive tumour cell component, we rather postulate that the tumour stroma plays a critical role in malignization after anti-angiogenic therapies and its targeting could offer significant advantages of less adaptation/resistance, and broader applicability to several different tumour types. Thus, we will initially dissect the mechanisms of stromal contribution to this malignization effect, followed by pharmacological targeting this event in transgenic and recently developed Tumorgraft mouse models of cancer, and later on applying this knowledge to the clinical setting in samples from two approved clinical studies with anti-angiogenic therapies currently ongoing at our Hospital.
By starting at the biology of animal models and later on validating these findings in the clinical setting we will tackle this current biomedical challenge by finding new stromal targets of malignancy that will ultimately benefit anti-angiogenic treated patients in the clinic.
Summary
Angiogenesis inhibition has proven to be a successful anti-cancer therapeutic approach and anti-angiogenic therapies are currently approved as standard therapy in several types of cancer for their clinically validated beneficial extension of overall survival, progression-free survival and/or time-to-progression in these cancer patients. Nevertheless, in preclinical mouse models of cancer, although these therapies also show significant anti-tumour effects and overall survival benefit, these therapies are also triggering increased tumour local invasion, with more distant dissemination and emergence of metastasis (overall tumour malignization). The clinical relevance of this malignization effect in patients that are currently being treated with antiangiogenic drugs still remains elusive, but the frequent tumour relapses and acquired resistance to therapy in some patients strongly suggests this insidious event should be exhaustively evaluated.
In this sense, many laboratories are rapidly advancing in the study of the molecular players and signalling pathways implicated in this event, although an important limitation of all these approaches is that they are all based on Tumour-centered studies of the causes of malignization and are not taking into account the Tumour Stroma contribution to tumour invasion and metastasis. Thus, we postulate that the tumour stroma indeed plays a critical role in malignization after anti-angiogenic therapies and its specific mechanisms should be determined in order to target these stromal components to impede tumour malignization after antiangiogenic therapies.
STROMALIGN is a groundbreaking project designed to overcome these current limitations from a novel perspective: instead of focusing on the genetically instable and highly adaptive tumour cell component, we rather postulate that the tumour stroma plays a critical role in malignization after anti-angiogenic therapies and its targeting could offer significant advantages of less adaptation/resistance, and broader applicability to several different tumour types. Thus, we will initially dissect the mechanisms of stromal contribution to this malignization effect, followed by pharmacological targeting this event in transgenic and recently developed Tumorgraft mouse models of cancer, and later on applying this knowledge to the clinical setting in samples from two approved clinical studies with anti-angiogenic therapies currently ongoing at our Hospital.
By starting at the biology of animal models and later on validating these findings in the clinical setting we will tackle this current biomedical challenge by finding new stromal targets of malignancy that will ultimately benefit anti-angiogenic treated patients in the clinic.
Max ERC Funding
1 495 796 €
Duration
Start date: 2012-06-01, End date: 2017-05-31
Project acronym STRP-DIFFERENTIATION
Project BIOMOLECULAR CHARACTERIZATION OF STREPTOMYCES DIFFERENTIATION AND ITS RELATIONSHIP WITH SECONDARY METABOLITE PRODUCTION
Researcher (PI) Ángel Manteca Fernández
Host Institution (HI) UNIVERSIDAD DE OVIEDO
Call Details Starting Grant (StG), LS9, ERC-2011-StG_20101109
Summary Streptomyces is a gram positive bacterium characterized by a complex developmental cycle. It is considered as a multicellular prokaryotic model that includes programmed cell death and sporulation. Streptomycetes are very important in industry, since they produce two thirds of clinically relevant secondary metabolites. Streptomyces and other bacteria with complex life cycles represent the evolutionary origin of some of the protein domains involved in the most important eukaryotic signalling pathways.
The classical Streptomyces developmental cycle focused in the sporulation. Industrial fermentations are mainly produced in liquid cultures (large bioreactors), conditions in which there is not sporulation, and it was traditionally considered that there was no differentiation. During his predoctoral training, A. Manteca re-evaluated Streptomyces development in solid sporulating cultures, laying the foundation of a new research line about Streptomyces differentiation totally independent to the investigations of his PhD supervisor (Streptomyces nucleases). During his postdoctoral training, he continued working in this emerging research line analyzing the relation between differentiation and secondary metabolite production, reporting the first study in which antibiotic production could be associated with hyphae differentiation in liquid. Later, he focused in the proteomic analysis of Streptomyces differentiation, creating the most complete database about proteome variations associated with hyphae differentiation.
The main objective of this project will be characterizing the biomolecular pathways behind Streptomyces differentiation, and their homologies and differences with eukaryotic signalling pathways. We will use the innovative developmental model elaborated by us and the information about the proteome differences during Streptomyces differentiation, to perform large scale mutagenesis and exhaustive phenotypic / bioinformatic characterization of these mutants.
Summary
Streptomyces is a gram positive bacterium characterized by a complex developmental cycle. It is considered as a multicellular prokaryotic model that includes programmed cell death and sporulation. Streptomycetes are very important in industry, since they produce two thirds of clinically relevant secondary metabolites. Streptomyces and other bacteria with complex life cycles represent the evolutionary origin of some of the protein domains involved in the most important eukaryotic signalling pathways.
The classical Streptomyces developmental cycle focused in the sporulation. Industrial fermentations are mainly produced in liquid cultures (large bioreactors), conditions in which there is not sporulation, and it was traditionally considered that there was no differentiation. During his predoctoral training, A. Manteca re-evaluated Streptomyces development in solid sporulating cultures, laying the foundation of a new research line about Streptomyces differentiation totally independent to the investigations of his PhD supervisor (Streptomyces nucleases). During his postdoctoral training, he continued working in this emerging research line analyzing the relation between differentiation and secondary metabolite production, reporting the first study in which antibiotic production could be associated with hyphae differentiation in liquid. Later, he focused in the proteomic analysis of Streptomyces differentiation, creating the most complete database about proteome variations associated with hyphae differentiation.
The main objective of this project will be characterizing the biomolecular pathways behind Streptomyces differentiation, and their homologies and differences with eukaryotic signalling pathways. We will use the innovative developmental model elaborated by us and the information about the proteome differences during Streptomyces differentiation, to perform large scale mutagenesis and exhaustive phenotypic / bioinformatic characterization of these mutants.
Max ERC Funding
1 341 985 €
Duration
Start date: 2012-01-01, End date: 2017-06-30
Project acronym SWCD
Project Development of super-wheat crops by introgressing agronomic traits from related wild species
Researcher (PI) Maria-Pilar Prieto
Host Institution (HI) AGENCIA ESTATAL CONSEJO SUPERIOR DEINVESTIGACIONES CIENTIFICAS
Call Details Starting Grant (StG), LS9, ERC-2009-StG
Summary Wheat is one of the most important food crops in the world and understanding its genetics and genome organisation is of great value for genetics and plant breeding purposes. Despite is genome complexity (polyploidy), hexaploid and tetraploid wheats behave as diploids during meiosis. This means that each chromosome only recognises its identical (homologue) to pair and not the related chromosomes (homeologues). There are several pairing homologous (Ph) genes controlling chromosome pairing in wheat during meiosis. The strongest effect is associated with the Ph1 locus, which is located on the long arm on chromosome 5B. Ph1 appears to sense homology prior to the chromosomes coming into contact with each other at early meiosis. Thus, if the chromosomes are true homologues, they perfectly recognise each other, chromatin remodelling is synchronized and allows pairing and recombination to occur. If the chromosomes are homoeologous (related), remodelling is not synchronized and the chromosomes fail to pair and recombine. In the absence of Ph1, all chromosomes can remodel without the requirement for the presence of an identical or near identical chromosome, and this increases the chance of pairing between related chromosomes in addition to pairing between true homologues. In this project we want to analyse deeper this Ph1 behaviour and we also want to exploit ph1 mutants as a tool for wheat breeding programs to promote inter-specific recombination between related wild species. In fact, Hordeum chilense (wild barley with interesting agronomic traits for wheat breeding) introgressions will be developed into durum wheat to transfer desirable agronomic traits from this wild barley into wheat, like resistance to diseases or the increment in carotene content. Introgression lines will be crosses with ph1 mutants to promote inter-specific recombination H. chilense-wheat, reduce the size of the H. chilense chromosome fragment. New wheat varieties will be generated.
Summary
Wheat is one of the most important food crops in the world and understanding its genetics and genome organisation is of great value for genetics and plant breeding purposes. Despite is genome complexity (polyploidy), hexaploid and tetraploid wheats behave as diploids during meiosis. This means that each chromosome only recognises its identical (homologue) to pair and not the related chromosomes (homeologues). There are several pairing homologous (Ph) genes controlling chromosome pairing in wheat during meiosis. The strongest effect is associated with the Ph1 locus, which is located on the long arm on chromosome 5B. Ph1 appears to sense homology prior to the chromosomes coming into contact with each other at early meiosis. Thus, if the chromosomes are true homologues, they perfectly recognise each other, chromatin remodelling is synchronized and allows pairing and recombination to occur. If the chromosomes are homoeologous (related), remodelling is not synchronized and the chromosomes fail to pair and recombine. In the absence of Ph1, all chromosomes can remodel without the requirement for the presence of an identical or near identical chromosome, and this increases the chance of pairing between related chromosomes in addition to pairing between true homologues. In this project we want to analyse deeper this Ph1 behaviour and we also want to exploit ph1 mutants as a tool for wheat breeding programs to promote inter-specific recombination between related wild species. In fact, Hordeum chilense (wild barley with interesting agronomic traits for wheat breeding) introgressions will be developed into durum wheat to transfer desirable agronomic traits from this wild barley into wheat, like resistance to diseases or the increment in carotene content. Introgression lines will be crosses with ph1 mutants to promote inter-specific recombination H. chilense-wheat, reduce the size of the H. chilense chromosome fragment. New wheat varieties will be generated.
Max ERC Funding
600 000 €
Duration
Start date: 2010-01-01, End date: 2015-12-31
Project acronym TEBLYM
Project Teleost B lymphocytes, the equivalent of mammalian B1 innate lymphocytes?
Researcher (PI) Carolina Tafalla Piñeiro
Host Institution (HI) INSTITUTO NACIONAL DE INVESTIGACION Y TECNOLOGIA AGRARIA Y ALIMENTARIA OA MP
Call Details Starting Grant (StG), LS9, ERC-2011-StG_20101109
Summary Teleost fish are the most primitive bony vertebrates that contain immunoglobulins, although belonging to different classes than those of mammals, having only IgM, IgD and IgT. No Ig maturation to more specific class is observed in fish, providing them with an antibody response that is slow and weak in terms of antigen affinity. This is the main reason why protection conferred by disease natural resistance or vaccination does not correlate with an antibody response. Despite these great differences, fish immunologists have always thought of fish B lymphocytes as the equivalent of mammalian conventional B2 lymphocytes, thus assuming many aspects of their functionality and leaving some possibilities unexplored.
However, many evidences strongly suggest that fish B lymphocytes do not act as mammalian B2 lymphocytes but closely resemble the B1 mammalian innate lymphocytes which are known to produce large amounts of IgM without previous exposure to the pathogen in a T-independent form.
Therefore, my main objective in this proposal is the phenotypical and functional characterization of fish B cells using the rainbow trout as a model. This will be performed with no restrictions derived from the assumption of roles ascribed in the basis of their homology to mammalian B2 lymphocytes, but in the light of the hypothesis that fish B lymphocytes resemble better a B1 model. This new context will enable me to explore possible functions and characteristics previously unexplored such as T cell independence, lack of antigen engagement, poly-reactivity of antibodies produced and role in pro-inflammatory responses.
The results obtained from this project may constitute a turning point on the field of fish immunology that should redefine previous unexplained results, having also practical repercussions for future vaccination strategies. Finally, fish B lymphocytes could become a model for humans B1 cells implicated in autoimmune diseases and leukaemias.
Summary
Teleost fish are the most primitive bony vertebrates that contain immunoglobulins, although belonging to different classes than those of mammals, having only IgM, IgD and IgT. No Ig maturation to more specific class is observed in fish, providing them with an antibody response that is slow and weak in terms of antigen affinity. This is the main reason why protection conferred by disease natural resistance or vaccination does not correlate with an antibody response. Despite these great differences, fish immunologists have always thought of fish B lymphocytes as the equivalent of mammalian conventional B2 lymphocytes, thus assuming many aspects of their functionality and leaving some possibilities unexplored.
However, many evidences strongly suggest that fish B lymphocytes do not act as mammalian B2 lymphocytes but closely resemble the B1 mammalian innate lymphocytes which are known to produce large amounts of IgM without previous exposure to the pathogen in a T-independent form.
Therefore, my main objective in this proposal is the phenotypical and functional characterization of fish B cells using the rainbow trout as a model. This will be performed with no restrictions derived from the assumption of roles ascribed in the basis of their homology to mammalian B2 lymphocytes, but in the light of the hypothesis that fish B lymphocytes resemble better a B1 model. This new context will enable me to explore possible functions and characteristics previously unexplored such as T cell independence, lack of antigen engagement, poly-reactivity of antibodies produced and role in pro-inflammatory responses.
The results obtained from this project may constitute a turning point on the field of fish immunology that should redefine previous unexplained results, having also practical repercussions for future vaccination strategies. Finally, fish B lymphocytes could become a model for humans B1 cells implicated in autoimmune diseases and leukaemias.
Max ERC Funding
1 390 000 €
Duration
Start date: 2011-12-01, End date: 2016-11-30
Project acronym TEMUBLYM
Project Teleost mucosal B1-like lymphocytes at the crossroad of tolerance and immunity
Researcher (PI) Carolina TAFALLA PINEIRO
Host Institution (HI) INSTITUTO NACIONAL DE INVESTIGACION Y TECNOLOGIA AGRARIA Y ALIMENTARIA OA MP
Call Details Consolidator Grant (CoG), LS9, ERC-2016-COG
Summary B cells are one of the main players of immunity, responsible for the production of immunoglobulins (Igs). In 2011, I was granted an ERC Starting grant to undertake the phenotypical and functional characterization of teleost B lymphocytes based on the hypothesis that they do not behave as mammalian B2 cells (conventional B cells) but closely resemble mammalian innate B1 lymphocytes involved in extrafollicular T-independent (TI) responses. Since then, my laboratory has gathered considerable evidences that strengthen this hypothesis. These studies were mostly carried out in central lymphoid compartments, but did not address how teleost B1-like cells regulate the delicate balance between immunity and tolerance at mucosal interfaces, in species lacking follicular structures. In this new project, I want to pursue my studies on B lymphocyte functionality, focusing on how teleost mucosal B cells are regulated, still under the assumption that fish B lymphocytes resemble better a B1 model. We will study how fish B cells differentiate to antibody secreting cells (ASCs) and establish extrafollicular long-term memory, taking into account novel results in mammals that have challenged traditional paradigms and revealed that long-term immunological memory can be established through TI IgM B1-like responses. Furthermore, we will also study the role of IgD in the gills, as previous studies from my group suggest that this Ig plays a key role in the regulation of immunity in this specific mucosa, as it seems to do in humans in areas such as the upper respiratory tract.
Addressing how fish B cells mount a protective mucosal immune response in the absence of T cell help from organized follicles could provide new mechanistic insights into IgM and IgD responses emerging in humans. From a practical view, our work will contribute to understand why satisfactory mucosal vaccination is still an unreached goal for most diseases in both mammals and fish, despite their strong demand.
Summary
B cells are one of the main players of immunity, responsible for the production of immunoglobulins (Igs). In 2011, I was granted an ERC Starting grant to undertake the phenotypical and functional characterization of teleost B lymphocytes based on the hypothesis that they do not behave as mammalian B2 cells (conventional B cells) but closely resemble mammalian innate B1 lymphocytes involved in extrafollicular T-independent (TI) responses. Since then, my laboratory has gathered considerable evidences that strengthen this hypothesis. These studies were mostly carried out in central lymphoid compartments, but did not address how teleost B1-like cells regulate the delicate balance between immunity and tolerance at mucosal interfaces, in species lacking follicular structures. In this new project, I want to pursue my studies on B lymphocyte functionality, focusing on how teleost mucosal B cells are regulated, still under the assumption that fish B lymphocytes resemble better a B1 model. We will study how fish B cells differentiate to antibody secreting cells (ASCs) and establish extrafollicular long-term memory, taking into account novel results in mammals that have challenged traditional paradigms and revealed that long-term immunological memory can be established through TI IgM B1-like responses. Furthermore, we will also study the role of IgD in the gills, as previous studies from my group suggest that this Ig plays a key role in the regulation of immunity in this specific mucosa, as it seems to do in humans in areas such as the upper respiratory tract.
Addressing how fish B cells mount a protective mucosal immune response in the absence of T cell help from organized follicles could provide new mechanistic insights into IgM and IgD responses emerging in humans. From a practical view, our work will contribute to understand why satisfactory mucosal vaccination is still an unreached goal for most diseases in both mammals and fish, despite their strong demand.
Max ERC Funding
1 866 046 €
Duration
Start date: 2017-04-01, End date: 2022-03-31
Project acronym THERACAN
Project Novel therapeutic strategies to treat pancreatic and lung cancer
Researcher (PI) Mariano BARBACID MONTALBAN
Host Institution (HI) FUNDACION CENTRO NACIONAL DE INVESTIGACIONES ONCOLOGICAS CARLOS III
Call Details Advanced Grant (AdG), LS4, ERC-2015-AdG
Summary This proposal is aimed at identifying and functionally validating targets with potential therapeutic value to devise novel strategies to treat two human cancers with unacceptable low survival rates and unmet medical needs: pancreatic ductal adenocarcinoma and K-RAS mutant lung adenocarcinoma. Although these tumor types have distinct pathological and clinical manifestations, they are both driven by K-RAS mutations. Hence, we expect that the proposed studies will generate synergies to accelerate the outcome of the expected results. In the first part of the proposal, we will identify those genes activated in the cancer initiating cells responsible for the onset of pancreatic and lung tumors. We reasoned that genes implicated in the initial stages of tumor development will be maintained during tumor evolution and not be affected by the intra-tumoral heterogeneity generated during tumor progression. We also propose to identify and validate genes capable of reprogramming the desmoplasic stroma characteristic of pancreatic tumors to hamper its pro-tumoral effects. Likewise, we intend to define the molecular events that control senescence, a naturally occurring process that serves as a barrier to tumor development. In the second part of the project, we will interrogate the role of known targets with suspected therapeutic value in tumor progression using a new generation of GEM tumor models that allow the temporal separation of tumor development from target ablation or inactivation. These studies will make it possible to design combination therapies capable of effectively eradicate advanced tumors. The last section of this proposal focuses on the pharmacological validation of these combination therapies using best-in-class inhibitors in state-of-the-art preclinical trial platforms based on GEM and PDX tumor models. The results derived from these studies will guide the design of new clinical trials that should have a positive impact in the treatment of these deadly diseases.
Summary
This proposal is aimed at identifying and functionally validating targets with potential therapeutic value to devise novel strategies to treat two human cancers with unacceptable low survival rates and unmet medical needs: pancreatic ductal adenocarcinoma and K-RAS mutant lung adenocarcinoma. Although these tumor types have distinct pathological and clinical manifestations, they are both driven by K-RAS mutations. Hence, we expect that the proposed studies will generate synergies to accelerate the outcome of the expected results. In the first part of the proposal, we will identify those genes activated in the cancer initiating cells responsible for the onset of pancreatic and lung tumors. We reasoned that genes implicated in the initial stages of tumor development will be maintained during tumor evolution and not be affected by the intra-tumoral heterogeneity generated during tumor progression. We also propose to identify and validate genes capable of reprogramming the desmoplasic stroma characteristic of pancreatic tumors to hamper its pro-tumoral effects. Likewise, we intend to define the molecular events that control senescence, a naturally occurring process that serves as a barrier to tumor development. In the second part of the project, we will interrogate the role of known targets with suspected therapeutic value in tumor progression using a new generation of GEM tumor models that allow the temporal separation of tumor development from target ablation or inactivation. These studies will make it possible to design combination therapies capable of effectively eradicate advanced tumors. The last section of this proposal focuses on the pharmacological validation of these combination therapies using best-in-class inhibitors in state-of-the-art preclinical trial platforms based on GEM and PDX tumor models. The results derived from these studies will guide the design of new clinical trials that should have a positive impact in the treatment of these deadly diseases.
Max ERC Funding
2 499 500 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym TRANSFR-Q
Project Transcriptional RegulAtory Network controlling Strawberry Fruit Ripening and Quality
Researcher (PI) David Posé Padilla
Host Institution (HI) UNIVERSIDAD DE MALAGA
Call Details Starting Grant (StG), LS9, ERC-2014-STG
Summary Ripening is the critical step for the development of flavour quality in fruit. This character has significantly declined in many fleshy fruits over recent decades, primarily due to the focus of current breeding programs on agronomic traits such as production, firmness, and postharvest shelf life. This strategy has caused a tunnelling effect on genetic variability in many crops. This is particularly significant in strawberry, where current cultivars are derived from a narrow germplasm stock. A notable feature of strawberry fruit is that undergoes a non-climacteric ripening program independent of the hormone ethylene, in contrast to well studied climacteric fruits such as tomato, where ethylene plays a central role. Therefore, improving fruit flavour in present strawberry varieties requires two important breakthroughs: 1) a precise understanding of non-climacteric fruit ripening regulation that will allow the targeting of relevant quality genes, and 2) the identification of unexploited allelic variants from wild germplasm to be introgressed through the generation of novel breeding lines. The first objective will be achieved by (i) focusing on the sequenced, diploid Fragaria vesca, a wild relative of the cultivated strawberry that will serve as a model, (ii) identifying key transcription factors (TFs) regulating fruit ripening by generating a stage- and tissue-specific gene expression map, (iii) using a candidate gene approach and reverse genetics based on gene silencing and TILLING to verify the role of these TFs, and (iv) defining the gene regulatory networks controlling the ripening process via integration of transcriptomic, metabolomic and ChIP-Seq data obtained from the stably silenced and/or TILLED lines. Finally, TRANSFR-Q plans to use this knowledge, combined with the identification of novel alleles from a core collection of Fragaria germplasm, to transfer flavour quality characters into current strawberry cultivars.
Summary
Ripening is the critical step for the development of flavour quality in fruit. This character has significantly declined in many fleshy fruits over recent decades, primarily due to the focus of current breeding programs on agronomic traits such as production, firmness, and postharvest shelf life. This strategy has caused a tunnelling effect on genetic variability in many crops. This is particularly significant in strawberry, where current cultivars are derived from a narrow germplasm stock. A notable feature of strawberry fruit is that undergoes a non-climacteric ripening program independent of the hormone ethylene, in contrast to well studied climacteric fruits such as tomato, where ethylene plays a central role. Therefore, improving fruit flavour in present strawberry varieties requires two important breakthroughs: 1) a precise understanding of non-climacteric fruit ripening regulation that will allow the targeting of relevant quality genes, and 2) the identification of unexploited allelic variants from wild germplasm to be introgressed through the generation of novel breeding lines. The first objective will be achieved by (i) focusing on the sequenced, diploid Fragaria vesca, a wild relative of the cultivated strawberry that will serve as a model, (ii) identifying key transcription factors (TFs) regulating fruit ripening by generating a stage- and tissue-specific gene expression map, (iii) using a candidate gene approach and reverse genetics based on gene silencing and TILLING to verify the role of these TFs, and (iv) defining the gene regulatory networks controlling the ripening process via integration of transcriptomic, metabolomic and ChIP-Seq data obtained from the stably silenced and/or TILLED lines. Finally, TRANSFR-Q plans to use this knowledge, combined with the identification of novel alleles from a core collection of Fragaria germplasm, to transfer flavour quality characters into current strawberry cultivars.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-06-01, End date: 2021-05-31
Project acronym YOUNGatHEART
Project YOUNGatHEART: CARDIAC REJUVENATION BY EPIGENETIC REMODELLING
Researcher (PI) SUSANA Gonzalez
Host Institution (HI) CENTRO NACIONAL DE INVESTIGACIONESCARDIOVASCULARES CARLOS III (F.S.P.)
Call Details Consolidator Grant (CoG), LS4, ERC-2014-CoG
Summary Aging poses the largest risk for cardiovascular disease (CVD) and is orchestrated, to some extent, by epigenetic changes. Despite the significant progress on many fronts in the cardiovascular field, non-inherited epigenetic regulation in cardiac aging and CVD remains unexplored. Dilated Cardiomyopathy (DCM) is a major contributor to healthcare costs and it is the leading indication for heart transplantation. We have recently discovered that adult cardiac-specific deletion of epigenetic regulator Bmi1 in mice induces DCM and heart failure. These unprecedented data support the idea that inadequate epigenetic regulation in adulthood is critical in CVD. In addition, our studies with parabiotic pairing of healthy and DCM-diagnosed mice show that the circulation of a healthy mouse significantly improve the cardiac performance of mouse with DCM. These ground-breaking discoveries suggest that DCM regression, or cardiac rejuvenation, is feasible in terms of epigenetic states. Therefore, YOUNGatHEART will unveil significant breakthrough on (1) how non-inherited epigenetic deregulation induces DCM and (2) how epigenetic remodeling reversed this process. For that, our challenges are: 1A. To decipher how aged-linked cardiac dysfunction contributes to CVD by identifying the epigenetic landscape regulating cardiac aging among species; 1B. To decode how epigenetic deregulation induces DCM by integrating clinical data and samples from DCM-transplanted patients with imaging, transcriptomic, proteomic, and functional approaches from DCM model; and, 2A. To identified systemic factors with anti-cardiomyopathic effects by systematic proteomic screenings after parabiosis and epigenome of the DCM hearts. In sum, YOUNGatHEART puts forward an ambitious but feasible and pioneering program to tackle the epigenetic hallmark in cardiac aging with the final aim (2B) of setting the molecular basis for future therapeutic interventions in CVD.
Summary
Aging poses the largest risk for cardiovascular disease (CVD) and is orchestrated, to some extent, by epigenetic changes. Despite the significant progress on many fronts in the cardiovascular field, non-inherited epigenetic regulation in cardiac aging and CVD remains unexplored. Dilated Cardiomyopathy (DCM) is a major contributor to healthcare costs and it is the leading indication for heart transplantation. We have recently discovered that adult cardiac-specific deletion of epigenetic regulator Bmi1 in mice induces DCM and heart failure. These unprecedented data support the idea that inadequate epigenetic regulation in adulthood is critical in CVD. In addition, our studies with parabiotic pairing of healthy and DCM-diagnosed mice show that the circulation of a healthy mouse significantly improve the cardiac performance of mouse with DCM. These ground-breaking discoveries suggest that DCM regression, or cardiac rejuvenation, is feasible in terms of epigenetic states. Therefore, YOUNGatHEART will unveil significant breakthrough on (1) how non-inherited epigenetic deregulation induces DCM and (2) how epigenetic remodeling reversed this process. For that, our challenges are: 1A. To decipher how aged-linked cardiac dysfunction contributes to CVD by identifying the epigenetic landscape regulating cardiac aging among species; 1B. To decode how epigenetic deregulation induces DCM by integrating clinical data and samples from DCM-transplanted patients with imaging, transcriptomic, proteomic, and functional approaches from DCM model; and, 2A. To identified systemic factors with anti-cardiomyopathic effects by systematic proteomic screenings after parabiosis and epigenome of the DCM hearts. In sum, YOUNGatHEART puts forward an ambitious but feasible and pioneering program to tackle the epigenetic hallmark in cardiac aging with the final aim (2B) of setting the molecular basis for future therapeutic interventions in CVD.
Max ERC Funding
1 861 910 €
Duration
Start date: 2015-11-01, End date: 2020-10-31