Project acronym 3CBIOTECH
Project Cold Carbon Catabolism of Microbial Communities underprinning a Sustainable Bioenergy and Biorefinery Economy
Researcher (PI) Gavin James Collins
Host Institution (HI) NATIONAL UNIVERSITY OF IRELAND GALWAY
Call Details Starting Grant (StG), LS9, ERC-2010-StG_20091118
Summary The applicant will collaborate with Irish, European and U.S.-based colleagues to develop a sustainable biorefinery and bioenergy industry in Ireland and Europe. The focus of this ERC Starting Grant will be the application of classical microbiological, physiological and real-time polymerase chain reaction (PCR)-based assays, to qualitatively and quantitatively characterize microbial communities underpinning novel and innovative, low-temperature, anaerobic waste (and other biomass) conversion technologies, including municipal wastewater treatment and, demonstration- and full-scale biorefinery applications.
Anaerobic digestion (AD) is a naturally-occurring process, which is widely applied for the conversion of waste to methane-containing biogas. Low-temperature (<20 degrees C) AD has been applied by the applicant as a cost-effective alternative to mesophilic (c. 35C) AD for the treatment of several waste categories. However, the microbiology of low-temperature AD is poorly understood. The applicant will work with microbial consortia isolated from anaerobic bioreactors, which have been operated for long-term experiments (>3.5 years), and include organic acid-oxidizing, hydrogen-producing syntrophic microbes and hydrogen-consuming methanogens. A major focus of the project will be the ecophysiology of psychrotolerant and psychrophilic methanogens already identified and cultivated by the applicant. The project will also investigate the role(s) of poorly-understood Crenarchaeota populations and homoacetogenic bacteria, in complex consortia. The host organization is a leading player in the microbiology of waste-to-energy applications. The applicant will train a team of scientists in all aspects of the microbiology and bioengineering of biomass conversion systems.
Summary
The applicant will collaborate with Irish, European and U.S.-based colleagues to develop a sustainable biorefinery and bioenergy industry in Ireland and Europe. The focus of this ERC Starting Grant will be the application of classical microbiological, physiological and real-time polymerase chain reaction (PCR)-based assays, to qualitatively and quantitatively characterize microbial communities underpinning novel and innovative, low-temperature, anaerobic waste (and other biomass) conversion technologies, including municipal wastewater treatment and, demonstration- and full-scale biorefinery applications.
Anaerobic digestion (AD) is a naturally-occurring process, which is widely applied for the conversion of waste to methane-containing biogas. Low-temperature (<20 degrees C) AD has been applied by the applicant as a cost-effective alternative to mesophilic (c. 35C) AD for the treatment of several waste categories. However, the microbiology of low-temperature AD is poorly understood. The applicant will work with microbial consortia isolated from anaerobic bioreactors, which have been operated for long-term experiments (>3.5 years), and include organic acid-oxidizing, hydrogen-producing syntrophic microbes and hydrogen-consuming methanogens. A major focus of the project will be the ecophysiology of psychrotolerant and psychrophilic methanogens already identified and cultivated by the applicant. The project will also investigate the role(s) of poorly-understood Crenarchaeota populations and homoacetogenic bacteria, in complex consortia. The host organization is a leading player in the microbiology of waste-to-energy applications. The applicant will train a team of scientists in all aspects of the microbiology and bioengineering of biomass conversion systems.
Max ERC Funding
1 499 797 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym A-DIET
Project Metabolomics based biomarkers of dietary intake- new tools for nutrition research
Researcher (PI) Lorraine Brennan
Host Institution (HI) UNIVERSITY COLLEGE DUBLIN, NATIONAL UNIVERSITY OF IRELAND, DUBLIN
Call Details Consolidator Grant (CoG), LS7, ERC-2014-CoG
Summary In todays advanced technological world, we can track the exact movement of individuals, analyse their genetic makeup and predict predisposition to certain diseases. However, we are unable to accurately assess an individual’s dietary intake. This is without a doubt one of the main stumbling blocks in assessing the link between diet and disease/health. The present proposal (A-DIET) will address this issue with the overarching objective to develop novel strategies for assessment of dietary intake.
Using approaches to (1) identify biomarkers of specific foods (2) classify people into dietary patterns (nutritypes) and (3) develop a tool for integration of dietary and biomarker data, A-DIET has the potential to dramatically enhance our ability to accurately assess dietary intake. The ultimate output from A-DIET will be a dietary assessment tool which can be used to obtain an accurate assessment of dietary intake by combining dietary and biomarker data which in turn will allow investigations into relationships between diet, health and disease. New biomarkers of specific foods will be identified and validated using intervention studies and metabolomic analyses. Methods will be developed to classify individuals into dietary patterns based on biomarker/metabolomic profiles thus demonstrating the novel concept of nutritypes. Strategies for integration of dietary and biomarker data will be developed and translated into a tool that will be made available to the wider scientific community.
Advances made in A-DIET will enable nutrition epidemiologist’s to properly examine the relationship between diet and disease and develop clear public health messages with regard to diet and health. Additionally results from A-DIET will allow researchers to accurately assess people’s diet and implement health promotion strategies and enable dieticians in a clinical environment to assess compliance to therapeutic diets such as adherence to a high fibre diet or a gluten free diet.
Summary
In todays advanced technological world, we can track the exact movement of individuals, analyse their genetic makeup and predict predisposition to certain diseases. However, we are unable to accurately assess an individual’s dietary intake. This is without a doubt one of the main stumbling blocks in assessing the link between diet and disease/health. The present proposal (A-DIET) will address this issue with the overarching objective to develop novel strategies for assessment of dietary intake.
Using approaches to (1) identify biomarkers of specific foods (2) classify people into dietary patterns (nutritypes) and (3) develop a tool for integration of dietary and biomarker data, A-DIET has the potential to dramatically enhance our ability to accurately assess dietary intake. The ultimate output from A-DIET will be a dietary assessment tool which can be used to obtain an accurate assessment of dietary intake by combining dietary and biomarker data which in turn will allow investigations into relationships between diet, health and disease. New biomarkers of specific foods will be identified and validated using intervention studies and metabolomic analyses. Methods will be developed to classify individuals into dietary patterns based on biomarker/metabolomic profiles thus demonstrating the novel concept of nutritypes. Strategies for integration of dietary and biomarker data will be developed and translated into a tool that will be made available to the wider scientific community.
Advances made in A-DIET will enable nutrition epidemiologist’s to properly examine the relationship between diet and disease and develop clear public health messages with regard to diet and health. Additionally results from A-DIET will allow researchers to accurately assess people’s diet and implement health promotion strategies and enable dieticians in a clinical environment to assess compliance to therapeutic diets such as adherence to a high fibre diet or a gluten free diet.
Max ERC Funding
1 995 548 €
Duration
Start date: 2015-08-01, End date: 2020-07-31
Project acronym ABC
Project Targeting Multidrug Resistant Cancer
Researcher (PI) Gergely Szakacs
Host Institution (HI) MAGYAR TUDOMANYOS AKADEMIA TERMESZETTUDOMANYI KUTATOKOZPONT
Call Details Starting Grant (StG), LS7, ERC-2010-StG_20091118
Summary Despite considerable advances in drug discovery, resistance to anticancer chemotherapy confounds the effective treatment of patients. Cancer cells can acquire broad cross-resistance to mechanistically and structurally unrelated drugs. P-glycoprotein (Pgp) actively extrudes many types of drugs from cancer cells, thereby conferring resistance to those agents. The central tenet of my work is that Pgp, a universally accepted biomarker of drug resistance, should in addition be considered as a molecular target of multidrug-resistant (MDR) cancer cells. Successful targeting of MDR cells would reduce the tumor burden and would also enable the elimination of ABC transporter-overexpressing cancer stem cells that are responsible for the replenishment of tumors. The proposed project is based on the following observations:
- First, by using a pharmacogenomic approach, I have revealed the hidden vulnerability of MDRcells (Szakács et al. 2004, Cancer Cell 6, 129-37);
- Second, I have identified a series of MDR-selective compounds with increased toxicity toPgp-expressing cells
(Turk et al.,Cancer Res, 2009. 69(21));
- Third, I have shown that MDR-selective compounds can be used to prevent theemergence of MDR (Ludwig, Szakács et al. 2006, Cancer Res 66, 4808-15);
- Fourth, we have generated initial pharmacophore models for cytotoxicity and MDR-selectivity (Hall et al. 2009, J Med Chem 52, 3191-3204).
I propose a comprehensive series of studies that will address thefollowing critical questions:
- First, what is the scope of MDR-selective compounds?
- Second, what is their mechanism of action?
- Third, what is the optimal therapeutic modality?
Extensive biological, pharmacological and bioinformatic analyses will be utilized to address four major specific aims. These aims address basic questions concerning the physiology of MDR ABC transporters in determining the mechanism of action of MDR-selective compounds, setting the stage for a fresh therapeutic approach that may eventually translate into improved patient care.
Summary
Despite considerable advances in drug discovery, resistance to anticancer chemotherapy confounds the effective treatment of patients. Cancer cells can acquire broad cross-resistance to mechanistically and structurally unrelated drugs. P-glycoprotein (Pgp) actively extrudes many types of drugs from cancer cells, thereby conferring resistance to those agents. The central tenet of my work is that Pgp, a universally accepted biomarker of drug resistance, should in addition be considered as a molecular target of multidrug-resistant (MDR) cancer cells. Successful targeting of MDR cells would reduce the tumor burden and would also enable the elimination of ABC transporter-overexpressing cancer stem cells that are responsible for the replenishment of tumors. The proposed project is based on the following observations:
- First, by using a pharmacogenomic approach, I have revealed the hidden vulnerability of MDRcells (Szakács et al. 2004, Cancer Cell 6, 129-37);
- Second, I have identified a series of MDR-selective compounds with increased toxicity toPgp-expressing cells
(Turk et al.,Cancer Res, 2009. 69(21));
- Third, I have shown that MDR-selective compounds can be used to prevent theemergence of MDR (Ludwig, Szakács et al. 2006, Cancer Res 66, 4808-15);
- Fourth, we have generated initial pharmacophore models for cytotoxicity and MDR-selectivity (Hall et al. 2009, J Med Chem 52, 3191-3204).
I propose a comprehensive series of studies that will address thefollowing critical questions:
- First, what is the scope of MDR-selective compounds?
- Second, what is their mechanism of action?
- Third, what is the optimal therapeutic modality?
Extensive biological, pharmacological and bioinformatic analyses will be utilized to address four major specific aims. These aims address basic questions concerning the physiology of MDR ABC transporters in determining the mechanism of action of MDR-selective compounds, setting the stage for a fresh therapeutic approach that may eventually translate into improved patient care.
Max ERC Funding
1 499 640 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym AGELESS
Project Comparative genomics / ‘wildlife’ transcriptomics uncovers the mechanisms of halted ageing in mammals
Researcher (PI) Emma Teeling
Host Institution (HI) UNIVERSITY COLLEGE DUBLIN, NATIONAL UNIVERSITY OF IRELAND, DUBLIN
Call Details Starting Grant (StG), LS2, ERC-2012-StG_20111109
Summary "Ageing is the gradual and irreversible breakdown of living systems associated with the advancement of time, which leads to an increase in vulnerability and eventual mortality. Despite recent advances in ageing research, the intrinsic complexity of the ageing process has prevented a full understanding of this process, therefore, ageing remains a grand challenge in contemporary biology. In AGELESS, we will tackle this challenge by uncovering the molecular mechanisms of halted ageing in a unique model system, the bats. Bats are the longest-lived mammals relative to their body size, and defy the ‘rate-of-living’ theories as they use twice as much the energy as other species of considerable size, but live far longer. This suggests that bats have some underlying mechanisms that may explain their exceptional longevity. In AGELESS, we will identify the molecular mechanisms that enable mammals to achieve extraordinary longevity, using state-of-the-art comparative genomic methodologies focused on bats. We will identify, using population transcriptomics and telomere/mtDNA genomics, the molecular changes that occur in an ageing wild population of bats to uncover how bats ‘age’ so slowly compared with other mammals. In silico whole genome analyses, field based ageing transcriptomic data, mtDNA and telomeric studies will be integrated and analysed using a networks approach, to ascertain how these systems interact to halt ageing. For the first time, we will be able to utilize the diversity seen within nature to identify key molecular targets and regions that regulate and control ageing in mammals. AGELESS will provide a deeper understanding of the causal mechanisms of ageing, potentially uncovering the crucial molecular pathways that can be modified to halt, alleviate and perhaps even reverse this process in man."
Summary
"Ageing is the gradual and irreversible breakdown of living systems associated with the advancement of time, which leads to an increase in vulnerability and eventual mortality. Despite recent advances in ageing research, the intrinsic complexity of the ageing process has prevented a full understanding of this process, therefore, ageing remains a grand challenge in contemporary biology. In AGELESS, we will tackle this challenge by uncovering the molecular mechanisms of halted ageing in a unique model system, the bats. Bats are the longest-lived mammals relative to their body size, and defy the ‘rate-of-living’ theories as they use twice as much the energy as other species of considerable size, but live far longer. This suggests that bats have some underlying mechanisms that may explain their exceptional longevity. In AGELESS, we will identify the molecular mechanisms that enable mammals to achieve extraordinary longevity, using state-of-the-art comparative genomic methodologies focused on bats. We will identify, using population transcriptomics and telomere/mtDNA genomics, the molecular changes that occur in an ageing wild population of bats to uncover how bats ‘age’ so slowly compared with other mammals. In silico whole genome analyses, field based ageing transcriptomic data, mtDNA and telomeric studies will be integrated and analysed using a networks approach, to ascertain how these systems interact to halt ageing. For the first time, we will be able to utilize the diversity seen within nature to identify key molecular targets and regions that regulate and control ageing in mammals. AGELESS will provide a deeper understanding of the causal mechanisms of ageing, potentially uncovering the crucial molecular pathways that can be modified to halt, alleviate and perhaps even reverse this process in man."
Max ERC Funding
1 499 768 €
Duration
Start date: 2013-01-01, End date: 2017-12-31
Project acronym ALH
Project Alternative life histories: linking genes to phenotypes to demography
Researcher (PI) Thomas Eric Reed
Host Institution (HI) UNIVERSITY COLLEGE CORK - NATIONAL UNIVERSITY OF IRELAND, CORK
Call Details Starting Grant (StG), LS8, ERC-2014-STG
Summary Understanding how and why individuals develop strikingly different life histories is a major goal in evolutionary biology. It is also a prerequisite for conserving important biodiversity within species and predicting the impacts of environmental change on populations. The aim of my study is to examine a key threshold phenotypic trait (alternative migratory tactics) in a series of large scale laboratory and field experiments, integrating several previously independent perspectives from evolutionary ecology, ecophysiology and genomics, to produce a downstream predictive model. My chosen study species, the brown trout Salmo trutta, has an extensive history of genetic and experimental work and exhibits ‘partial migration’: individuals either migrate to sea (‘sea trout’) or remain in freshwater their whole lives. Recent advances in molecular parentage assignment, quantitative genetics and genomics (next generation sequencing and bioinformatics) will allow unprecedented insight into how alternative life history phenotypes are moulded by the interaction between genes and environment. To provide additional mechanistic understanding of these processes, the balance between metabolic requirements during growth and available extrinsic resources will be investigated as the major physiological driver of migratory behaviour. Together these results will be used to develop a predictive model to explore the consequences of rapid environmental change, accounting for the effects of genetics and environment on phenotype and on population demographics. In addition to their value for conservation and management of an iconic and key species in European freshwaters and coastal seas, these results will generate novel insight into the evolution of migratory behaviour generally, providing a text book example of how alternative life histories are shaped and maintained in wild populations.
Summary
Understanding how and why individuals develop strikingly different life histories is a major goal in evolutionary biology. It is also a prerequisite for conserving important biodiversity within species and predicting the impacts of environmental change on populations. The aim of my study is to examine a key threshold phenotypic trait (alternative migratory tactics) in a series of large scale laboratory and field experiments, integrating several previously independent perspectives from evolutionary ecology, ecophysiology and genomics, to produce a downstream predictive model. My chosen study species, the brown trout Salmo trutta, has an extensive history of genetic and experimental work and exhibits ‘partial migration’: individuals either migrate to sea (‘sea trout’) or remain in freshwater their whole lives. Recent advances in molecular parentage assignment, quantitative genetics and genomics (next generation sequencing and bioinformatics) will allow unprecedented insight into how alternative life history phenotypes are moulded by the interaction between genes and environment. To provide additional mechanistic understanding of these processes, the balance between metabolic requirements during growth and available extrinsic resources will be investigated as the major physiological driver of migratory behaviour. Together these results will be used to develop a predictive model to explore the consequences of rapid environmental change, accounting for the effects of genetics and environment on phenotype and on population demographics. In addition to their value for conservation and management of an iconic and key species in European freshwaters and coastal seas, these results will generate novel insight into the evolution of migratory behaviour generally, providing a text book example of how alternative life histories are shaped and maintained in wild populations.
Max ERC Funding
1 499 202 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym ANICOLEVO
Project Animal coloration through deep time: evolutionary novelty, homology and taphonomy
Researcher (PI) Maria McNamara
Host Institution (HI) UNIVERSITY COLLEGE CORK - NATIONAL UNIVERSITY OF IRELAND, CORK
Call Details Starting Grant (StG), LS8, ERC-2014-STG
Summary What does the fossil record tell us about the evolution of colour in animals through deep time? Evidence of colour in fossils can inform on the visual signalling strategies used by ancient animals. Research to date often has a narrow focus, lacks a broad phylogenetic and temporal context, and rarely incorporates information on taphonomy. This proposal represents a bold new holistic approach to the study of fossil colour: it will couple powerful imaging- and chemical analytical techniques with a rigorous programme of fossilisation experiments simulating decay, burial, and transport, and analysis of fossils and their sedimentary context, to construct the first robust models for the evolution of colour in animals through deep time. The research will resolve the original integumentary colours of fossil higher vertebrates, and the original colours of fossil hair; the fossil record of non-melanin pigments in feathers and insects; the biological significance of monotonal patterning in fossil insects; and the evolutionary history of scales and 3D photonic crystals in insects. Critically, the research will test, for the first time, whether evidence of fossil colour can solve broader evolutionary questions, e.g. the true affinities of enigmatic Cambrian chordate-like metazoans, and feather-like integumentary filaments in dinosaurs. The proposal entails construction of a dedicated experimental maturation laboratory for simulating the impact of burial on tissues. This laboratory will form the core of the world’s first integrated ‘experimental fossilisation facility’, consolidating the PI’s team as the global hub for fossil colour research. The research team comprises the PI, three postdoctoral researchers, and three PhD students, and will form an extensive research network via collaborations with 13 researchers from Europe and beyond. The project will reach out to diverse scientists and will inspire a positive attitude to science among the general public and policymakers alike.
Summary
What does the fossil record tell us about the evolution of colour in animals through deep time? Evidence of colour in fossils can inform on the visual signalling strategies used by ancient animals. Research to date often has a narrow focus, lacks a broad phylogenetic and temporal context, and rarely incorporates information on taphonomy. This proposal represents a bold new holistic approach to the study of fossil colour: it will couple powerful imaging- and chemical analytical techniques with a rigorous programme of fossilisation experiments simulating decay, burial, and transport, and analysis of fossils and their sedimentary context, to construct the first robust models for the evolution of colour in animals through deep time. The research will resolve the original integumentary colours of fossil higher vertebrates, and the original colours of fossil hair; the fossil record of non-melanin pigments in feathers and insects; the biological significance of monotonal patterning in fossil insects; and the evolutionary history of scales and 3D photonic crystals in insects. Critically, the research will test, for the first time, whether evidence of fossil colour can solve broader evolutionary questions, e.g. the true affinities of enigmatic Cambrian chordate-like metazoans, and feather-like integumentary filaments in dinosaurs. The proposal entails construction of a dedicated experimental maturation laboratory for simulating the impact of burial on tissues. This laboratory will form the core of the world’s first integrated ‘experimental fossilisation facility’, consolidating the PI’s team as the global hub for fossil colour research. The research team comprises the PI, three postdoctoral researchers, and three PhD students, and will form an extensive research network via collaborations with 13 researchers from Europe and beyond. The project will reach out to diverse scientists and will inspire a positive attitude to science among the general public and policymakers alike.
Max ERC Funding
1 562 000 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym ARCHAIC ADAPT
Project Admixture accelerated adaptation: signals from modern, ancient and archaic DNA.
Researcher (PI) Emilia HUERTA-SANCHEZ
Host Institution (HI) THE PROVOST, FELLOWS, FOUNDATION SCHOLARS & THE OTHER MEMBERS OF BOARD OF THE COLLEGE OF THE HOLY & UNDIVIDED TRINITY OF QUEEN ELIZABETH NEAR DUBLIN
Call Details Starting Grant (StG), LS8, ERC-2018-STG
Summary With the advent of new sequencing technologies, population geneticists now have access to more data than ever before. We have access to thousands of human genomes from a diverse set of populations around the globe, and, thanks to advances in DNA extraction and library preparation, we now are beginning to have access to ancient DNA sequence data. These data have greatly improved our knowledge of human history, human adaptation to different environments and human disease. Genome-wide studies have highlighted many genes or genomic loci that may play a role in adaptive or disease related phenotypes of biological importance.
With these collections of modern and ancient sequence data we want to answer a key evolutionary question: how do human adaptations arise? We strongly believe that the state-of-the-art methodologies for uncovering signatures of adaptation are blind to potential modes of adaptation because they are lacking two critical components – more complete integration of multiple population haplotype data (including archaic, ancient and modern samples), and an account of population interactions that facilitate adaptation.
Therefore I plan to develop new methods to detect shared selective events across populations by creating novel statistical summaries, and to detect admixture-facilitated adaptation which we believe is likely a common mode of natural selection. We will apply these tools to new datasets to characterize the interplay of natural selection, archaic and modern admixture in populations in the Americas and make a comparative analysis of modern and ancient European samples to understand the origin and changing profile of adaptive archaic alleles. As a result our work will reveal evolutionary processes that have played an important role in human evolution and disease.
Summary
With the advent of new sequencing technologies, population geneticists now have access to more data than ever before. We have access to thousands of human genomes from a diverse set of populations around the globe, and, thanks to advances in DNA extraction and library preparation, we now are beginning to have access to ancient DNA sequence data. These data have greatly improved our knowledge of human history, human adaptation to different environments and human disease. Genome-wide studies have highlighted many genes or genomic loci that may play a role in adaptive or disease related phenotypes of biological importance.
With these collections of modern and ancient sequence data we want to answer a key evolutionary question: how do human adaptations arise? We strongly believe that the state-of-the-art methodologies for uncovering signatures of adaptation are blind to potential modes of adaptation because they are lacking two critical components – more complete integration of multiple population haplotype data (including archaic, ancient and modern samples), and an account of population interactions that facilitate adaptation.
Therefore I plan to develop new methods to detect shared selective events across populations by creating novel statistical summaries, and to detect admixture-facilitated adaptation which we believe is likely a common mode of natural selection. We will apply these tools to new datasets to characterize the interplay of natural selection, archaic and modern admixture in populations in the Americas and make a comparative analysis of modern and ancient European samples to understand the origin and changing profile of adaptive archaic alleles. As a result our work will reveal evolutionary processes that have played an important role in human evolution and disease.
Max ERC Funding
1 500 000 €
Duration
Start date: 2020-01-01, End date: 2024-12-31
Project acronym BIOELECPRO
Project Frontier Research on the Dielectric Properties of Biological Tissue
Researcher (PI) Martin James O'Halloran
Host Institution (HI) NATIONAL UNIVERSITY OF IRELAND GALWAY
Call Details Starting Grant (StG), LS7, ERC-2014-STG
Summary The dielectric properties of biological tissues are of fundamental importance to the understanding of the interaction of electromagnetic fields with the human body. These properties are used to determine the safety of electronic devices, and in the design, development and refinement of electromagnetic medical imaging and therapeutic devices. Many historical studies have aimed to establish the dielectric properties of a broad range of tissues. A growing number of recent studies have sought to more accurately estimate these dielectric properties by standardising measurement procedures, and in some cases, measuring the dielectric properties in-vivo. However, these studies have often produced results in direct conflict with historical studies, casting doubt on the accuracy of the currently utilised dielectric properties. At best, this uncertainty could significantly delay the development of electromagnetic imaging or therapeutic medical devices. At worst, the health dangers of electromagnetic radiation could be under-estimated. The applicant will embark upon frontier research to develop improved methods and standards for the measurement of the dielectric properties of biological tissue. The research programme will accelerate the design and development of electromagnetic imaging and therapeutic devices, at a time when the technology is gaining significant momentum. The primary objective of the research is to develop a deep understanding of the fundamental factors which contribute to errors in dielectric property measurement. These factors will include in-vivo/ex-vivo measurements and dielectric measurement method used, amongst many others. Secondly, a new open-access repository of dielectric measurements will be created based on a greatly enhanced understanding of the mechanisms underlying dielectric property measurement. Finally, new electromagnetic-based imaging and therapeutic medical devices will be investigated, based on the solid foundation of dielectric data.
Summary
The dielectric properties of biological tissues are of fundamental importance to the understanding of the interaction of electromagnetic fields with the human body. These properties are used to determine the safety of electronic devices, and in the design, development and refinement of electromagnetic medical imaging and therapeutic devices. Many historical studies have aimed to establish the dielectric properties of a broad range of tissues. A growing number of recent studies have sought to more accurately estimate these dielectric properties by standardising measurement procedures, and in some cases, measuring the dielectric properties in-vivo. However, these studies have often produced results in direct conflict with historical studies, casting doubt on the accuracy of the currently utilised dielectric properties. At best, this uncertainty could significantly delay the development of electromagnetic imaging or therapeutic medical devices. At worst, the health dangers of electromagnetic radiation could be under-estimated. The applicant will embark upon frontier research to develop improved methods and standards for the measurement of the dielectric properties of biological tissue. The research programme will accelerate the design and development of electromagnetic imaging and therapeutic devices, at a time when the technology is gaining significant momentum. The primary objective of the research is to develop a deep understanding of the fundamental factors which contribute to errors in dielectric property measurement. These factors will include in-vivo/ex-vivo measurements and dielectric measurement method used, amongst many others. Secondly, a new open-access repository of dielectric measurements will be created based on a greatly enhanced understanding of the mechanisms underlying dielectric property measurement. Finally, new electromagnetic-based imaging and therapeutic medical devices will be investigated, based on the solid foundation of dielectric data.
Max ERC Funding
1 499 329 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym BRAINCANNABINOIDS
Project Understanding the molecular blueprint and functional complexity of the endocannabinoid metabolome in the brain
Researcher (PI) István Katona
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Starting Grant (StG), LS5, ERC-2009-StG
Summary We and others have recently delineated the molecular architecture of a new feedback pathway in brain synapses, which operates as a synaptic circuit breaker. This pathway is supposed to use a group of lipid messengers as retrograde synaptic signals, the so-called endocannabinoids. Although heterogeneous in their chemical structures, these molecules along with the psychoactive compound in cannabis are thought to target the same effector in the brain, the CB1 receptor. However, the molecular catalog of these bioactive lipids and their metabolic enzymes has been expanding rapidly by recent advances in lipidomics and proteomics raising the possibility that these lipids may also serve novel, yet unidentified physiological functions. Thus, the overall aim of our research program is to define the molecular and anatomical organization of these endocannabinoid-mediated pathways and to determine their functional significance. In the present proposal, we will focus on understanding how these novel pathways regulate synaptic and extrasynaptic signaling in hippocampal neurons. Using combination of lipidomic, genetic and high-resolution anatomical approaches, we will identify distinct chemical species of endocannabinoids and will show how their metabolic enzymes are segregated into different subcellular compartments in cell type- and synapse-specific manner. Subsequently, we will use genetically encoded gain-of-function, loss-of-function and reporter constructs in imaging experiments and electrophysiological recordings to gain insights into the diverse tasks that these new pathways serve in synaptic transmission and extrasynaptic signal processing. Our proposed experiments will reveal fundamental principles of intercellular and intracellular endocannabinoid signaling in the brain.
Summary
We and others have recently delineated the molecular architecture of a new feedback pathway in brain synapses, which operates as a synaptic circuit breaker. This pathway is supposed to use a group of lipid messengers as retrograde synaptic signals, the so-called endocannabinoids. Although heterogeneous in their chemical structures, these molecules along with the psychoactive compound in cannabis are thought to target the same effector in the brain, the CB1 receptor. However, the molecular catalog of these bioactive lipids and their metabolic enzymes has been expanding rapidly by recent advances in lipidomics and proteomics raising the possibility that these lipids may also serve novel, yet unidentified physiological functions. Thus, the overall aim of our research program is to define the molecular and anatomical organization of these endocannabinoid-mediated pathways and to determine their functional significance. In the present proposal, we will focus on understanding how these novel pathways regulate synaptic and extrasynaptic signaling in hippocampal neurons. Using combination of lipidomic, genetic and high-resolution anatomical approaches, we will identify distinct chemical species of endocannabinoids and will show how their metabolic enzymes are segregated into different subcellular compartments in cell type- and synapse-specific manner. Subsequently, we will use genetically encoded gain-of-function, loss-of-function and reporter constructs in imaging experiments and electrophysiological recordings to gain insights into the diverse tasks that these new pathways serve in synaptic transmission and extrasynaptic signal processing. Our proposed experiments will reveal fundamental principles of intercellular and intracellular endocannabinoid signaling in the brain.
Max ERC Funding
1 638 000 €
Duration
Start date: 2009-11-01, End date: 2014-10-31
Project acronym BugTheDrug
Project Predicting the effects of gut microbiota and diet on an individual’s drug response and safety
Researcher (PI) Ines THIELE
Host Institution (HI) NATIONAL UNIVERSITY OF IRELAND GALWAY
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary Precision medicine is an emerging paradigm that aims at maximizing the benefits and minimizing the harm of drugs. Realistic mechanistic models are needed to understand and limit heterogeneity in drug responses. Consequently, novel approaches are required that explicitly account for individual variations in response to environmental influences, in addition to genetic variation. The human gut microbiota metabolizes drugs and is modulated by diet, and it exhibits significant variation among individuals. However, the influence of the gut microbiota on drug failure or drug side effects is under-researched. In this study, I will combine whole-body, genome-scale molecular resolution modeling of human metabolism and human gut microbial metabolism, which represents a network of genes, proteins, and biochemical reactions, with physiological, clinically relevant modeling of drug responses. I will perform two pilot studies on human subjects to illustrate that this innovative, versatile computational modeling framework can be used to stratify patients prior to drug prescription and to optimize drug bioavailability through personalized dietary intervention. With these studies, BugTheDrug will advance mechanistic understanding of drug-microbiota-diet interactions and their contribution to individual drug responses. I will perform the first integration of cutting-edge approaches and novel insights from four distinct research areas: systems biology, quantitative systems pharmacology, microbiology, and nutrition. BugTheDrug conceptually and technologically addresses the demand for novel approaches to the study of individual variability, thereby providing breakthrough support for progress in precision medicine.
Summary
Precision medicine is an emerging paradigm that aims at maximizing the benefits and minimizing the harm of drugs. Realistic mechanistic models are needed to understand and limit heterogeneity in drug responses. Consequently, novel approaches are required that explicitly account for individual variations in response to environmental influences, in addition to genetic variation. The human gut microbiota metabolizes drugs and is modulated by diet, and it exhibits significant variation among individuals. However, the influence of the gut microbiota on drug failure or drug side effects is under-researched. In this study, I will combine whole-body, genome-scale molecular resolution modeling of human metabolism and human gut microbial metabolism, which represents a network of genes, proteins, and biochemical reactions, with physiological, clinically relevant modeling of drug responses. I will perform two pilot studies on human subjects to illustrate that this innovative, versatile computational modeling framework can be used to stratify patients prior to drug prescription and to optimize drug bioavailability through personalized dietary intervention. With these studies, BugTheDrug will advance mechanistic understanding of drug-microbiota-diet interactions and their contribution to individual drug responses. I will perform the first integration of cutting-edge approaches and novel insights from four distinct research areas: systems biology, quantitative systems pharmacology, microbiology, and nutrition. BugTheDrug conceptually and technologically addresses the demand for novel approaches to the study of individual variability, thereby providing breakthrough support for progress in precision medicine.
Max ERC Funding
1 687 458 €
Duration
Start date: 2018-04-01, End date: 2023-03-31
Project acronym CholAminCo
Project Synergy and antagonism of cholinergic and dopaminergic systems in associative learning
Researcher (PI) Balazs Gyoergy HANGYA
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Starting Grant (StG), LS5, ERC-2016-STG
Summary Neuromodulators such as acetylcholine and dopamine are able to rapidly reprogram neuronal information processing and dynamically change brain states. Degeneration or dysfunction of cholinergic and dopaminergic neurons can lead to neuropsychiatric conditions like schizophrenia and addiction or cognitive diseases such as Alzheimer’s. Neuromodulatory systems control overlapping cognitive processes and often have similar modes of action; therefore it is important to reveal cooperation and competition between different systems to understand their unique contributions to cognitive functions like learning, memory and attention. This is only possible by direct comparison, which necessitates monitoring multiple neuromodulatory systems under identical experimental conditions. Moreover, simultaneous recording of different neuromodulatory cell types goes beyond phenomenological description of similarities and differences by revealing the underlying correlation structure at the level of action potential timing. However, such data allowing direct comparison of neuromodulatory actions are still sparse. As a first step to bridge this gap, I propose to elucidate the unique versus complementary roles of two “classical” neuromodulatory systems, the cholinergic and dopaminergic projection system implicated in various cognitive functions including associative learning and plasticity. First, we will record optogenetically identified cholinergic and dopaminergic neurons simultaneously using chronic extracellular recording in mice undergoing classical and operant conditioning. Second, we will determine the postsynaptic impact of cholinergic and dopaminergic neurons by manipulating them both separately and simultaneously while recording consequential changes in cortical neuronal activity and learning behaviour. These experiments will reveal how major neuromodulatory systems interact to mediate similar or different aspects of the same cognitive functions.
Summary
Neuromodulators such as acetylcholine and dopamine are able to rapidly reprogram neuronal information processing and dynamically change brain states. Degeneration or dysfunction of cholinergic and dopaminergic neurons can lead to neuropsychiatric conditions like schizophrenia and addiction or cognitive diseases such as Alzheimer’s. Neuromodulatory systems control overlapping cognitive processes and often have similar modes of action; therefore it is important to reveal cooperation and competition between different systems to understand their unique contributions to cognitive functions like learning, memory and attention. This is only possible by direct comparison, which necessitates monitoring multiple neuromodulatory systems under identical experimental conditions. Moreover, simultaneous recording of different neuromodulatory cell types goes beyond phenomenological description of similarities and differences by revealing the underlying correlation structure at the level of action potential timing. However, such data allowing direct comparison of neuromodulatory actions are still sparse. As a first step to bridge this gap, I propose to elucidate the unique versus complementary roles of two “classical” neuromodulatory systems, the cholinergic and dopaminergic projection system implicated in various cognitive functions including associative learning and plasticity. First, we will record optogenetically identified cholinergic and dopaminergic neurons simultaneously using chronic extracellular recording in mice undergoing classical and operant conditioning. Second, we will determine the postsynaptic impact of cholinergic and dopaminergic neurons by manipulating them both separately and simultaneously while recording consequential changes in cortical neuronal activity and learning behaviour. These experiments will reveal how major neuromodulatory systems interact to mediate similar or different aspects of the same cognitive functions.
Max ERC Funding
1 499 463 €
Duration
Start date: 2017-05-01, End date: 2022-04-30
Project acronym CHROMARRANGE
Project Programmed and unprogrammed genomic rearrangements during the evolution of yeast species
Researcher (PI) Kenneth Henry Wolfe
Host Institution (HI) UNIVERSITY COLLEGE DUBLIN, NATIONAL UNIVERSITY OF IRELAND, DUBLIN
Call Details Advanced Grant (AdG), LS2, ERC-2010-AdG_20100317
Summary By detailed evolutionary comparisons among multiple sequenced yeast genomes, we have identified several unusual regions where our preliminary evidence suggests that previously unknown molecular biology phenomena, involving rearrangement of genomic DNA, are occurring. I now propose to use a combination of dry-lab and wet-lab experimental approaches to characterize these regions and phenomena further. One region is a 24-kb section of chromosome XIV that appears to undergo recurrent 'flip/flop' inversion between two isomers at a fairly high rate in five species as diverse as Saccharomyces cerevisiae and Naumovia castellii, leading to a 1:1 ratio of the two isomers in each species. We hypothesize that this region is the site of a programmed DNA rearrangement analogous to mating-type switching. We have also identified two new genes related to the mating-type switching endonuclease HO, but different from it, that are potentially involved in rearrangement processes though not necessarily the inversion described above. We will determine the sites of action of these endonucleases. Separately, we have found evidence for a process of recurrent deletion of DNA from regions flanking the mating-type (MAT) locus in all yeast species that are descended from the whole-genome duplication (WGD) event, causing continual transpositions of genes from beside MAT to other locations in the genome. In related computational work, we propose to investigate an hypothesis that evolutionary loss of the MATa2 transcriptional activator may have been the cause of the WGD event.
Summary
By detailed evolutionary comparisons among multiple sequenced yeast genomes, we have identified several unusual regions where our preliminary evidence suggests that previously unknown molecular biology phenomena, involving rearrangement of genomic DNA, are occurring. I now propose to use a combination of dry-lab and wet-lab experimental approaches to characterize these regions and phenomena further. One region is a 24-kb section of chromosome XIV that appears to undergo recurrent 'flip/flop' inversion between two isomers at a fairly high rate in five species as diverse as Saccharomyces cerevisiae and Naumovia castellii, leading to a 1:1 ratio of the two isomers in each species. We hypothesize that this region is the site of a programmed DNA rearrangement analogous to mating-type switching. We have also identified two new genes related to the mating-type switching endonuclease HO, but different from it, that are potentially involved in rearrangement processes though not necessarily the inversion described above. We will determine the sites of action of these endonucleases. Separately, we have found evidence for a process of recurrent deletion of DNA from regions flanking the mating-type (MAT) locus in all yeast species that are descended from the whole-genome duplication (WGD) event, causing continual transpositions of genes from beside MAT to other locations in the genome. In related computational work, we propose to investigate an hypothesis that evolutionary loss of the MATa2 transcriptional activator may have been the cause of the WGD event.
Max ERC Funding
1 516 960 €
Duration
Start date: 2011-06-01, End date: 2016-05-31
Project acronym CODEKILLER
Project Killer plasmids as drivers of genetic code changes during yeast evolution
Researcher (PI) Kenneth WOLFE
Host Institution (HI) UNIVERSITY COLLEGE DUBLIN, NATIONAL UNIVERSITY OF IRELAND, DUBLIN
Call Details Advanced Grant (AdG), LS8, ERC-2017-ADG
Summary The genetic code was established at a very early stage during the evolution of life on Earth and is nearly universal. In eukaryotic nuclear genes, the only known examples of a sense codon that underwent an evolutionary change of meaning, from one amino acid to another, occur in yeast species. The codon CUG is translated as Leu in the universal genetic code, but it has long been known to be translated as Ser in some Candida species. In recent work, we discovered that this switch is one of three parallel reassignments of CUG that occurred in three closely related clades of yeasts. CUG was reassigned once from Leu to Ala, and twice from Leu to Ser, in three separate events. The meaning of sense codons in the nuclear genetic code has otherwise remained completely stable during all of eukaryotic evolution, so why was CUG so unstable in yeasts? CODEKILLER will test a radical new hypothesis that the genetic code changes were caused by a killer toxin that specifically attacked the tRNA that translated CUG as Leu. The hypothesis implies that the reassignments of CUG were not driven by selection in favor of their effects on the proteome, as commonly assumed, but by selection against the existence of a particular tRNA. As well as searching for this killer toxin, we will study the detailed mechanism of genetic code change by engineering a reversal of a CUG-Ser species back to CUG-Leu translation, and investigate translation in some species that naturally contain both tRNA-Leu and tRNA-Ser molecules capable of decoding CUG.
Summary
The genetic code was established at a very early stage during the evolution of life on Earth and is nearly universal. In eukaryotic nuclear genes, the only known examples of a sense codon that underwent an evolutionary change of meaning, from one amino acid to another, occur in yeast species. The codon CUG is translated as Leu in the universal genetic code, but it has long been known to be translated as Ser in some Candida species. In recent work, we discovered that this switch is one of three parallel reassignments of CUG that occurred in three closely related clades of yeasts. CUG was reassigned once from Leu to Ala, and twice from Leu to Ser, in three separate events. The meaning of sense codons in the nuclear genetic code has otherwise remained completely stable during all of eukaryotic evolution, so why was CUG so unstable in yeasts? CODEKILLER will test a radical new hypothesis that the genetic code changes were caused by a killer toxin that specifically attacked the tRNA that translated CUG as Leu. The hypothesis implies that the reassignments of CUG were not driven by selection in favor of their effects on the proteome, as commonly assumed, but by selection against the existence of a particular tRNA. As well as searching for this killer toxin, we will study the detailed mechanism of genetic code change by engineering a reversal of a CUG-Ser species back to CUG-Leu translation, and investigate translation in some species that naturally contain both tRNA-Leu and tRNA-Ser molecules capable of decoding CUG.
Max ERC Funding
2 368 356 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym COSIP
Project Clarifying Optimal Sodium Intake Project
Researcher (PI) Martin James O'Donnell
Host Institution (HI) NATIONAL UNIVERSITY OF IRELAND GALWAY
Call Details Starting Grant (StG), LS7, ERC-2014-STG
Summary Hypertension is a leading risk factor for cardiovascular disease (CVD) globally, accounting for 25-35% of the population-attributable fraction. Sodium (salt) intake is a key determinant of blood pressure, and reducing sodium intake has emerged as an important target for population-based interventions to prevent CVD. However, there is considerable uncertainty about the optimal level of sodium (salt) intake that is associated with lowest CVD risk, and whether optimal levels differ for different populations and individuals. In this proposal, we will answer key fundamental research questions about the association of sodium intake with blood pressure and CVD risk. Our research challenges current guideline recommendations of low-sodium intake for all populations. Specifically, we will: a) determine whether sustained (long-term) low sodium intake is associated with beneficial (or adverse) effects on established and novel CV biomarkers. b) explore whether inter-daily ‘pattern’ of sodium intake is an important determinant of 24-hour blood pressure pattern; c) determine whether the association between sodium intake and CVD varies by ethnicity, sex, age, other dietary factors (e.g. potassium intake), or other factors in 2 large international epidemiologic studies (PURE and INTERSTROKE; n>125,000 individuals). d) quantify the population-attributable fraction of excess sodium intake on global burden of CVD (stroke, myocardial infarction, heart failure and CV death), and model the potential impact of various population-based approaches to reducing sodium intake; e) determine whether sodium intake is associated with other vascular-related clinical conditions, namely including atrial fibrillation, cognitive impairment and falls (providing novel information); f) determine whether genetic variants associated with ‘salt sensitivity’ and hypertension are association with blood pressure and stroke, and whether these associations are modified by sodium intake.
Summary
Hypertension is a leading risk factor for cardiovascular disease (CVD) globally, accounting for 25-35% of the population-attributable fraction. Sodium (salt) intake is a key determinant of blood pressure, and reducing sodium intake has emerged as an important target for population-based interventions to prevent CVD. However, there is considerable uncertainty about the optimal level of sodium (salt) intake that is associated with lowest CVD risk, and whether optimal levels differ for different populations and individuals. In this proposal, we will answer key fundamental research questions about the association of sodium intake with blood pressure and CVD risk. Our research challenges current guideline recommendations of low-sodium intake for all populations. Specifically, we will: a) determine whether sustained (long-term) low sodium intake is associated with beneficial (or adverse) effects on established and novel CV biomarkers. b) explore whether inter-daily ‘pattern’ of sodium intake is an important determinant of 24-hour blood pressure pattern; c) determine whether the association between sodium intake and CVD varies by ethnicity, sex, age, other dietary factors (e.g. potassium intake), or other factors in 2 large international epidemiologic studies (PURE and INTERSTROKE; n>125,000 individuals). d) quantify the population-attributable fraction of excess sodium intake on global burden of CVD (stroke, myocardial infarction, heart failure and CV death), and model the potential impact of various population-based approaches to reducing sodium intake; e) determine whether sodium intake is associated with other vascular-related clinical conditions, namely including atrial fibrillation, cognitive impairment and falls (providing novel information); f) determine whether genetic variants associated with ‘salt sensitivity’ and hypertension are association with blood pressure and stroke, and whether these associations are modified by sodium intake.
Max ERC Funding
1 499 431 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym CREST
Project Enrichment of macular pigment, and its impact on vision and blindness
Researcher (PI) John Michael Nolan
Host Institution (HI) WATERFORD INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Age-related macular degeneration (AMD) is the leading cause of blindness in the developed world. The macula, the central part of the retina, is responsible for optimal spatial vision. There is a growing body of evidence that a lack of a dietary pigment at the macula, known as macular pigment (MP), is associated with increased risk of AMD.
MP contains the carotenoids lutein (L), zeaxanthin (Z) and meso-zeaxanthin (meso-Z). The typical western diet contains around 60 carotenoids, and 18 have been identified in human serum. However, only three are found at the macula, indicating the unique biological selectivity for their uptake at this location. The function of MP remains undetermined. It is likely that the accumulation of MP has evolved because of its optical and antioxidant properties; for example, MP limits retinal oxidative damage passively (through filtration of blue light) and actively (by quenching free radicals). Furthermore, its optical properties suggest a key role for MP in enhancing visual performance and supporting ‘super’ vision by reducing the effects of chromatic aberration and light scatter.
Recent research has shown that MP can be augmented by dietary supplementation in most (but not all) subjects, suggesting that the macular concentrations of these carotenoids are suboptimal in many people. My laboratory has discovered that a dip in the central portion of this pigment, seen in around 12% of individuals, is an undesirable feature of its spatial profile and may be linked to an inability to generate meso-Z at the macula. However, we have identified that enrichment of MP can be achieved by inclusion of meso-Z in a dietary supplement.
We propose to uniquely enrich MP and assess its impact on visual performance in normal subjects and visual function in patients with AMD. This groundbreaking study will advance our understanding of the protective and optical hypothesis of MP, and potentially improve normal vision and prevent or delay blindness due to AMD.
Summary
Age-related macular degeneration (AMD) is the leading cause of blindness in the developed world. The macula, the central part of the retina, is responsible for optimal spatial vision. There is a growing body of evidence that a lack of a dietary pigment at the macula, known as macular pigment (MP), is associated with increased risk of AMD.
MP contains the carotenoids lutein (L), zeaxanthin (Z) and meso-zeaxanthin (meso-Z). The typical western diet contains around 60 carotenoids, and 18 have been identified in human serum. However, only three are found at the macula, indicating the unique biological selectivity for their uptake at this location. The function of MP remains undetermined. It is likely that the accumulation of MP has evolved because of its optical and antioxidant properties; for example, MP limits retinal oxidative damage passively (through filtration of blue light) and actively (by quenching free radicals). Furthermore, its optical properties suggest a key role for MP in enhancing visual performance and supporting ‘super’ vision by reducing the effects of chromatic aberration and light scatter.
Recent research has shown that MP can be augmented by dietary supplementation in most (but not all) subjects, suggesting that the macular concentrations of these carotenoids are suboptimal in many people. My laboratory has discovered that a dip in the central portion of this pigment, seen in around 12% of individuals, is an undesirable feature of its spatial profile and may be linked to an inability to generate meso-Z at the macula. However, we have identified that enrichment of MP can be achieved by inclusion of meso-Z in a dietary supplement.
We propose to uniquely enrich MP and assess its impact on visual performance in normal subjects and visual function in patients with AMD. This groundbreaking study will advance our understanding of the protective and optical hypothesis of MP, and potentially improve normal vision and prevent or delay blindness due to AMD.
Max ERC Funding
1 493 342 €
Duration
Start date: 2011-10-01, End date: 2016-09-30
Project acronym DeCode
Project Dendrites and memory: role of dendritic spikes in information coding by hippocampal CA3 pyramidal neurons
Researcher (PI) Judit MAKARA
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Consolidator Grant (CoG), LS5, ERC-2017-COG
Summary The hippocampus is essential for building episodic memories. Coding of locations, contexts or events in the hippocampus is based on the correlated activity of neuronal ensembles; however, the mechanisms promoting the recruitment of individual neurons into information-coding ensembles are poorly understood.
In particular, the recurrent synaptic network of pyramidal cells (PCs) in the hippocampal CA3 area, receiving external inputs from the entorhinal cortex and the dentate gyrus, is thought to be essential for associative memory. Current models of the associative functions of CA3 are mainly based on plasticity of these synaptic connections. Recent work by us and others however suggests that active, voltage-dependent properties of CA3PC dendrites may also promote ensemble functions. Dendritic voltage-dependent ion channels allow nonlinear amplification of spatiotemporally correlated synaptic inputs (such as those produced by ensemble activity) and can even generate local dendritic spikes, which may elicit specific action potential patterns and induce synaptic plasticity. Furthermore, dendritic processing may be modulated by activity-dependent regulation of dendritic ion channels. However, still little is known about the active properties of CA3PC dendrites and their functions during spatial coding or memory tasks.
The general aim of my research program is to understand the cellular mechanisms that underlie the formation of hippocampal memory-coding neuronal ensembles. Specifically, we will test the hypothesis that active input integration by dendrites of individual CA3PCs plays an important role in their recruitment into specific context-coding ensembles. By combining in vitro (patch-clamp electrophysiology and two-photon (2P) microscopy in slices) and in vivo (2P imaging and activity-dependent labelling in behaving rodents) approaches, we will provide an in-depth understanding of the dendritic components contributing to the generation of the CA3 ensemble code.
Summary
The hippocampus is essential for building episodic memories. Coding of locations, contexts or events in the hippocampus is based on the correlated activity of neuronal ensembles; however, the mechanisms promoting the recruitment of individual neurons into information-coding ensembles are poorly understood.
In particular, the recurrent synaptic network of pyramidal cells (PCs) in the hippocampal CA3 area, receiving external inputs from the entorhinal cortex and the dentate gyrus, is thought to be essential for associative memory. Current models of the associative functions of CA3 are mainly based on plasticity of these synaptic connections. Recent work by us and others however suggests that active, voltage-dependent properties of CA3PC dendrites may also promote ensemble functions. Dendritic voltage-dependent ion channels allow nonlinear amplification of spatiotemporally correlated synaptic inputs (such as those produced by ensemble activity) and can even generate local dendritic spikes, which may elicit specific action potential patterns and induce synaptic plasticity. Furthermore, dendritic processing may be modulated by activity-dependent regulation of dendritic ion channels. However, still little is known about the active properties of CA3PC dendrites and their functions during spatial coding or memory tasks.
The general aim of my research program is to understand the cellular mechanisms that underlie the formation of hippocampal memory-coding neuronal ensembles. Specifically, we will test the hypothesis that active input integration by dendrites of individual CA3PCs plays an important role in their recruitment into specific context-coding ensembles. By combining in vitro (patch-clamp electrophysiology and two-photon (2P) microscopy in slices) and in vivo (2P imaging and activity-dependent labelling in behaving rodents) approaches, we will provide an in-depth understanding of the dendritic components contributing to the generation of the CA3 ensemble code.
Max ERC Funding
1 990 314 €
Duration
Start date: 2018-06-01, End date: 2023-05-31
Project acronym DOSE
Project Dosage sensitive genes in evolution and disease
Researcher (PI) Aoife Mclysaght
Host Institution (HI) THE PROVOST, FELLOWS, FOUNDATION SCHOLARS & THE OTHER MEMBERS OF BOARD OF THE COLLEGE OF THE HOLY & UNDIVIDED TRINITY OF QUEEN ELIZABETH NEAR DUBLIN
Call Details Starting Grant (StG), LS8, ERC-2012-StG_20111109
Summary Evolutionary change of gene copy number through gene duplication is a relatively pervasive phenomenon in eukaryotic genomes. However, for a subset of genes such changes are deleterious because they result in imbalances in the cell. Such dosage-sensitive genes have been increasingly implicated in disease, particularly through the association of copy number variants (CNVs) with pathogenicity.
In my lab we have previously discovered that many genes in the human genome which were retained after whole genome duplication (WGD) are refractory to gene duplication both over evolutionary timescales and within populations. These are expected characteristics of dosage-balanced genes. Many of these genes are implicated in human disease. I now propose to take a computational (dry-lab) approach to examine the evolution of dosage-balanced genes further and to develop a sophisticated model of evolutionary constraint of copy number. These models will enable the identification of dosage-balanced genes and their consideration as novel candidate disease loci.
Recognising and interpreting patterns of constraint is the cornerstone of molecular evolution. Through careful analysis of genome sequences with respect to gene duplication over evolutionary times and within populations, we will develop a formal and generalised model of copy-number evolution and constraint. We will use these models to identify candidate disease loci within pathogenic CNVs. We will also study the characteristics of known disease genes in order to identify novel candidate loci for dosage-dependent disease.
This is an ambitious and high impact project that has the potential to yield major insights into gene copy-number constraint and its relationship to complex disease.
Summary
Evolutionary change of gene copy number through gene duplication is a relatively pervasive phenomenon in eukaryotic genomes. However, for a subset of genes such changes are deleterious because they result in imbalances in the cell. Such dosage-sensitive genes have been increasingly implicated in disease, particularly through the association of copy number variants (CNVs) with pathogenicity.
In my lab we have previously discovered that many genes in the human genome which were retained after whole genome duplication (WGD) are refractory to gene duplication both over evolutionary timescales and within populations. These are expected characteristics of dosage-balanced genes. Many of these genes are implicated in human disease. I now propose to take a computational (dry-lab) approach to examine the evolution of dosage-balanced genes further and to develop a sophisticated model of evolutionary constraint of copy number. These models will enable the identification of dosage-balanced genes and their consideration as novel candidate disease loci.
Recognising and interpreting patterns of constraint is the cornerstone of molecular evolution. Through careful analysis of genome sequences with respect to gene duplication over evolutionary times and within populations, we will develop a formal and generalised model of copy-number evolution and constraint. We will use these models to identify candidate disease loci within pathogenic CNVs. We will also study the characteristics of known disease genes in order to identify novel candidate loci for dosage-dependent disease.
This is an ambitious and high impact project that has the potential to yield major insights into gene copy-number constraint and its relationship to complex disease.
Max ERC Funding
1 358 534 €
Duration
Start date: 2013-01-01, End date: 2018-12-31
Project acronym DOUBLE EXPRESS
Project Gene expression level as a keystone to understanding gene duplication: evolutionary constraints, opportunities, and disease
Researcher (PI) Aoife MCLYSAGHT
Host Institution (HI) THE PROVOST, FELLOWS, FOUNDATION SCHOLARS & THE OTHER MEMBERS OF BOARD OF THE COLLEGE OF THE HOLY & UNDIVIDED TRINITY OF QUEEN ELIZABETH NEAR DUBLIN
Call Details Consolidator Grant (CoG), LS8, ERC-2017-COG
Summary Duplicate genes are important in disease, are a hugely important source of evolutionary novelty, and for many years we thought we understood them. We thought that duplication relieved selective constraints. We thought that gene knockout neutrality was due to redundancy. We thought that a duplicate is a duplicate is a duplicate. Evidence is accumulating challenging each of these views. Rather than being the result of an unbiased process, the genes that tend to duplicate in our genome and others are quickly evolving, non-essential genes, irrespective of current duplication status. Conversely, genes retained after whole genome duplication (WGD) are slowly evolving, important genes.
I propose that different resolution of the evolutionary constraints imposed by the demands of gene expression can explain these contrasting relationships. I propose that the opposing constraints on gene-by-gene duplications as compared to WGD channel these different sets of genes into remarkably different evolutionary trajectories. In particular, in much the same way that individual gene duplication creates an opportunity for the evolution of a new gene, the co-evolution of expression of sets of interacting genes after WGD creates an opportunity for the evolution of new biochemical pathways and protein complexes. Furthermore, I suggest a common mechanism of pathogenicity for many duplication events independent of the biochemical function of the encoded genes.
With the availability of abundant high-quality genomics data, now is an opportune time to address these questions. Primarily through computational and statistical analysis I will reveal the relationship between gene duplication and expression and test a model that the indirect costs of gene expression are a major determinant of the outcome of gene duplication. I will explore the effects this has on gene and genome evolution. Finally, I will link the patterns of gene expression and duplicability to pathogenic effects.
Summary
Duplicate genes are important in disease, are a hugely important source of evolutionary novelty, and for many years we thought we understood them. We thought that duplication relieved selective constraints. We thought that gene knockout neutrality was due to redundancy. We thought that a duplicate is a duplicate is a duplicate. Evidence is accumulating challenging each of these views. Rather than being the result of an unbiased process, the genes that tend to duplicate in our genome and others are quickly evolving, non-essential genes, irrespective of current duplication status. Conversely, genes retained after whole genome duplication (WGD) are slowly evolving, important genes.
I propose that different resolution of the evolutionary constraints imposed by the demands of gene expression can explain these contrasting relationships. I propose that the opposing constraints on gene-by-gene duplications as compared to WGD channel these different sets of genes into remarkably different evolutionary trajectories. In particular, in much the same way that individual gene duplication creates an opportunity for the evolution of a new gene, the co-evolution of expression of sets of interacting genes after WGD creates an opportunity for the evolution of new biochemical pathways and protein complexes. Furthermore, I suggest a common mechanism of pathogenicity for many duplication events independent of the biochemical function of the encoded genes.
With the availability of abundant high-quality genomics data, now is an opportune time to address these questions. Primarily through computational and statistical analysis I will reveal the relationship between gene duplication and expression and test a model that the indirect costs of gene expression are a major determinant of the outcome of gene duplication. I will explore the effects this has on gene and genome evolution. Finally, I will link the patterns of gene expression and duplicability to pathogenic effects.
Max ERC Funding
1 824 794 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym EVOLECOCOG
Project The evolutionary ecology of cognition across a heterogeneous landscape
Researcher (PI) John Leo Quinn
Host Institution (HI) UNIVERSITY COLLEGE CORK - NATIONAL UNIVERSITY OF IRELAND, CORK
Call Details Consolidator Grant (CoG), LS8, ERC-2013-CoG
Summary "Why do individuals vary in their cognitive abilities? This proposal takes the disciplines of cognition and evolutionary biology into a natural setting to answer this question by investigating a variety of proximate causes and population-level consequences of individual variation in cognitive ability. It represents the first large-scale integrative study of cognitive ability on any wild population. State of the art observational (remote sensing and automated self-administration trials of learning in the wild), chemical (stable isotope analysis of diet), physiological (stress, energetics, immunocompetence), molecular (DNA fingerprinting and metabarcoding) and analytical (reaction norm, quantitative genetic) techniques will be used. The chosen study system, the great tit Parus major, is one of the most widely used in Europe, but uniquely here will consist of 12 subpopulations across deciduous and conifer woodland fragments. The proposal’s broad scope is captured in three objectives: 1) To characterise proximate causes of variation in cognitive (associative/reversal learning; problem solving; brain size) and other traits (the reactive-proactive personality axis; bill morphology), all of which can influence similar ecologically important behaviour. Quantitative genetic, social, parasite-mediated, and physiological causes will be explored. 2) To examine links between these traits, and key behaviours and trade-offs, e.g., space use, niche specialization, predation, parental care and promiscuity; and 3) To examine the consequences of this variation for life histories and fitness. The research team consists of the PI, five early career biologists, and three PhD students, and will collaborate with eight researchers from Europe and further afield. The project will reveal ground-breaking insight into why individuals vary in their cognitive ability. It aims to impact a wide scientific community, to raise public interest in science, and to inform EU biodiversity policy."
Summary
"Why do individuals vary in their cognitive abilities? This proposal takes the disciplines of cognition and evolutionary biology into a natural setting to answer this question by investigating a variety of proximate causes and population-level consequences of individual variation in cognitive ability. It represents the first large-scale integrative study of cognitive ability on any wild population. State of the art observational (remote sensing and automated self-administration trials of learning in the wild), chemical (stable isotope analysis of diet), physiological (stress, energetics, immunocompetence), molecular (DNA fingerprinting and metabarcoding) and analytical (reaction norm, quantitative genetic) techniques will be used. The chosen study system, the great tit Parus major, is one of the most widely used in Europe, but uniquely here will consist of 12 subpopulations across deciduous and conifer woodland fragments. The proposal’s broad scope is captured in three objectives: 1) To characterise proximate causes of variation in cognitive (associative/reversal learning; problem solving; brain size) and other traits (the reactive-proactive personality axis; bill morphology), all of which can influence similar ecologically important behaviour. Quantitative genetic, social, parasite-mediated, and physiological causes will be explored. 2) To examine links between these traits, and key behaviours and trade-offs, e.g., space use, niche specialization, predation, parental care and promiscuity; and 3) To examine the consequences of this variation for life histories and fitness. The research team consists of the PI, five early career biologists, and three PhD students, and will collaborate with eight researchers from Europe and further afield. The project will reveal ground-breaking insight into why individuals vary in their cognitive ability. It aims to impact a wide scientific community, to raise public interest in science, and to inform EU biodiversity policy."
Max ERC Funding
1 993 189 €
Duration
Start date: 2015-03-01, End date: 2020-12-31
Project acronym EVOLOR
Project Cognitive Ageing in Dogs
Researcher (PI) Eniko Kubinyi
Host Institution (HI) EOTVOS LORAND TUDOMANYEGYETEM
Call Details Starting Grant (StG), LS9, ERC-2015-STG
Summary The aim of this project is to understand the causal factors contributing to the cognitive decline during senescence and to develop sensitive and standardized behaviour tests for early detection in order to increase the welfare of affected species. With the rapidly ageing population of Europe, related research is a priority in the European Union.
We will focus both on characterising the ageing phenotype and the underlying biological processes in dogs as a well-established natural animal model. We develop a reliable and valid test battery applying innovative multidisciplinary methods (e.g. eye-tracking, motion path analysis, identification of behaviour using inertial sensors, EEG, fMRI, candidate gene, and epigenetics) in both longitudinal and cross-sectional studies. We expect to reveal specific environmental risk factors which hasten ageing and also protective factors which may postpone it. We aim to provide objective criteria (behavioural, physiological and genetic biomarkers) to assess and predict the ageing trajectory for specific individual dogs. This would help veterinarians to recognise the symptoms early, and initiate necessary counter actions.
This approach establishes the framework for answering the broad question that how we can extend the healthy life of ageing dogs which indirectly also contributes to the welfare of the owner and decreases veterinary expenses. The detailed description of the ageing phenotype may also facilitate the use of dogs as a natural model for human senescence, including the development and application of pharmaceutical interventions.
We expect that our approach offers the scientific foundation to delay the onset of cognitive ageing in dog populations by 1-2 years, and also increase the proportion of dogs that enjoy healthy ageing.
Summary
The aim of this project is to understand the causal factors contributing to the cognitive decline during senescence and to develop sensitive and standardized behaviour tests for early detection in order to increase the welfare of affected species. With the rapidly ageing population of Europe, related research is a priority in the European Union.
We will focus both on characterising the ageing phenotype and the underlying biological processes in dogs as a well-established natural animal model. We develop a reliable and valid test battery applying innovative multidisciplinary methods (e.g. eye-tracking, motion path analysis, identification of behaviour using inertial sensors, EEG, fMRI, candidate gene, and epigenetics) in both longitudinal and cross-sectional studies. We expect to reveal specific environmental risk factors which hasten ageing and also protective factors which may postpone it. We aim to provide objective criteria (behavioural, physiological and genetic biomarkers) to assess and predict the ageing trajectory for specific individual dogs. This would help veterinarians to recognise the symptoms early, and initiate necessary counter actions.
This approach establishes the framework for answering the broad question that how we can extend the healthy life of ageing dogs which indirectly also contributes to the welfare of the owner and decreases veterinary expenses. The detailed description of the ageing phenotype may also facilitate the use of dogs as a natural model for human senescence, including the development and application of pharmaceutical interventions.
We expect that our approach offers the scientific foundation to delay the onset of cognitive ageing in dog populations by 1-2 years, and also increase the proportion of dogs that enjoy healthy ageing.
Max ERC Funding
1 202 500 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
Project acronym FORCEMAP
Project Intramolecular force mapping of enzymes in action: the role of strain in motor mechanisms
Researcher (PI) András Málnási-Csizmadia
Host Institution (HI) EOTVOS LORAND TUDOMANYEGYETEM
Call Details Starting Grant (StG), LS1, ERC-2007-StG
Summary A fundamental but unexplored problem in biology is whether and how enzymes use mechanical strain during their functioning. It is now evident that the knowledge of atomic structures and chemical interactions is not sufficient to understand the intricate mechanisms underlying enzyme specificity and efficiency. Several lines of evidence suggest that mechanical effects play crucial roles in enzyme activity. Therefore we aim to create detailed force maps that reveal how the intramolecular distribution of mechanical strains changes during the enzyme cycle and how these rearrangements drive the enzyme processes. The applicability of current nanotechniques for the investigation of this problem is limited because they do not allow simultaneous measurement of mechanical and enzymatic parameters. Thus we seek to open new avenues of research by developing site-specific sensors and passive or photoinducible molecular springs to measure force-dependent chemical/structural changes with high spatiotemporal resolution in myosin. Since force perturbations occur very rapidly, we are able to combine experimental studies with quasi-realistic in silico simulations to describe the physical background of enzyme function. We expect that our research will yield fundamental insights into the role of intramolecular strains in enzymes and thus greatly aid the design and control of enzyme processes (specificity, activity, regulation). Our studies may also lead to new paradigms in the understanding of motor systems.
Summary
A fundamental but unexplored problem in biology is whether and how enzymes use mechanical strain during their functioning. It is now evident that the knowledge of atomic structures and chemical interactions is not sufficient to understand the intricate mechanisms underlying enzyme specificity and efficiency. Several lines of evidence suggest that mechanical effects play crucial roles in enzyme activity. Therefore we aim to create detailed force maps that reveal how the intramolecular distribution of mechanical strains changes during the enzyme cycle and how these rearrangements drive the enzyme processes. The applicability of current nanotechniques for the investigation of this problem is limited because they do not allow simultaneous measurement of mechanical and enzymatic parameters. Thus we seek to open new avenues of research by developing site-specific sensors and passive or photoinducible molecular springs to measure force-dependent chemical/structural changes with high spatiotemporal resolution in myosin. Since force perturbations occur very rapidly, we are able to combine experimental studies with quasi-realistic in silico simulations to describe the physical background of enzyme function. We expect that our research will yield fundamental insights into the role of intramolecular strains in enzymes and thus greatly aid the design and control of enzyme processes (specificity, activity, regulation). Our studies may also lead to new paradigms in the understanding of motor systems.
Max ERC Funding
750 000 €
Duration
Start date: 2008-09-01, End date: 2014-08-31
Project acronym FRONTHAL
Project Specificity of cortico-thalamic interactions and its role in frontal cortical functions
Researcher (PI) Laszlo ACSADY
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Advanced Grant (AdG), LS5, ERC-2016-ADG
Summary Frontal cortical areas are responsible for a wide range of executive and cognitive functions. Frontal cortices communicate with the thalamus via bidirectional pathways and these connections are indispensable for frontal cortical operations. Still, we have very little information about the specificity of connections, synaptic interactions and plasticity between frontal cortex and thalamus and the roles of these interactions in frontal cortical functions.
In the present proposal, we will test the hypothesis that frontal cortical areas developed a highly specialized connectivity pattern with the thalamus. This supports unique interactions between the cortex and the thalamus according to the specific requirements of frontal cortical activity, including experience-dependent plastic changes.
The project will use cell type-specific viral tracing in mice and 3D electron microscopic reconstructions in mice and humans to identify circuit motifs that are evolutionarily conserved, yet, still specific to fronto-thalamic pathways. The physiological approach will employ in vivo optogenetics combined with intra-, juxta- and extracellular recordings. We will perform behavioral experiments by bidirectional modulation of well-defined elements in the network, in learning paradigms, which depend on the integrity of frontal cortex.
The project is the first systematic approach which aims to understand the nature of interaction between the frontal cortex and the thalamus. It will not only fill the tremendous gap in our knowledge regarding these pathways but will help us elucidate the functional organization of non-sensory thalamus in general.
Frontal cortices are involved in a wide range of major neurological disorders (e.g. Parkinson’s disease, epilepsy, schizophrenia, chronic pain) which affect executive functions and involve fronto-thalamic pathways. We believe that understanding fronto-thalamic interactions will lead to fundamentally novel insight into the nature of these diseases.
Summary
Frontal cortical areas are responsible for a wide range of executive and cognitive functions. Frontal cortices communicate with the thalamus via bidirectional pathways and these connections are indispensable for frontal cortical operations. Still, we have very little information about the specificity of connections, synaptic interactions and plasticity between frontal cortex and thalamus and the roles of these interactions in frontal cortical functions.
In the present proposal, we will test the hypothesis that frontal cortical areas developed a highly specialized connectivity pattern with the thalamus. This supports unique interactions between the cortex and the thalamus according to the specific requirements of frontal cortical activity, including experience-dependent plastic changes.
The project will use cell type-specific viral tracing in mice and 3D electron microscopic reconstructions in mice and humans to identify circuit motifs that are evolutionarily conserved, yet, still specific to fronto-thalamic pathways. The physiological approach will employ in vivo optogenetics combined with intra-, juxta- and extracellular recordings. We will perform behavioral experiments by bidirectional modulation of well-defined elements in the network, in learning paradigms, which depend on the integrity of frontal cortex.
The project is the first systematic approach which aims to understand the nature of interaction between the frontal cortex and the thalamus. It will not only fill the tremendous gap in our knowledge regarding these pathways but will help us elucidate the functional organization of non-sensory thalamus in general.
Frontal cortices are involved in a wide range of major neurological disorders (e.g. Parkinson’s disease, epilepsy, schizophrenia, chronic pain) which affect executive functions and involve fronto-thalamic pathways. We believe that understanding fronto-thalamic interactions will lead to fundamentally novel insight into the nature of these diseases.
Max ERC Funding
1 597 575 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym FunctionalProteomics
Project Proteomic fingerprinting of functionally characterized single synapses
Researcher (PI) Zoltan NUSSER
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Advanced Grant (AdG), LS5, ERC-2017-ADG
Summary Our astonishing cognitive abilities are the consequence of complex connectivity within our neuronal networks and the large functional diversity of excitable nerve cells and their synapses. Investigations over the past half a century revealed dramatic diversity in shape, size and functional properties among synapses established by distinct cell types in different brain regions and demonstrated that the functional differences are partly due to different molecular mechanisms. However, synaptic diversity is also observed among synapses established by molecularly and morphologically uniform presynaptic cells on molecularly and morphologically uniform postsynaptic cells. Our hypothesis is that quantitative molecular differences underlie the functional diversity of such synapses. We will focus on hippocampal CA1 pyramidal cell (PC) to mGluR1α+ O-LM cell synapses, which show remarkable functional and molecular heterogeneity. In vitro multiple cell patch-clamp recordings followed by quantal analysis will be performed to quantify well-defined biophysical properties of these synapses. The molecular composition of the functionally characterized single synapses will be determined following the development of a novel postembedding immunolocalization method. Correlations between the molecular content and functional properties will be established and genetic up- and downregulation of individual synaptic proteins will be conducted to reveal causal relationships. Finally, correlations of the activity history and the functional properties of the synapses will be established by performing in vivo two-photon Ca2+ imaging in head-fixed behaving animals followed by in vitro functional characterization of their synapses. Our results will reveal quantitative molecular fingerprints of functional properties, allowing us to render dynamic behaviour to billions of synapses when the connectome of the hippocampal circuit is created using array tomography.
Summary
Our astonishing cognitive abilities are the consequence of complex connectivity within our neuronal networks and the large functional diversity of excitable nerve cells and their synapses. Investigations over the past half a century revealed dramatic diversity in shape, size and functional properties among synapses established by distinct cell types in different brain regions and demonstrated that the functional differences are partly due to different molecular mechanisms. However, synaptic diversity is also observed among synapses established by molecularly and morphologically uniform presynaptic cells on molecularly and morphologically uniform postsynaptic cells. Our hypothesis is that quantitative molecular differences underlie the functional diversity of such synapses. We will focus on hippocampal CA1 pyramidal cell (PC) to mGluR1α+ O-LM cell synapses, which show remarkable functional and molecular heterogeneity. In vitro multiple cell patch-clamp recordings followed by quantal analysis will be performed to quantify well-defined biophysical properties of these synapses. The molecular composition of the functionally characterized single synapses will be determined following the development of a novel postembedding immunolocalization method. Correlations between the molecular content and functional properties will be established and genetic up- and downregulation of individual synaptic proteins will be conducted to reveal causal relationships. Finally, correlations of the activity history and the functional properties of the synapses will be established by performing in vivo two-photon Ca2+ imaging in head-fixed behaving animals followed by in vitro functional characterization of their synapses. Our results will reveal quantitative molecular fingerprints of functional properties, allowing us to render dynamic behaviour to billions of synapses when the connectome of the hippocampal circuit is created using array tomography.
Max ERC Funding
2 498 750 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym GENECLOCKS
Project Reconstructing a dated tree of life using phylogenetic incongruence
Researcher (PI) Gergely Janos SZOLLOSI
Host Institution (HI) EOTVOS LORAND TUDOMANYEGYETEM
Call Details Starting Grant (StG), LS8, ERC-2016-STG
Summary With the advent of genome-scale sequencing, molecular phylogeny, which reconstructs gene trees from homologous sequences, has reached an impasse. Instead of answering open questions, new genomes have reignited old debates. The problem is clear, gene trees are not species trees, each is the unique result of series of evolutionary events. If, however, we model these differences in the context of a common species tree, we can access a wealth of information on genome evolution and the diversification of species that is not available to traditional methods. For example, as horizontal gene transfer (HGT) can only occur between coexisting species, HGTs provide information on the order of speciations. When HGT is rare, lineage sorting can generate incongruence between gene trees and the dating problem can be formulated in terms of biologically meaningful parameters (such as population size), that are informative on the rate of evolution and hence invaluable to molecular dating.
My first goal is to develop methods that systematically extract information on the pattern and timing of genomic evolution by explaining differences between gene trees. This will allow us to, for the first time, reconstruct a dated tree of life from genome-scale data. We will use parallel programming to maximise the number of genomes analysed.
My second goal is to apply these methods to open problems, e.g.: i) to resolve the timing of microbial evolution and its relationship to Earth history, where the extreme paucity of fossils limits the use of molecular dating methods, by using HGT events as “molecular fossils”; ii) to reconstruct rooted phylogenies from complete genomes and harness phylogenetic incongruence to answer long standing questions, such as the of diversification of animals or the position of eukaryotes among archaea; and iii) for eukaryotic groups such as Fungi, where evidence of significant amounts of HGT is emerging our methods will also allow the quantification of the extent of HGT.
Summary
With the advent of genome-scale sequencing, molecular phylogeny, which reconstructs gene trees from homologous sequences, has reached an impasse. Instead of answering open questions, new genomes have reignited old debates. The problem is clear, gene trees are not species trees, each is the unique result of series of evolutionary events. If, however, we model these differences in the context of a common species tree, we can access a wealth of information on genome evolution and the diversification of species that is not available to traditional methods. For example, as horizontal gene transfer (HGT) can only occur between coexisting species, HGTs provide information on the order of speciations. When HGT is rare, lineage sorting can generate incongruence between gene trees and the dating problem can be formulated in terms of biologically meaningful parameters (such as population size), that are informative on the rate of evolution and hence invaluable to molecular dating.
My first goal is to develop methods that systematically extract information on the pattern and timing of genomic evolution by explaining differences between gene trees. This will allow us to, for the first time, reconstruct a dated tree of life from genome-scale data. We will use parallel programming to maximise the number of genomes analysed.
My second goal is to apply these methods to open problems, e.g.: i) to resolve the timing of microbial evolution and its relationship to Earth history, where the extreme paucity of fossils limits the use of molecular dating methods, by using HGT events as “molecular fossils”; ii) to reconstruct rooted phylogenies from complete genomes and harness phylogenetic incongruence to answer long standing questions, such as the of diversification of animals or the position of eukaryotes among archaea; and iii) for eukaryotic groups such as Fungi, where evidence of significant amounts of HGT is emerging our methods will also allow the quantification of the extent of HGT.
Max ERC Funding
1 453 542 €
Duration
Start date: 2017-07-01, End date: 2022-06-30
Project acronym Human Decisions
Project The Neural Determinants of Perceptual Decision Making in the Human Brain
Researcher (PI) Redmond O'connell
Host Institution (HI) THE PROVOST, FELLOWS, FOUNDATION SCHOLARS & THE OTHER MEMBERS OF BOARD OF THE COLLEGE OF THE HOLY & UNDIVIDED TRINITY OF QUEEN ELIZABETH NEAR DUBLIN
Call Details Starting Grant (StG), LS5, ERC-2014-STG
Summary How do we make reliable decisions given sensory information that is often weak or ambiguous? Current theories center on a brain mechanism whereby sensory evidence is integrated over time into a “decision variable” which triggers the appropriate action upon reaching a criterion. Neural signals fitting this role have been identified in monkey electrophysiology but efforts to study the neural dynamics underpinning human decision making have been hampered by technical challenges associated with non-invasive recording. This proposal builds on a recent paradigm breakthrough made by the applicant that enables parallel tracking of discrete neural signals that can be unambiguously linked to the three key information processing stages necessary for simple perceptual decisions: sensory encoding, decision formation and motor preparation. Chief among these is a freely-evolving decision variable signal which builds at an evidence-dependent rate up to an action-triggering threshold and precisely determines the timing and accuracy of perceptual reports at the single-trial level. This provides an unprecedented neurophysiological window onto the distinct parameters of the human decision process such that the underlying mechanisms of several major behavioral phenomena can finally be investigated. This proposal seeks to develop a systems-level understanding of perceptual decision making in the human brain by tackling three core questions: 1) what are the neural adaptations that allow us to deal with speed pressure and variations in the reliability of the physically presented evidence? 2) What neural mechanism determines our subjective confidence in a decision? and 3) How does aging impact on the distinct neural components underpinning perceptual decision making? Each of the experiments described in this proposal will definitively test key predictions from prominent theoretical models using a combination of temporally precise neurophysiological measurement and psychophysical modelling.
Summary
How do we make reliable decisions given sensory information that is often weak or ambiguous? Current theories center on a brain mechanism whereby sensory evidence is integrated over time into a “decision variable” which triggers the appropriate action upon reaching a criterion. Neural signals fitting this role have been identified in monkey electrophysiology but efforts to study the neural dynamics underpinning human decision making have been hampered by technical challenges associated with non-invasive recording. This proposal builds on a recent paradigm breakthrough made by the applicant that enables parallel tracking of discrete neural signals that can be unambiguously linked to the three key information processing stages necessary for simple perceptual decisions: sensory encoding, decision formation and motor preparation. Chief among these is a freely-evolving decision variable signal which builds at an evidence-dependent rate up to an action-triggering threshold and precisely determines the timing and accuracy of perceptual reports at the single-trial level. This provides an unprecedented neurophysiological window onto the distinct parameters of the human decision process such that the underlying mechanisms of several major behavioral phenomena can finally be investigated. This proposal seeks to develop a systems-level understanding of perceptual decision making in the human brain by tackling three core questions: 1) what are the neural adaptations that allow us to deal with speed pressure and variations in the reliability of the physically presented evidence? 2) What neural mechanism determines our subjective confidence in a decision? and 3) How does aging impact on the distinct neural components underpinning perceptual decision making? Each of the experiments described in this proposal will definitively test key predictions from prominent theoretical models using a combination of temporally precise neurophysiological measurement and psychophysical modelling.
Max ERC Funding
1 382 643 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym iHEAR
Project Investigating the meanings and mechanisms of psychotic experiences in young people: a novel, mixed-methods approach
Researcher (PI) Mary CANNON
Host Institution (HI) ROYAL COLLEGE OF SURGEONS IN IRELAND
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary Up to one fifth of young people have had the experience of psychotic symptoms, such as hearing voices when there is no-one around, or seeing visions. We now know that young people who experience these symptoms are at increased risk of developing psychotic disorders in adulthood. We also know that these young people are at higher risk of a range of co-morbid disorders such as depression and anxiety, and particularly suicidal behaviours. On the other hand, many of these young people will remain well and, for them, the psychotic experiences were merely a transitory phenomenon.
Childhood trauma is known to be associated with increased risk for psychotic symptoms and is a promising target for intervention. However we do not yet know enough about what types or timing of stressors are involved in the pathogenesis of psychotic symptoms, nor the mechanism by which early life stress may lead to changes in brain structure and function resulting in symptoms such as hallucinations. We also need to be able to identify those young people who will benefit most from intervention.
This ground-breaking, multi-disciplinary programme of work sets out to address these issues by drawing together epidemiology, social science, anthropology and neuroscience to devise a comprehensive programme of work examining the relationship between early life stress and psychotic symptoms among young people.
Designed as three inter-related work packages, this iHEAR programme will exploit a large population-based cohort and will capitalise on my existing unique cohort of young people, who were known to have experienced psychotic symptoms in childhood, as they enter young adulthood. This iHEAR programme will result in new information which will allow the development of innovative interventions to prevent or pre-empt severe mental illness in later life.
Summary
Up to one fifth of young people have had the experience of psychotic symptoms, such as hearing voices when there is no-one around, or seeing visions. We now know that young people who experience these symptoms are at increased risk of developing psychotic disorders in adulthood. We also know that these young people are at higher risk of a range of co-morbid disorders such as depression and anxiety, and particularly suicidal behaviours. On the other hand, many of these young people will remain well and, for them, the psychotic experiences were merely a transitory phenomenon.
Childhood trauma is known to be associated with increased risk for psychotic symptoms and is a promising target for intervention. However we do not yet know enough about what types or timing of stressors are involved in the pathogenesis of psychotic symptoms, nor the mechanism by which early life stress may lead to changes in brain structure and function resulting in symptoms such as hallucinations. We also need to be able to identify those young people who will benefit most from intervention.
This ground-breaking, multi-disciplinary programme of work sets out to address these issues by drawing together epidemiology, social science, anthropology and neuroscience to devise a comprehensive programme of work examining the relationship between early life stress and psychotic symptoms among young people.
Designed as three inter-related work packages, this iHEAR programme will exploit a large population-based cohort and will capitalise on my existing unique cohort of young people, who were known to have experienced psychotic symptoms in childhood, as they enter young adulthood. This iHEAR programme will result in new information which will allow the development of innovative interventions to prevent or pre-empt severe mental illness in later life.
Max ERC Funding
1 781 623 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym INTERIMPACT
Project Impact of identified interneurons on cellular network mechanisms in the human and rodent neocortex
Researcher (PI) Gábor Tamás
Host Institution (HI) Szegedi Tudomanyegyetem - Hungarian-Netherlands School of Educational Management
Call Details Advanced Grant (AdG), LS5, ERC-2010-AdG_20100317
Summary This application addresses mechanisms linking the activity of single neurons with network events by defining the function of identified cell types in the cerebral cortex. The key hypotheses emerged from our experiments and propose that neurogliaform cells and axo-axonic cells achieve their function in the cortex through extreme forms of unspecificity and specificity, respectively. The project capitalizes on our discovery that neurogliaform cells reach GABAA and GABAB receptors on target cells through unitary volume transmission going beyond the classical theory which states that single cortical neurons act in or around synaptic junctions. We propose that the spatial unspecificity of neurotransmitter action leads to unprecedented functional capabilities for a single neuron simultaneously acting on neuronal, glial and vascular components of the surrounding area allowing neurogliaform cells to synchronize metabolic demand and supply in microcircuits. In contrast, axo-axonic cells represent extreme spatial specificity in the brain: terminals of axo-axonic cells exclusively target the axon initial segment of pyramidal neurons. Axo-axonic cells were considered as the most potent inhibitory neurons of the cortex. However, our experiments suggested that axo-axonic cells can be the most powerful excitatory neurons known to date by triggering complex network events. Our unprecedented recordings in the human cortex show that axo-axonic cells are crucial in activating functional assemblies which were implicated in higher order cognitive representations. We aim to define interactions between active cortical networks and axo-axonic cell triggered assemblies with an emphasis on mechanisms modulated by neurogliaform cells and commonly prescribed drugs.
Summary
This application addresses mechanisms linking the activity of single neurons with network events by defining the function of identified cell types in the cerebral cortex. The key hypotheses emerged from our experiments and propose that neurogliaform cells and axo-axonic cells achieve their function in the cortex through extreme forms of unspecificity and specificity, respectively. The project capitalizes on our discovery that neurogliaform cells reach GABAA and GABAB receptors on target cells through unitary volume transmission going beyond the classical theory which states that single cortical neurons act in or around synaptic junctions. We propose that the spatial unspecificity of neurotransmitter action leads to unprecedented functional capabilities for a single neuron simultaneously acting on neuronal, glial and vascular components of the surrounding area allowing neurogliaform cells to synchronize metabolic demand and supply in microcircuits. In contrast, axo-axonic cells represent extreme spatial specificity in the brain: terminals of axo-axonic cells exclusively target the axon initial segment of pyramidal neurons. Axo-axonic cells were considered as the most potent inhibitory neurons of the cortex. However, our experiments suggested that axo-axonic cells can be the most powerful excitatory neurons known to date by triggering complex network events. Our unprecedented recordings in the human cortex show that axo-axonic cells are crucial in activating functional assemblies which were implicated in higher order cognitive representations. We aim to define interactions between active cortical networks and axo-axonic cell triggered assemblies with an emphasis on mechanisms modulated by neurogliaform cells and commonly prescribed drugs.
Max ERC Funding
2 391 695 €
Duration
Start date: 2011-06-01, End date: 2017-05-31
Project acronym MEME
Project Memory Engram Maintenance and Expression
Researcher (PI) Tomas RYAN
Host Institution (HI) THE PROVOST, FELLOWS, FOUNDATION SCHOLARS & THE OTHER MEMBERS OF BOARD OF THE COLLEGE OF THE HOLY & UNDIVIDED TRINITY OF QUEEN ELIZABETH NEAR DUBLIN
Call Details Starting Grant (StG), LS5, ERC-2016-STG
Summary The goal of this project is to understand how specific memory engrams are physically stored in the brain. Connectionist theories of memory storage have guided research into the neuroscience of memory for over a half century, but have received little direct proof due to experimental limitations. The major confound that has limited direct testing of such theories has been an inability to identify the cells and circuits that store specific memories. Memory engram technology, which allows the tagging and in vivo manipulation of specific engram cells, has recently allowed us to overcome this empirical limitation and has revolutionised the way memory can be studied in rodent models. Based on our research it is now known that sparse populations of hippocampal neurons that were active during a defined learning experience are both sufficient and necessary for retrieval of specific contextual memories. More recently we have established that hippocampal engram cells preferentially synapse directly onto postsynaptic engram cells. This “engram cell connectivity” could provide the neurobiological substrate for the storage of multimodal memories through a distributed engram circuit. However it is currently unknown whether engram cell connectivity itself is important for memory function. The proposed integrative neuroscience project will employ inter-disciplinary methods to directly probe the importance of engram cell connectivity for memory retrieval, storage, and encoding. The outcomes will directly inform a novel and comprehensive neurobiological model of memory engram storage.
Summary
The goal of this project is to understand how specific memory engrams are physically stored in the brain. Connectionist theories of memory storage have guided research into the neuroscience of memory for over a half century, but have received little direct proof due to experimental limitations. The major confound that has limited direct testing of such theories has been an inability to identify the cells and circuits that store specific memories. Memory engram technology, which allows the tagging and in vivo manipulation of specific engram cells, has recently allowed us to overcome this empirical limitation and has revolutionised the way memory can be studied in rodent models. Based on our research it is now known that sparse populations of hippocampal neurons that were active during a defined learning experience are both sufficient and necessary for retrieval of specific contextual memories. More recently we have established that hippocampal engram cells preferentially synapse directly onto postsynaptic engram cells. This “engram cell connectivity” could provide the neurobiological substrate for the storage of multimodal memories through a distributed engram circuit. However it is currently unknown whether engram cell connectivity itself is important for memory function. The proposed integrative neuroscience project will employ inter-disciplinary methods to directly probe the importance of engram cell connectivity for memory retrieval, storage, and encoding. The outcomes will directly inform a novel and comprehensive neurobiological model of memory engram storage.
Max ERC Funding
1 500 000 €
Duration
Start date: 2017-02-01, End date: 2022-01-31
Project acronym MicroCONtACT
Project Microglial control of neuronal activity in the healthy and the injured brain
Researcher (PI) Adam DENES
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Consolidator Grant (CoG), LS5, ERC-2016-COG
Summary Microglia are the main immune cells of the brain, but their role in brain injury is highly controversial due to the difficulties in selectively manipulating and imaging microglial actions in real time. Specifically, it is unclear whether microglia control neuronal survival after injury via shaping the activity of complex neuronal networks in vivo. To this end, we have combined fast in vivo two-photon imaging of neuronal calcium responses with selective microglial manipulation for the first time. Our data suggest that microglia constantly monitor and control neuronal network activity and these actions are essential to limit excitotoxicity and neuronal death after acute brain injury. We also identify microglia as key regulators of spreading depolarization in vivo. However, the underlying mechanisms remained unexplored. Here, I propose that microglia control neuronal excitability and based on preliminary data I set out to investigate how this occurs. We will combine selective, CSF1R-mediated microglia depletion with advanced neurophysiological methods such as in vivo calcium imaging and intracranial EEG for the first time, to reveal how microglia shape activity of complex neuronal networks in the healthy and the injured brain. Then, we will study microglia-neuron interactions from the network level to nanoscale level using in vivo two-photon imaging and super-resolution microscopy. We will apply novel chemogenic and optogenetic approaches to manipulate microglia in real time, assess their role in neuronal activity changes and investigate the molecular mechanisms in vitro and in vivo. Our unpublished data also suggest that inflammation – a key contributor to brain diseases – could disrupt microglia-neuron signaling and we set out to investigate the underlying mechanisms. By using state-of the-art research tools that had not been applied previously in this context, our studies are likely to reveal novel pathophysiological mechanisms relevant for common brain diseases.
Summary
Microglia are the main immune cells of the brain, but their role in brain injury is highly controversial due to the difficulties in selectively manipulating and imaging microglial actions in real time. Specifically, it is unclear whether microglia control neuronal survival after injury via shaping the activity of complex neuronal networks in vivo. To this end, we have combined fast in vivo two-photon imaging of neuronal calcium responses with selective microglial manipulation for the first time. Our data suggest that microglia constantly monitor and control neuronal network activity and these actions are essential to limit excitotoxicity and neuronal death after acute brain injury. We also identify microglia as key regulators of spreading depolarization in vivo. However, the underlying mechanisms remained unexplored. Here, I propose that microglia control neuronal excitability and based on preliminary data I set out to investigate how this occurs. We will combine selective, CSF1R-mediated microglia depletion with advanced neurophysiological methods such as in vivo calcium imaging and intracranial EEG for the first time, to reveal how microglia shape activity of complex neuronal networks in the healthy and the injured brain. Then, we will study microglia-neuron interactions from the network level to nanoscale level using in vivo two-photon imaging and super-resolution microscopy. We will apply novel chemogenic and optogenetic approaches to manipulate microglia in real time, assess their role in neuronal activity changes and investigate the molecular mechanisms in vitro and in vivo. Our unpublished data also suggest that inflammation – a key contributor to brain diseases – could disrupt microglia-neuron signaling and we set out to investigate the underlying mechanisms. By using state-of the-art research tools that had not been applied previously in this context, our studies are likely to reveal novel pathophysiological mechanisms relevant for common brain diseases.
Max ERC Funding
2 000 000 €
Duration
Start date: 2017-04-01, End date: 2022-03-31
Project acronym MOLECMAP
Project Quantitative Molecular Map of the Neuronal Surface
Researcher (PI) Zoltan Jozsef Nusser
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Advanced Grant (AdG), LS5, ERC-2011-ADG_20110310
Summary The most fundamental roles of nerve cells are the detection of chemical neurotransmitters to generate synaptic potentials; the summation of these potentials to create their output signals; and the consequent release of their own neurotransmitter molecules. All of these functions require the orchestrated work of hundreds of molecules targeted to specialized regions of the cells. In nerve cells, more than in any other cell type, a single molecule could fulfill very different functional roles depending on its subcellular location. For example, dendritic voltage-gated Ca2+ channels play a role in the integration and plasticity of synaptic inputs, whereas the same channels when concentrated in presynaptic active zones are essential for neurotransmitter release. Thus, the function of a protein in nerve cells cannot be understood from its expression or lack of it, but its precise subcellular location, density and molecular environment needs to be determined. The major aim of the present proposal is to create a quantitative molecular map of the surface of hippocampal pyramidal cells (PCs). We will start by examining voltage-gated ion channels due to their pivotal roles in input summation, output generation and neurotransmitter release. We will apply high resolution quantitative molecular neuroanatomical techniques to reveal their densities in 19 different axo-somato-dendritic plasma membrane compartments of CA1 PCs. Functional predictions will be generated using detailed, morphologically realistic multicompartmental PC models with experimentally determined ion channel distributions and densities. Such predictions will be tested by combining in vitro patch-clamp electrophysiology and imaging techniques with correlated light- and electron microscopy. Our results will provide the first quantitative molecular map of the neuronal surface and will reveal new mechanisms that increase the computational power and the functional diversity of nerve cells.
Summary
The most fundamental roles of nerve cells are the detection of chemical neurotransmitters to generate synaptic potentials; the summation of these potentials to create their output signals; and the consequent release of their own neurotransmitter molecules. All of these functions require the orchestrated work of hundreds of molecules targeted to specialized regions of the cells. In nerve cells, more than in any other cell type, a single molecule could fulfill very different functional roles depending on its subcellular location. For example, dendritic voltage-gated Ca2+ channels play a role in the integration and plasticity of synaptic inputs, whereas the same channels when concentrated in presynaptic active zones are essential for neurotransmitter release. Thus, the function of a protein in nerve cells cannot be understood from its expression or lack of it, but its precise subcellular location, density and molecular environment needs to be determined. The major aim of the present proposal is to create a quantitative molecular map of the surface of hippocampal pyramidal cells (PCs). We will start by examining voltage-gated ion channels due to their pivotal roles in input summation, output generation and neurotransmitter release. We will apply high resolution quantitative molecular neuroanatomical techniques to reveal their densities in 19 different axo-somato-dendritic plasma membrane compartments of CA1 PCs. Functional predictions will be generated using detailed, morphologically realistic multicompartmental PC models with experimentally determined ion channel distributions and densities. Such predictions will be tested by combining in vitro patch-clamp electrophysiology and imaging techniques with correlated light- and electron microscopy. Our results will provide the first quantitative molecular map of the neuronal surface and will reveal new mechanisms that increase the computational power and the functional diversity of nerve cells.
Max ERC Funding
2 494 446 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym MOLINFLAM
Project Molecular dissection of inflammatory pathways
Researcher (PI) Attila Mocsai
Host Institution (HI) SEMMELWEIS EGYETEM
Call Details Starting Grant (StG), LS3, ERC-2007-StG
Summary Inflammatory diseases are highly prevalent, often chronic diseases that cause diminished quality of life and are connected with major causes of death in Western societies. Despite their societal impact, their pathomechanism is incompletely understood, hindering development of novel therapeutic strategies. In particular, little is known about the intracellular signal transduction processes involved in the tissue destruction phase of aggressive autoimmune diseases such as rheumatoid arthritis. The present proposal aims to clarify this issue using in vivo and in vitro studies on genetically manipulated mice. During the proposed studies, mice deficient in various signal transduction molecules such as Syk, PLCg2, Gab2 and p190 RhoGAPs will be used to test their contribution to inflammatory responses. In vitro studies will test the activation of major effector cells of inflammation (neutrophils, macrophages and osteoclasts) while in vivo studies will utilize mouse models such as autoantibody- and cytokine-induced inflammatory arthritis or autoantibody-induced glomerulonephritis. Further studies will be performed to test the contribution of the above signaling molecules to disease pathogenesis in a lineage-restricted manner, using the Cre-lox approach. Finally, wild type and mutant versions of the signaling molecules tested will be retrovirally re-expressed into the relevant knockout hematopoietic stem cells in vivo to allow structure-function studies during in vivo inflammation. Two novel transgenic strains and a knock-in (floxed) mutant will also be generated during the course of the project. Using state-of-the-art approaches and techniques, this project will provide information at unprecedented molecular detail on signal transduction mechanisms involved in inflammatory diseases, and is expected to point to possible future targets of novel anti-inflammatory therapies.
Summary
Inflammatory diseases are highly prevalent, often chronic diseases that cause diminished quality of life and are connected with major causes of death in Western societies. Despite their societal impact, their pathomechanism is incompletely understood, hindering development of novel therapeutic strategies. In particular, little is known about the intracellular signal transduction processes involved in the tissue destruction phase of aggressive autoimmune diseases such as rheumatoid arthritis. The present proposal aims to clarify this issue using in vivo and in vitro studies on genetically manipulated mice. During the proposed studies, mice deficient in various signal transduction molecules such as Syk, PLCg2, Gab2 and p190 RhoGAPs will be used to test their contribution to inflammatory responses. In vitro studies will test the activation of major effector cells of inflammation (neutrophils, macrophages and osteoclasts) while in vivo studies will utilize mouse models such as autoantibody- and cytokine-induced inflammatory arthritis or autoantibody-induced glomerulonephritis. Further studies will be performed to test the contribution of the above signaling molecules to disease pathogenesis in a lineage-restricted manner, using the Cre-lox approach. Finally, wild type and mutant versions of the signaling molecules tested will be retrovirally re-expressed into the relevant knockout hematopoietic stem cells in vivo to allow structure-function studies during in vivo inflammation. Two novel transgenic strains and a knock-in (floxed) mutant will also be generated during the course of the project. Using state-of-the-art approaches and techniques, this project will provide information at unprecedented molecular detail on signal transduction mechanisms involved in inflammatory diseases, and is expected to point to possible future targets of novel anti-inflammatory therapies.
Max ERC Funding
1 200 000 €
Duration
Start date: 2008-10-01, End date: 2014-03-31
Project acronym Multicellularity
Project The genetic basis of the convergent evolution of fungal multicellularity
Researcher (PI) Laszlo NAGY
Host Institution (HI) MAGYAR TUDOMANYOS AKADEMIA SZEGEDIBIOLOGIAI KUTATOKOZPONT
Call Details Starting Grant (StG), LS8, ERC-2017-STG
Summary The evolution of multicellularity (MC) has been one of the major transitions in the history of life. Despite immense interest in its evolutionary origins, the genomic changes leading to the emergence of MC, especially that of complex MC (differentiated 3-dimensional structures) are poorly known. Previous comparative genomics projects aiming to understand the genetic bases of MC in one way or another relied on gene content-based analyses. However, a pattern emerging from these studies is that gene content provides only an incomplete explanation for the evolution of MC even at ancient timescales. We hypothesize that besides gene duplications, changes to cis-regulatory elements and gene expression patterns (including protein isoforms) have significantly contributed to the evolution of MC. To test this hypothesis, we will deploy a combination of computational methods, phylogenomics, comparative transcriptomics and genome-wide assays of regulatory elements. Our research focuses on fungi as a model system, where complex MC evolved convergently and in subsequent two steps. Fungi are ideal models to tackle this question for several reasons: a) multicellularity in fungi evolved multiple times, b) there are rich genomic resources (>500 complete genomes), c) complex multicellular structures can be routinely grown in the lab and d) genetic manipulations are feasible for several cornerstone species. We set out to examine which genes participate in the building of simple and complex multicellular structures and whether the evolution of regulome complexity and gene expression patterns can explain the evolution of MC better than can traditionally assayed sources of genetic innovations (e.g. gene duplications). Ultimately, our goal is to reach a general synthesis on the genetic bases of the evolution of MC and that of organismal complexity.
Summary
The evolution of multicellularity (MC) has been one of the major transitions in the history of life. Despite immense interest in its evolutionary origins, the genomic changes leading to the emergence of MC, especially that of complex MC (differentiated 3-dimensional structures) are poorly known. Previous comparative genomics projects aiming to understand the genetic bases of MC in one way or another relied on gene content-based analyses. However, a pattern emerging from these studies is that gene content provides only an incomplete explanation for the evolution of MC even at ancient timescales. We hypothesize that besides gene duplications, changes to cis-regulatory elements and gene expression patterns (including protein isoforms) have significantly contributed to the evolution of MC. To test this hypothesis, we will deploy a combination of computational methods, phylogenomics, comparative transcriptomics and genome-wide assays of regulatory elements. Our research focuses on fungi as a model system, where complex MC evolved convergently and in subsequent two steps. Fungi are ideal models to tackle this question for several reasons: a) multicellularity in fungi evolved multiple times, b) there are rich genomic resources (>500 complete genomes), c) complex multicellular structures can be routinely grown in the lab and d) genetic manipulations are feasible for several cornerstone species. We set out to examine which genes participate in the building of simple and complex multicellular structures and whether the evolution of regulome complexity and gene expression patterns can explain the evolution of MC better than can traditionally assayed sources of genetic innovations (e.g. gene duplications). Ultimately, our goal is to reach a general synthesis on the genetic bases of the evolution of MC and that of organismal complexity.
Max ERC Funding
1 486 500 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym nanoAXON
Project Nano-physiology of small glutamatergic axon terminals
Researcher (PI) Janos SZABADICS
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Consolidator Grant (CoG), LS5, ERC-2017-COG
Summary We will reveal the neuronal mechanisms of fundamental hippocampal and axonal functions using direct patch clamp recordings from the small axon terminals of the major glutamatergic afferent and efferent pathways of the dentate gyrus region. Specifically, we will investigate the intrinsic axonal properties and unitary synaptic functions of the axons in the dentate gyrus that originate from the entorhinal cortex, the hilar mossy cells and the hypothalamic supramammillary nucleus. The fully controlled access to the activity of individual neuronal projections allows us to address the crucial questions how upstream regions of the dentate gyrus convey physiologically relevant spike activities and how these activities are translated to unitary synaptic responses in individual dentate gyrus neurons. The successful information transfers by these mechanisms ultimately generate specific dentate gyrus cell activity that contributes to hippocampal memory functions. Comprehensive mechanistic insights are essential to understand the impacts of the activity patterns associated with fundamental physiological functions and attainable with the necessary details only with direct recordings from individual axons. For example, these knowledge are necessary to understand how single cell activities in the entorhinal cortex (carrying primary spatial information) contribute to spatial representation in the dentate (i.e. place fields). Furthermore, because the size of these recorded axon terminals matches that of the majority of cortical synapses, our discoveries will demonstrate basic biophysical and neuronal principles of axonal signaling that are relevant for universal neuronal functions throughout the CNS. Thus, an exceptional repertoire of methods, including recording from anatomically identified individual small axon terminals, voltage- and calcium imaging and computational simulations, places us in an advantaged position for revealing unprecedented information about neuronal circuits.
Summary
We will reveal the neuronal mechanisms of fundamental hippocampal and axonal functions using direct patch clamp recordings from the small axon terminals of the major glutamatergic afferent and efferent pathways of the dentate gyrus region. Specifically, we will investigate the intrinsic axonal properties and unitary synaptic functions of the axons in the dentate gyrus that originate from the entorhinal cortex, the hilar mossy cells and the hypothalamic supramammillary nucleus. The fully controlled access to the activity of individual neuronal projections allows us to address the crucial questions how upstream regions of the dentate gyrus convey physiologically relevant spike activities and how these activities are translated to unitary synaptic responses in individual dentate gyrus neurons. The successful information transfers by these mechanisms ultimately generate specific dentate gyrus cell activity that contributes to hippocampal memory functions. Comprehensive mechanistic insights are essential to understand the impacts of the activity patterns associated with fundamental physiological functions and attainable with the necessary details only with direct recordings from individual axons. For example, these knowledge are necessary to understand how single cell activities in the entorhinal cortex (carrying primary spatial information) contribute to spatial representation in the dentate (i.e. place fields). Furthermore, because the size of these recorded axon terminals matches that of the majority of cortical synapses, our discoveries will demonstrate basic biophysical and neuronal principles of axonal signaling that are relevant for universal neuronal functions throughout the CNS. Thus, an exceptional repertoire of methods, including recording from anatomically identified individual small axon terminals, voltage- and calcium imaging and computational simulations, places us in an advantaged position for revealing unprecedented information about neuronal circuits.
Max ERC Funding
1 994 025 €
Duration
Start date: 2018-04-01, End date: 2023-03-31
Project acronym NETWORK EVOLUTION
Project Integrated evolutionary analyses of genetic and drug interaction networks in yeast
Researcher (PI) Csaba Pal
Host Institution (HI) MAGYAR TUDOMANYOS AKADEMIA SZEGEDIBIOLOGIAI KUTATOKOZPONT
Call Details Starting Grant (StG), LS5, ERC-2007-StG
Summary The ability of cellular systems to adapt to genetic and environmental perturbations is a fundamental but poorly understood process both at the molecular and evolutionary level. There are both physiological and evolutionary reasonings why mutations often have limited impact on cellular growth. First, perturbations that hit one target often have no effect on the overall performance of a complex system (such as metabolic networks), as perturbations can be adjusted by reorganizing fluxes in metabolic networks, or changing regulation and expression of genes. Second, due to the fast evolvability of microbes, the effect of a perturbation can readily be alleviated by the evolution of compensatory mutations at other sites of the network. Understanding the extent of intrinsic and evolved robustness in cellular systems demands integrated analyses that combine functional genomics and computational systems biology with microbial evolutionary experiments. In collaboration with several leading research teams in the field, we plan to investigate the following issues. First, we will ask how accurately genome-scale metabolic network models can predict the impact of genetic deletions and other non-heritable perturbations. Second, to understand how the impact of genetic and drug perturbations can be mitigated during evolution, we will pursue a large-scale lab evolutionary protocol, and compare the results with predictions of computational models. Our work may suggest avenues of research on the general rules of acquired drug resistance in microbes.
Summary
The ability of cellular systems to adapt to genetic and environmental perturbations is a fundamental but poorly understood process both at the molecular and evolutionary level. There are both physiological and evolutionary reasonings why mutations often have limited impact on cellular growth. First, perturbations that hit one target often have no effect on the overall performance of a complex system (such as metabolic networks), as perturbations can be adjusted by reorganizing fluxes in metabolic networks, or changing regulation and expression of genes. Second, due to the fast evolvability of microbes, the effect of a perturbation can readily be alleviated by the evolution of compensatory mutations at other sites of the network. Understanding the extent of intrinsic and evolved robustness in cellular systems demands integrated analyses that combine functional genomics and computational systems biology with microbial evolutionary experiments. In collaboration with several leading research teams in the field, we plan to investigate the following issues. First, we will ask how accurately genome-scale metabolic network models can predict the impact of genetic deletions and other non-heritable perturbations. Second, to understand how the impact of genetic and drug perturbations can be mitigated during evolution, we will pursue a large-scale lab evolutionary protocol, and compare the results with predictions of computational models. Our work may suggest avenues of research on the general rules of acquired drug resistance in microbes.
Max ERC Funding
1 280 000 €
Duration
Start date: 2008-07-01, End date: 2013-06-30
Project acronym NewSpindleForce
Project A new class of microtubules in the spindle exerting forces on kinetochores
Researcher (PI) Iva Marija Tolic
Host Institution (HI) RUDER BOSKOVIC INSTITUTE
Call Details Consolidator Grant (CoG), LS3, ERC-2014-CoG
Summary At the onset of division the cell forms a spindle, a micro-machine made of microtubules, which divide the chromosomes by pulling on kinetochores, protein complexes on the chromosome. The central question in the field is how accurate chromosome segregation results from the interactions between kinetochores, microtubules and the associated proteins. According to the current paradigm, the forces on kinetochores are produced by k-fibers, bundles of microtubules extending between the spindle pole and the kinetochore. The proposed project is built upon a groundbreaking hypothesis that a new class of microtubules, which we term bridging microtubules, bridge sister kinetochores. Our preliminary results show that bridging microtubules are responsible for the positioning of kinetochores in HeLa and PtK1 cells. Bridging microtubules have not been studied before because this requires cutting-edge microscopy and laser microsurgery techniques. By applying these methods, with which I have extensive expertise, we will determine the organization of these microtubules, identify the proteins that link them with k-fibers, and uncover where and how the forces for kinetochore positioning and movement are generated. My strength is in taking an interdisciplinary approach, which I will use in this project by combining laser microsurgery with genetic perturbations, quantitative measurements of the responses and comparison with theoretical models. Understanding the role of bridging microtubules in force generation and chromosome movements will not only shed light on the mechanism of chromosome segregation, but may also increase the potential of mitotic anticancer strategies, as the spindle is a major target for chemotherapy. The proposed ERC funding is essential for the success of these timely and ambitious experiments, allowing me to strengthen my position as an international leader in research on cell division, thereby increasing Europe's foremost position in this field.
Summary
At the onset of division the cell forms a spindle, a micro-machine made of microtubules, which divide the chromosomes by pulling on kinetochores, protein complexes on the chromosome. The central question in the field is how accurate chromosome segregation results from the interactions between kinetochores, microtubules and the associated proteins. According to the current paradigm, the forces on kinetochores are produced by k-fibers, bundles of microtubules extending between the spindle pole and the kinetochore. The proposed project is built upon a groundbreaking hypothesis that a new class of microtubules, which we term bridging microtubules, bridge sister kinetochores. Our preliminary results show that bridging microtubules are responsible for the positioning of kinetochores in HeLa and PtK1 cells. Bridging microtubules have not been studied before because this requires cutting-edge microscopy and laser microsurgery techniques. By applying these methods, with which I have extensive expertise, we will determine the organization of these microtubules, identify the proteins that link them with k-fibers, and uncover where and how the forces for kinetochore positioning and movement are generated. My strength is in taking an interdisciplinary approach, which I will use in this project by combining laser microsurgery with genetic perturbations, quantitative measurements of the responses and comparison with theoretical models. Understanding the role of bridging microtubules in force generation and chromosome movements will not only shed light on the mechanism of chromosome segregation, but may also increase the potential of mitotic anticancer strategies, as the spindle is a major target for chemotherapy. The proposed ERC funding is essential for the success of these timely and ambitious experiments, allowing me to strengthen my position as an international leader in research on cell division, thereby increasing Europe's foremost position in this field.
Max ERC Funding
2 150 000 €
Duration
Start date: 2015-04-01, End date: 2020-03-31
Project acronym OCULUS
Project A radical approach for improved glaucoma treatment
Researcher (PI) Peter Humphries
Host Institution (HI) THE PROVOST, FELLOWS, FOUNDATION SCHOLARS & THE OTHER MEMBERS OF BOARD OF THE COLLEGE OF THE HOLY & UNDIVIDED TRINITY OF QUEEN ELIZABETH NEAR DUBLIN
Call Details Advanced Grant (AdG), LS7, ERC-2012-ADG_20120314
Summary Open angle glaucoma (OAG) is the second leading cause of world blindness. Treatments involving topically applied pressure-reducing medications or surgery targeting ocular drainage channels are effective, although significant complications exist. We propose to address the hypothesis that it is possible to develop a radical approach to management of intraocular pressure employing an AAV-mediated system for increasing the permeability of Schlemm’s canal endothelial cells (SCEC), based on published supportive data from this laboratory showing that RNAi-mediated down regulation of mRNA encoding components of tight junctions of neuronal vascular endothelia induces increased cell permeability, a process which has been used to validate a procedure for acute treatment of neuronal edema. While tight junctions of neuronal vascular endothelial cells have been extensively studied and comprise of a series of up to 30 protein components, less is known of the organization of adherence mechanisms of SCEC, although electron- and immunofluorescence microscopy show the presence of tight junctions. We propose a comprehensive analysis of tight junction protein expression in SCEC in vitro. In vivo studies will involve introduction of AAV vectors into the anterior chamber of the eye in rodent models of elevated IOP. The vectors will be designed to express shRNAs targeting a variety of tight junction transcripts expressed in SCEC using an inducible system. The effect of RNAi-mediated increase in the permeability of SCEC will be assessed using aqueous humour outflow measurement methods and we will also explore the utility of high resolution and diffusion-weighted MRI for this purpose, which may prove to be a simpler, non-invasive and clinically relevant method. This research will provide further fundamental insights into the mechanisms of ocular pressure maintenance and could provide benefit to those patients not responsive to conventional means of therapy.
Summary
Open angle glaucoma (OAG) is the second leading cause of world blindness. Treatments involving topically applied pressure-reducing medications or surgery targeting ocular drainage channels are effective, although significant complications exist. We propose to address the hypothesis that it is possible to develop a radical approach to management of intraocular pressure employing an AAV-mediated system for increasing the permeability of Schlemm’s canal endothelial cells (SCEC), based on published supportive data from this laboratory showing that RNAi-mediated down regulation of mRNA encoding components of tight junctions of neuronal vascular endothelia induces increased cell permeability, a process which has been used to validate a procedure for acute treatment of neuronal edema. While tight junctions of neuronal vascular endothelial cells have been extensively studied and comprise of a series of up to 30 protein components, less is known of the organization of adherence mechanisms of SCEC, although electron- and immunofluorescence microscopy show the presence of tight junctions. We propose a comprehensive analysis of tight junction protein expression in SCEC in vitro. In vivo studies will involve introduction of AAV vectors into the anterior chamber of the eye in rodent models of elevated IOP. The vectors will be designed to express shRNAs targeting a variety of tight junction transcripts expressed in SCEC using an inducible system. The effect of RNAi-mediated increase in the permeability of SCEC will be assessed using aqueous humour outflow measurement methods and we will also explore the utility of high resolution and diffusion-weighted MRI for this purpose, which may prove to be a simpler, non-invasive and clinically relevant method. This research will provide further fundamental insights into the mechanisms of ocular pressure maintenance and could provide benefit to those patients not responsive to conventional means of therapy.
Max ERC Funding
2 499 838 €
Duration
Start date: 2013-04-01, End date: 2018-09-30
Project acronym OscillInterference
Project Therapeutic Mechanisms and Long Term Effects of Directed Transcranial Alternating Current Stimulation in Epileptic Seizures
Researcher (PI) Antal Berényi
Host Institution (HI) Szegedi Tudomanyegyetem - Hungarian-Netherlands School of Educational Management
Call Details Starting Grant (StG), LS5, ERC-2013-StG
Summary A significant proportion of patients with epilepsy are refractive to pharmaceutical treatments. Recurrent, untreated epileptic seizures are associated with risk of adverse neurological, cognitive, and psychological outcomes. Despite years of study, there are still significant barriers to the management of these disorders. In my proposal I advance the hypothesis that time-targeted perturbation of neural network oscillations by transcranial electric stimulation (TES) decreases the duration of seizures. I hypothesize further that spatially focused TES and chronically applied TES intervention can also permanently reduce seizure occurrence. Our specific aims are designed to perform in vivo studies in rodent models of two seizure types (absence seizures and complex partial seizures) to evaluate the effectiveness of TES in abrogating pathologic network activity, and to use high resolution recording techniques and optogenetical methods to assess the neural mechanisms involved. Our results may help to establish general principles of the diverse epilepsy pathophysiology and introduce novel therapeutic approaches. We will establish a focal TES stimulation protocol to selectively interfere with brain regions previously identified as key structures in the pathomechanism of epilepsy. The deliverables of these experiments will make a significant advancement in the understanding of the pathomechanisms of these disorders, and will offer a new alternative treatment option as a complimentary therapeutic approach to the state of the art pharmaceutical products. The methods used in this project are unique and advanced as the first attempt to perform 512 channel extracellular recordings in the behaving animal to investigate the evolution of epileptic seizures at the neuronal network and cellular levels and by achieving spatially selective TES. The combination of these methods are deployed for both understanding the mechanisms of seizure evolution, and termination of seizures.
Summary
A significant proportion of patients with epilepsy are refractive to pharmaceutical treatments. Recurrent, untreated epileptic seizures are associated with risk of adverse neurological, cognitive, and psychological outcomes. Despite years of study, there are still significant barriers to the management of these disorders. In my proposal I advance the hypothesis that time-targeted perturbation of neural network oscillations by transcranial electric stimulation (TES) decreases the duration of seizures. I hypothesize further that spatially focused TES and chronically applied TES intervention can also permanently reduce seizure occurrence. Our specific aims are designed to perform in vivo studies in rodent models of two seizure types (absence seizures and complex partial seizures) to evaluate the effectiveness of TES in abrogating pathologic network activity, and to use high resolution recording techniques and optogenetical methods to assess the neural mechanisms involved. Our results may help to establish general principles of the diverse epilepsy pathophysiology and introduce novel therapeutic approaches. We will establish a focal TES stimulation protocol to selectively interfere with brain regions previously identified as key structures in the pathomechanism of epilepsy. The deliverables of these experiments will make a significant advancement in the understanding of the pathomechanisms of these disorders, and will offer a new alternative treatment option as a complimentary therapeutic approach to the state of the art pharmaceutical products. The methods used in this project are unique and advanced as the first attempt to perform 512 channel extracellular recordings in the behaving animal to investigate the evolution of epileptic seizures at the neuronal network and cellular levels and by achieving spatially selective TES. The combination of these methods are deployed for both understanding the mechanisms of seizure evolution, and termination of seizures.
Max ERC Funding
1 419 000 €
Duration
Start date: 2013-11-01, End date: 2018-10-31
Project acronym resistance evolution
Project Bacterial evolution of hypersensitivity and resistance against antimicrobial peptides
Researcher (PI) Csaba Pal
Host Institution (HI) MAGYAR TUDOMANYOS AKADEMIA SZEGEDIBIOLOGIAI KUTATOKOZPONT
Call Details Consolidator Grant (CoG), LS8, ERC-2014-CoG
Summary Evolution of resistance towards a single drug simultaneously increases (cross-resistance) or decreases (collateral sensitivity) fitness to multiple other antimicrobial agents. The molecular mechanisms driving cross-resistance are relatively well described, but it remains largely unclear how frequently does genetic adaptation to a single drug increase the sensitivity to others and what the underlying molecular mechanisms of collateral sensitivity are. This proposal focuses on studying the bacterial evolution of resistance and collateral sensitivity against antimicrobial peptides (AMPs). Beyond their modulatory roles in the immune system, these naturally occurring peptides provide protection against pathogenic microbes, and are considered as promising novel alternatives to traditional antibiotics. However, there are concerns that evolution against therapeutic AMPs can readily develop and as a by-product this might compromise natural immunity. Our knowledge of these issues is limited due to the shortage of systematic evolutionary studies. Therefore, the three central questions we address are: Do bacteria resistant to multiple antibiotics become hypersensitive to certain antimicrobial peptides? What are the evolutionary mechanisms leading to AMP resistance and to what extent does this process induce cross-resistance/collateral sensitivity against other drugs? Last, are these evolutionary trade-offs predictable based on chemical and functional peptide properties? To investigate these issues rigorously, we integrate tools of laboratory evolution, high-throughput phenotypic assays, functional genomics, and computational systems biology. Our project will provide an insight into the evolutionary mechanisms that drive cross-resistance and collateral sensitivities with the aim to explore the vulnerable points of resistant bacteria. Another goal is to provide guidelines for the future design of antimicrobial peptides with desirable properties against bacterial pathogens.
Summary
Evolution of resistance towards a single drug simultaneously increases (cross-resistance) or decreases (collateral sensitivity) fitness to multiple other antimicrobial agents. The molecular mechanisms driving cross-resistance are relatively well described, but it remains largely unclear how frequently does genetic adaptation to a single drug increase the sensitivity to others and what the underlying molecular mechanisms of collateral sensitivity are. This proposal focuses on studying the bacterial evolution of resistance and collateral sensitivity against antimicrobial peptides (AMPs). Beyond their modulatory roles in the immune system, these naturally occurring peptides provide protection against pathogenic microbes, and are considered as promising novel alternatives to traditional antibiotics. However, there are concerns that evolution against therapeutic AMPs can readily develop and as a by-product this might compromise natural immunity. Our knowledge of these issues is limited due to the shortage of systematic evolutionary studies. Therefore, the three central questions we address are: Do bacteria resistant to multiple antibiotics become hypersensitive to certain antimicrobial peptides? What are the evolutionary mechanisms leading to AMP resistance and to what extent does this process induce cross-resistance/collateral sensitivity against other drugs? Last, are these evolutionary trade-offs predictable based on chemical and functional peptide properties? To investigate these issues rigorously, we integrate tools of laboratory evolution, high-throughput phenotypic assays, functional genomics, and computational systems biology. Our project will provide an insight into the evolutionary mechanisms that drive cross-resistance and collateral sensitivities with the aim to explore the vulnerable points of resistant bacteria. Another goal is to provide guidelines for the future design of antimicrobial peptides with desirable properties against bacterial pathogens.
Max ERC Funding
1 846 250 €
Duration
Start date: 2015-10-01, End date: 2021-09-30
Project acronym RLPHARMFMRI
Project Beyond dopamine: Characterizing the computational functions of midbrain modulatory neurotransmitter systems in human reinforcement learning using model-based pharmacological fMRI
Researcher (PI) John O'doherty
Host Institution (HI) THE PROVOST, FELLOWS, FOUNDATION SCHOLARS & THE OTHER MEMBERS OF BOARD OF THE COLLEGE OF THE HOLY & UNDIVIDED TRINITY OF QUEEN ELIZABETH NEAR DUBLIN
Call Details Starting Grant (StG), LS5, ERC-2009-StG
Summary Understanding how humans and other animals are able to learn from experience and use this information to select future behavioural strategies to obtain the reinforcers necessary for survival, is a fundamental research question in biology. Considerable progress has been made in recent years on the neural computational underpinnings of this process following the observation that the phasic activity of dopamine neurons in the midbrain resembles a prediction error from a formal computational theory known as reinforcement learning (RL). While much is known about the functions of dopamine in RL, much less is known about the computational functions of other modulatory neurotransmitter systems in the midbrain such as the cholinergic, norcpinephrine, and serotonergic systems. The goal of this research proposal to the ERC, is to begin a systematic study of the computational functions of these other neurotransmitter systems (beyond dopamine) in RL. To do this we will combine functional magnetic resonance imaging in human subjects while they perform simple decision making tasks and undergo pharmacological manipulations to modulate systemic levels of these different neurotransmitter systems. We will combine computational model-based analyses with fMRI and behavioural data in order to explore the effects that these pharmacological modulations exert on different parameters and modules within RL. Specifically, we will test the contributions that the cholinergic system makes in setting the learning rate during RL and in mediating computations of expected uncertainty in the distribution of rewards available, we will test for the role of norepinephrine in balancing the rate of exploration and exploitation during decision making, as well as in encoding the level of unexpected uncertainty, and we will explore the possible role of serotonin in setting the rate of temporal discounting for reward, or in encoding prediction errors during aversive as opposed to reward-learning.
Summary
Understanding how humans and other animals are able to learn from experience and use this information to select future behavioural strategies to obtain the reinforcers necessary for survival, is a fundamental research question in biology. Considerable progress has been made in recent years on the neural computational underpinnings of this process following the observation that the phasic activity of dopamine neurons in the midbrain resembles a prediction error from a formal computational theory known as reinforcement learning (RL). While much is known about the functions of dopamine in RL, much less is known about the computational functions of other modulatory neurotransmitter systems in the midbrain such as the cholinergic, norcpinephrine, and serotonergic systems. The goal of this research proposal to the ERC, is to begin a systematic study of the computational functions of these other neurotransmitter systems (beyond dopamine) in RL. To do this we will combine functional magnetic resonance imaging in human subjects while they perform simple decision making tasks and undergo pharmacological manipulations to modulate systemic levels of these different neurotransmitter systems. We will combine computational model-based analyses with fMRI and behavioural data in order to explore the effects that these pharmacological modulations exert on different parameters and modules within RL. Specifically, we will test the contributions that the cholinergic system makes in setting the learning rate during RL and in mediating computations of expected uncertainty in the distribution of rewards available, we will test for the role of norepinephrine in balancing the rate of exploration and exploitation during decision making, as well as in encoding the level of unexpected uncertainty, and we will explore the possible role of serotonin in setting the rate of temporal discounting for reward, or in encoding prediction errors during aversive as opposed to reward-learning.
Max ERC Funding
1 841 404 €
Duration
Start date: 2010-01-01, End date: 2010-09-30
Project acronym SERRACO
Project Modulation of cortical activity by median raphe neuronal assemblies with identified behavioural effects
Researcher (PI) Tamás Freund
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Advanced Grant (AdG), LS5, ERC-2011-ADG_20110310
Summary Cortical operations are built up from states associated with distinct behaviour-dependent network activity patterns that subserve information aquisition, encoding, memory consolidation and retrieval. Thus, they can be considered as manifestations of different processing modes. Groups of modulatory, largely monoaminergic neurons located in subcortical nuclei innervating all forebrain areas are indispensable for the generation, stabilization and termination of cortical activity states. In recent years the concept of subcortical modulation has been expanded by the discovery of a fast type of modulatory action driving the rapid readjustment of cortical activity and associated behaviours. Thus, cortical networks are under the influence of a tonic, slow, as well as a phasic, rapid component of subcortical modulation that are acting in parallel. Results from our laboratory revealed that the median raphe (MR) nucleus, one of the main sources of serotonergic innervation of the limbic system , besides the non-synaptic diffuse action, also exerts a fast type of modulation via the selective innervation of cortical GABAergic interneurons. This selective effect on local inhibition may be ideal for the synchronous resetting of the target principal cell circuits, or for the continuous tuning of their activity. These discoveries, together with the methodological advances of recent years, enable us to map the neuronal network mechanisms behind transitions of brain states, as well as associated behaviours, induced by subcortical inputs. We will focus on the MR – limbic connection with the aim to unravel the physiological, pharmacological and anatomical features of MR neuronal assemblies, both the slow- and fast-acting, as well as the serotonergic and glutamatergic components (together with their cortical target circuits) that will have been shown - using optic stimulation of ChR2/eGFP virus-infected MR neurons - to evoke characteristic behaviours, such as anxiety and conditioned fear.
Summary
Cortical operations are built up from states associated with distinct behaviour-dependent network activity patterns that subserve information aquisition, encoding, memory consolidation and retrieval. Thus, they can be considered as manifestations of different processing modes. Groups of modulatory, largely monoaminergic neurons located in subcortical nuclei innervating all forebrain areas are indispensable for the generation, stabilization and termination of cortical activity states. In recent years the concept of subcortical modulation has been expanded by the discovery of a fast type of modulatory action driving the rapid readjustment of cortical activity and associated behaviours. Thus, cortical networks are under the influence of a tonic, slow, as well as a phasic, rapid component of subcortical modulation that are acting in parallel. Results from our laboratory revealed that the median raphe (MR) nucleus, one of the main sources of serotonergic innervation of the limbic system , besides the non-synaptic diffuse action, also exerts a fast type of modulation via the selective innervation of cortical GABAergic interneurons. This selective effect on local inhibition may be ideal for the synchronous resetting of the target principal cell circuits, or for the continuous tuning of their activity. These discoveries, together with the methodological advances of recent years, enable us to map the neuronal network mechanisms behind transitions of brain states, as well as associated behaviours, induced by subcortical inputs. We will focus on the MR – limbic connection with the aim to unravel the physiological, pharmacological and anatomical features of MR neuronal assemblies, both the slow- and fast-acting, as well as the serotonergic and glutamatergic components (together with their cortical target circuits) that will have been shown - using optic stimulation of ChR2/eGFP virus-infected MR neurons - to evoke characteristic behaviours, such as anxiety and conditioned fear.
Max ERC Funding
2 700 000 €
Duration
Start date: 2012-03-01, End date: 2017-02-28
Project acronym SYM-BIOTICS
Project Dual exploitation of natural plant strategies in agriculture and public health: enhancing nitrogen-fixation and surmounting microbial infections
Researcher (PI) Eva Kondorosi
Host Institution (HI) MAGYAR TUDOMANYOS AKADEMIA SZEGEDIBIOLOGIAI KUTATOKOZPONT
Call Details Advanced Grant (AdG), LS9, ERC-2010-AdG_20100317
Summary With an unprecedented increase in the human population, higher agricultural production, enhanced food safety and the protection against alarming rise of antibiotic resistant pathogenic bacteria are amongst the main challenges of this century. This proposal centered on Rhizobium-legume symbiosis aims at contributing to these tasks by i) understanding the development of symbiotic nitrogen fixing cells for improvement of the eco-friendly biological nitrogen fixation, ii) gaining a comprehensive knowledge on polyploidy having a great impact on crop yields and iii) exploiting the strategies of symbiotic plant cells for the development of novel antibiotics. Symbiotic nitrogen fixation in Rhizobium-legume interactions is a major contributor to the combined nitrogen pool in the biosphere. It takes place in root nodules where giant plant cells host the nitrogen fixing bacteria. In Medicago nodules both the plant cells and the bacteria are polyploids and incapable for cell division. These polyploid plant cells produce hundreds of symbiotic peptides (symPEPs) that provoke terminal differentiation of bacteria in symbiosis and exhibit broad range antimicrobial activities in vitro. Permanent generation of polyploid cells is essential for the nodule development. It will be studied whether the complete genome is duplicated in consecutive endocycles, how different ploidy levels affect DNA methylation and expression profile and whether polyploidy is required for the expression of symPEP genes. The activity and mode of actions of symPEPs are in the focus of the proposal; i) how symPEPs achieve bacteroid differentiation and affect nitrogen fixation and ii) whether symPEP antimicrobial activities provide novel modes of antimicrobial actions and iii) whether ¿Sym-Biotics¿ could become widely used novel antibiotics. Their applicability as plant protecting and meat decontaminating agents as well as their in vivo efficiency in mouse septicemia models will be tested.
Summary
With an unprecedented increase in the human population, higher agricultural production, enhanced food safety and the protection against alarming rise of antibiotic resistant pathogenic bacteria are amongst the main challenges of this century. This proposal centered on Rhizobium-legume symbiosis aims at contributing to these tasks by i) understanding the development of symbiotic nitrogen fixing cells for improvement of the eco-friendly biological nitrogen fixation, ii) gaining a comprehensive knowledge on polyploidy having a great impact on crop yields and iii) exploiting the strategies of symbiotic plant cells for the development of novel antibiotics. Symbiotic nitrogen fixation in Rhizobium-legume interactions is a major contributor to the combined nitrogen pool in the biosphere. It takes place in root nodules where giant plant cells host the nitrogen fixing bacteria. In Medicago nodules both the plant cells and the bacteria are polyploids and incapable for cell division. These polyploid plant cells produce hundreds of symbiotic peptides (symPEPs) that provoke terminal differentiation of bacteria in symbiosis and exhibit broad range antimicrobial activities in vitro. Permanent generation of polyploid cells is essential for the nodule development. It will be studied whether the complete genome is duplicated in consecutive endocycles, how different ploidy levels affect DNA methylation and expression profile and whether polyploidy is required for the expression of symPEP genes. The activity and mode of actions of symPEPs are in the focus of the proposal; i) how symPEPs achieve bacteroid differentiation and affect nitrogen fixation and ii) whether symPEP antimicrobial activities provide novel modes of antimicrobial actions and iii) whether ¿Sym-Biotics¿ could become widely used novel antibiotics. Their applicability as plant protecting and meat decontaminating agents as well as their in vivo efficiency in mouse septicemia models will be tested.
Max ERC Funding
2 320 000 €
Duration
Start date: 2011-07-01, End date: 2017-06-30
Project acronym VISONby3DSTIM
Project Restoration of visual perception by artificial stimulation performed by 3D EAO microscopy
Researcher (PI) Jozsef Balázs Rózsa
Host Institution (HI) INSTITUTE OF EXPERIMENTAL MEDICINE - HUNGARIAN ACADEMY OF SCIENCES
Call Details Consolidator Grant (CoG), LS5, ERC-2015-CoG
Summary The long-term aim of the investigation is to assess the feasibility of creating an “artificial sense” and, thereby, a possible sensory (visual) prosthetic. While working towards this goal, we will have to address the question of how neural assembly activity relates to subjective perceptions. Finding and understanding these functional assemblies will make it possible to reactivate them in a precise, biologically relevant manner to elicit similar cortical activation as visual stimulation. Recent publications suggest that cortical connectivity can be mapped by two-photon microscopy. Here we want, therefore, to develop a novel 3D Electro-Acousto-Optical microscope for high-throughput assembly mapping. The microscope will be capable of scanning neuronal activity with one order of magnitude higher speed (300-500 kHz/ROI) and simultaneously photoactivate neurons with three order of magnitude higher efficiency (2,500 – 25,000 neurons/ms) than existing 3D microscopes while preserving the subcellular resolution required to simultaneously measure the somatic, the dendritic and axonal computation units in the entire V1 region of the cortex. The microscope will be based on our current 3D AO technology; on novel ultra-fast scanning technologies; new, 10-fold faster AO deflectors; and novel (multi-ROI) scanning strategies. Using our microscope in combination with novel caged neurotransmitters and optogenetic tools, we want to map cell assemblies and to understand how they form larger clusters and how they are associated with visual features. Furthermore, as a proof-of-concept of this grant, we want to restore visual perception by recreating previously mapped assembly patterns with 3D artificial photositmulation in behaving mice and see if the animal responds to the artificial stimulus in the same way as to the visual stimulus. Moreover, we want to restore visual information based spatial navigation in head restrained animals orienting and moving in a virtual labyrinth for reward.
Summary
The long-term aim of the investigation is to assess the feasibility of creating an “artificial sense” and, thereby, a possible sensory (visual) prosthetic. While working towards this goal, we will have to address the question of how neural assembly activity relates to subjective perceptions. Finding and understanding these functional assemblies will make it possible to reactivate them in a precise, biologically relevant manner to elicit similar cortical activation as visual stimulation. Recent publications suggest that cortical connectivity can be mapped by two-photon microscopy. Here we want, therefore, to develop a novel 3D Electro-Acousto-Optical microscope for high-throughput assembly mapping. The microscope will be capable of scanning neuronal activity with one order of magnitude higher speed (300-500 kHz/ROI) and simultaneously photoactivate neurons with three order of magnitude higher efficiency (2,500 – 25,000 neurons/ms) than existing 3D microscopes while preserving the subcellular resolution required to simultaneously measure the somatic, the dendritic and axonal computation units in the entire V1 region of the cortex. The microscope will be based on our current 3D AO technology; on novel ultra-fast scanning technologies; new, 10-fold faster AO deflectors; and novel (multi-ROI) scanning strategies. Using our microscope in combination with novel caged neurotransmitters and optogenetic tools, we want to map cell assemblies and to understand how they form larger clusters and how they are associated with visual features. Furthermore, as a proof-of-concept of this grant, we want to restore visual perception by recreating previously mapped assembly patterns with 3D artificial photositmulation in behaving mice and see if the animal responds to the artificial stimulus in the same way as to the visual stimulus. Moreover, we want to restore visual information based spatial navigation in head restrained animals orienting and moving in a virtual labyrinth for reward.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-05-01, End date: 2021-04-30