Project acronym 2DNANOCAPS
Project Next Generation of 2D-Nanomaterials: Enabling Supercapacitor Development
Researcher (PI) Valeria Nicolosi
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), PE8, ERC-2011-StG_20101014
Summary Climate change and the decreasing availability of fossil fuels require society to move towards sustainable and renewable resources. 2DNanoCaps will focus on electrochemical energy storage, specifically supercapacitors. In terms of performance supercapacitors fill up the gap between batteries and the classical capacitors. Whereas batteries possess a high energy density but low power density, supercapacitors possess high power density but low energy density. Efforts are currently dedicated to move supercapacitors towards high energy density and high power density performance. Improvements have been achieved in the last few years due to the use of new electrode nanomaterials and the design of new hybrid faradic/capacitive systems. We recognize, however, that we are reaching a newer limit beyond which we will only see small incremental improvements. The main reason for this being the intrinsic difficulty in handling and processing materials at the nano-scale and the lack of communication across different scientific disciplines. I plan to use a multidisciplinary approach, where novel nanomaterials, existing knowledge on nano-scale processing and established expertise in device fabrication and testing will be brought together to focus on creating more efficient supercapacitor technologies. 2DNanoCaps will exploit liquid phase exfoliated two-dimensional nanomaterials such as transition metal oxides, layered metal chalcogenides and graphene as electrode materials. Electrodes will be ultra-thin (capacitance and thickness of the electrodes are inversely proportional), conductive, with high dielectric constants. Intercalation of ions between the assembled 2D flakes will be also achievable, providing pseudo-capacitance. The research here proposed will be initially based on fundamental laboratory studies, recognising that this holds the key to achieving step-change in supercapacitors, but also includes scaling-up and hybridisation as final objectives.
Summary
Climate change and the decreasing availability of fossil fuels require society to move towards sustainable and renewable resources. 2DNanoCaps will focus on electrochemical energy storage, specifically supercapacitors. In terms of performance supercapacitors fill up the gap between batteries and the classical capacitors. Whereas batteries possess a high energy density but low power density, supercapacitors possess high power density but low energy density. Efforts are currently dedicated to move supercapacitors towards high energy density and high power density performance. Improvements have been achieved in the last few years due to the use of new electrode nanomaterials and the design of new hybrid faradic/capacitive systems. We recognize, however, that we are reaching a newer limit beyond which we will only see small incremental improvements. The main reason for this being the intrinsic difficulty in handling and processing materials at the nano-scale and the lack of communication across different scientific disciplines. I plan to use a multidisciplinary approach, where novel nanomaterials, existing knowledge on nano-scale processing and established expertise in device fabrication and testing will be brought together to focus on creating more efficient supercapacitor technologies. 2DNanoCaps will exploit liquid phase exfoliated two-dimensional nanomaterials such as transition metal oxides, layered metal chalcogenides and graphene as electrode materials. Electrodes will be ultra-thin (capacitance and thickness of the electrodes are inversely proportional), conductive, with high dielectric constants. Intercalation of ions between the assembled 2D flakes will be also achievable, providing pseudo-capacitance. The research here proposed will be initially based on fundamental laboratory studies, recognising that this holds the key to achieving step-change in supercapacitors, but also includes scaling-up and hybridisation as final objectives.
Max ERC Funding
1 501 296 €
Duration
Start date: 2011-10-01, End date: 2016-09-30
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 3D2DPrint
Project 3D Printing of Novel 2D Nanomaterials: Adding Advanced 2D Functionalities to Revolutionary Tailored 3D Manufacturing
Researcher (PI) Valeria Nicolosi
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), PE8, ERC-2015-CoG
Summary My vision is to establish, within the framework of an ERC CoG, a multidisciplinary group which will work in concert towards pioneering the integration of novel 2-Dimensional nanomaterials with novel additive fabrication techniques to develop a unique class of energy storage devices.
Batteries and supercapacitors are two very complementary types of energy storage devices. Batteries store much higher energy densities; supercapacitors, on the other hand, hold one tenth of the electricity per unit of volume or weight as compared to batteries but can achieve much higher power densities. Technology is currently striving to improve the power density of batteries and the energy density of supercapacitors. To do so it is imperative to develop new materials, chemistries and manufacturing strategies.
3D2DPrint aims to develop micro-energy devices (both supercapacitors and batteries), technologies particularly relevant in the context of the emergent industry of micro-electro-mechanical systems and constantly downsized electronics. We plan to use novel two-dimensional (2D) nanomaterials obtained by liquid-phase exfoliation. This method offers a new, economic and easy way to prepare ink of a variety of 2D systems, allowing to produce wide device performance window through elegant and simple constituent control at the point of fabrication. 3D2DPrint will use our expertise and know-how to allow development of advanced AM methods to integrate dissimilar nanomaterial blends and/or “hybrids” into fully embedded 3D printed energy storage devices, with the ultimate objective to realise a range of products that contain the above described nanomaterials subcomponent devices, electrical connections and traditional micro-fabricated subcomponents (if needed) ideally using a single tool.
Summary
My vision is to establish, within the framework of an ERC CoG, a multidisciplinary group which will work in concert towards pioneering the integration of novel 2-Dimensional nanomaterials with novel additive fabrication techniques to develop a unique class of energy storage devices.
Batteries and supercapacitors are two very complementary types of energy storage devices. Batteries store much higher energy densities; supercapacitors, on the other hand, hold one tenth of the electricity per unit of volume or weight as compared to batteries but can achieve much higher power densities. Technology is currently striving to improve the power density of batteries and the energy density of supercapacitors. To do so it is imperative to develop new materials, chemistries and manufacturing strategies.
3D2DPrint aims to develop micro-energy devices (both supercapacitors and batteries), technologies particularly relevant in the context of the emergent industry of micro-electro-mechanical systems and constantly downsized electronics. We plan to use novel two-dimensional (2D) nanomaterials obtained by liquid-phase exfoliation. This method offers a new, economic and easy way to prepare ink of a variety of 2D systems, allowing to produce wide device performance window through elegant and simple constituent control at the point of fabrication. 3D2DPrint will use our expertise and know-how to allow development of advanced AM methods to integrate dissimilar nanomaterial blends and/or “hybrids” into fully embedded 3D printed energy storage devices, with the ultimate objective to realise a range of products that contain the above described nanomaterials subcomponent devices, electrical connections and traditional micro-fabricated subcomponents (if needed) ideally using a single tool.
Max ERC Funding
2 499 942 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym A-BINGOS
Project Accreting binary populations in Nearby Galaxies: Observations and Simulations
Researcher (PI) Andreas Zezas
Host Institution (HI) IDRYMA TECHNOLOGIAS KAI EREVNAS
Call Details Consolidator Grant (CoG), PE9, ERC-2013-CoG
Summary "High-energy observations of our Galaxy offer a good, albeit not complete, picture of the X-ray source populations, in particular the accreting binary sources. Recent ability to study accreting binaries in nearby galaxies has shown that we would be short-sighted if we restricted ourselves to our Galaxy or to a few nearby ones. I propose an ambitious project that involves a comprehensive study of all the galaxies within 10 Mpc for which we can study in detail their X-ray sources and stellar populations. The study will combine data from a unique suite of observatories (Chandra, XMM-Newton, HST, Spitzer) with state-of-the-art theoretical modelling of binary systems. I propose a novel approach that links the accreting binary populations to their parent stellar populations and surpasses any current studies of X-ray binary populations, both in scale and in scope, by: (a) combining methods and results from several different areas of astrophysics (compact objects, binary systems, stellar populations, galaxy evolution); (b) using data from almost the whole electromagnetic spectrum (infrared to X-ray bands); (c) identifying and studying the different sub-populations of accreting binaries; and (d) performing direct comparison between observations and theoretical predictions, over a broad parameter space. The project: (a) will answer the long-standing question of the formation efficiency of accreting binaries in different environments; and (b) will constrain their evolutionary paths. As by-products the project will provide eagerly awaited input to the fields of gravitational-wave sources, γ-ray bursts, and X-ray emitting galaxies at cosmological distances and it will produce a heritage multi-wavelength dataset and library of models for future studies of galaxies and accreting binaries."
Summary
"High-energy observations of our Galaxy offer a good, albeit not complete, picture of the X-ray source populations, in particular the accreting binary sources. Recent ability to study accreting binaries in nearby galaxies has shown that we would be short-sighted if we restricted ourselves to our Galaxy or to a few nearby ones. I propose an ambitious project that involves a comprehensive study of all the galaxies within 10 Mpc for which we can study in detail their X-ray sources and stellar populations. The study will combine data from a unique suite of observatories (Chandra, XMM-Newton, HST, Spitzer) with state-of-the-art theoretical modelling of binary systems. I propose a novel approach that links the accreting binary populations to their parent stellar populations and surpasses any current studies of X-ray binary populations, both in scale and in scope, by: (a) combining methods and results from several different areas of astrophysics (compact objects, binary systems, stellar populations, galaxy evolution); (b) using data from almost the whole electromagnetic spectrum (infrared to X-ray bands); (c) identifying and studying the different sub-populations of accreting binaries; and (d) performing direct comparison between observations and theoretical predictions, over a broad parameter space. The project: (a) will answer the long-standing question of the formation efficiency of accreting binaries in different environments; and (b) will constrain their evolutionary paths. As by-products the project will provide eagerly awaited input to the fields of gravitational-wave sources, γ-ray bursts, and X-ray emitting galaxies at cosmological distances and it will produce a heritage multi-wavelength dataset and library of models for future studies of galaxies and accreting binaries."
Max ERC Funding
1 242 000 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
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 Active-DNA
Project Computationally Active DNA Nanostructures
Researcher (PI) Damien WOODS
Host Institution (HI) NATIONAL UNIVERSITY OF IRELAND MAYNOOTH
Call Details Consolidator Grant (CoG), PE6, ERC-2017-COG
Summary During the 20th century computer technology evolved from bulky, slow, special purpose mechanical engines to the now ubiquitous silicon chips and software that are one of the pinnacles of human ingenuity. The goal of the field of molecular programming is to take the next leap and build a new generation of matter-based computers using DNA, RNA and proteins. This will be accomplished by computer scientists, physicists and chemists designing molecules to execute ``wet'' nanoscale programs in test tubes. The workflow includes proposing theoretical models, mathematically proving their computational properties, physical modelling and implementation in the wet-lab.
The past decade has seen remarkable progress at building static 2D and 3D DNA nanostructures. However, unlike biological macromolecules and complexes that are built via specified self-assembly pathways, that execute robotic-like movements, and that undergo evolution, the activity of human-engineered nanostructures is severely limited. We will need sophisticated algorithmic ideas to build structures that rival active living systems. Active-DNA, aims to address this challenge by achieving a number of objectives on computation, DNA-based self-assembly and molecular robotics. Active-DNA research work will range from defining models and proving theorems that characterise the computational and expressive capabilities of such active programmable materials to experimental work implementing active DNA nanostructures in the wet-lab.
Summary
During the 20th century computer technology evolved from bulky, slow, special purpose mechanical engines to the now ubiquitous silicon chips and software that are one of the pinnacles of human ingenuity. The goal of the field of molecular programming is to take the next leap and build a new generation of matter-based computers using DNA, RNA and proteins. This will be accomplished by computer scientists, physicists and chemists designing molecules to execute ``wet'' nanoscale programs in test tubes. The workflow includes proposing theoretical models, mathematically proving their computational properties, physical modelling and implementation in the wet-lab.
The past decade has seen remarkable progress at building static 2D and 3D DNA nanostructures. However, unlike biological macromolecules and complexes that are built via specified self-assembly pathways, that execute robotic-like movements, and that undergo evolution, the activity of human-engineered nanostructures is severely limited. We will need sophisticated algorithmic ideas to build structures that rival active living systems. Active-DNA, aims to address this challenge by achieving a number of objectives on computation, DNA-based self-assembly and molecular robotics. Active-DNA research work will range from defining models and proving theorems that characterise the computational and expressive capabilities of such active programmable materials to experimental work implementing active DNA nanostructures in the wet-lab.
Max ERC Funding
2 349 603 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym ADAPTIVES
Project Algorithmic Development and Analysis of Pioneer Techniques for Imaging with waVES
Researcher (PI) Chrysoula Tsogka
Host Institution (HI) IDRYMA TECHNOLOGIAS KAI EREVNAS
Call Details Starting Grant (StG), PE1, ERC-2009-StG
Summary The proposed work concerns the theoretical and numerical development of robust and adaptive methodologies for broadband imaging in clutter. The word clutter expresses our uncertainty on the wave speed of the propagation medium. Our results are expected to have a strong impact in a wide range of applications, including underwater acoustics, exploration geophysics and ultrasound non-destructive testing. Our machinery is coherent interferometry (CINT), a state-of-the-art statistically stable imaging methodology, highly suitable for the development of imaging methods in clutter. We aim to extend CINT along two complementary directions: novel types of applications, and further mathematical and numerical development so as to assess and extend its range of applicability. CINT is designed for imaging with partially coherent array data recorded in richly scattering media. It uses statistical smoothing techniques to obtain results that are independent of the clutter realization. Quantifying the amount of smoothing needed is difficult, especially when there is no a priori knowledge about the propagation medium. We intend to address this question by coupling the imaging process with the estimation of the medium's large scale features. Our algorithms rely on the residual coherence in the data. When the coherent signal is too weak, the CINT results are unsatisfactory. We propose two ways for enhancing the resolution of CINT: filter the data prior to imaging (noise reduction) and waveform design (optimize the source distribution). Finally, we propose to extend the applicability of our imaging-in-clutter methodologies by investigating the possibility of utilizing ambient noise sources to perform passive sensor imaging, as well as by studying the imaging problem in random waveguides.
Summary
The proposed work concerns the theoretical and numerical development of robust and adaptive methodologies for broadband imaging in clutter. The word clutter expresses our uncertainty on the wave speed of the propagation medium. Our results are expected to have a strong impact in a wide range of applications, including underwater acoustics, exploration geophysics and ultrasound non-destructive testing. Our machinery is coherent interferometry (CINT), a state-of-the-art statistically stable imaging methodology, highly suitable for the development of imaging methods in clutter. We aim to extend CINT along two complementary directions: novel types of applications, and further mathematical and numerical development so as to assess and extend its range of applicability. CINT is designed for imaging with partially coherent array data recorded in richly scattering media. It uses statistical smoothing techniques to obtain results that are independent of the clutter realization. Quantifying the amount of smoothing needed is difficult, especially when there is no a priori knowledge about the propagation medium. We intend to address this question by coupling the imaging process with the estimation of the medium's large scale features. Our algorithms rely on the residual coherence in the data. When the coherent signal is too weak, the CINT results are unsatisfactory. We propose two ways for enhancing the resolution of CINT: filter the data prior to imaging (noise reduction) and waveform design (optimize the source distribution). Finally, we propose to extend the applicability of our imaging-in-clutter methodologies by investigating the possibility of utilizing ambient noise sources to perform passive sensor imaging, as well as by studying the imaging problem in random waveguides.
Max ERC Funding
690 000 €
Duration
Start date: 2010-06-01, End date: 2015-11-30
Project acronym AFFIRM
Project Analysis of Biofilm Mediated Fouling of Nanofiltration Membranes
Researcher (PI) Eoin Casey
Host Institution (HI) UNIVERSITY COLLEGE DUBLIN, NATIONAL UNIVERSITY OF IRELAND, DUBLIN
Call Details Starting Grant (StG), PE8, ERC-2011-StG_20101014
Summary 1.2 billion people worldwide lack access to safe drinking water. Drinking water quality is threatened by newly emerging organic micro-pollutants (pesticides, pharmaceuticals, industrial chemicals) in source waters. Nanofiltration is a technology that is expected to play a key role in future water treatment processes due to its effectiveness in removal of micropollutants. However, the loss of membrane flux due to fouling is one of the main impediments in the development of membrane processes for use in drinking water treatment. Currently there is a wholly inadequate mechanistic understanding of the role of biofilm on the fouling of nanofiltration membranes.
Applying techniques including confocal microscopy, force spectroscopy, and infrared spectroscopy using an experimental programme informed by a technique known as scale-down together with mathematical modelling, it is confidently expected that significant advances will be gained in the mechanistic understanding of nanofiltration biofouling.
The specific objectives are 1. How is the rate of formation and extent of such biofilms influenced by the biological response to the local microenvironment? 2 Elucidate the effect of extracellular polysaccharide substances on physical properties, composition and structure of these biofilms. 3: Investigate mechanisms to enhance biofilm removal by a physical detachment process complemented by techniques that alter biofilm material properties.
A more fundamental insight into the mechanisms of nanofiltration operation will help in further development of this treatment method in future water treatment processes.
Summary
1.2 billion people worldwide lack access to safe drinking water. Drinking water quality is threatened by newly emerging organic micro-pollutants (pesticides, pharmaceuticals, industrial chemicals) in source waters. Nanofiltration is a technology that is expected to play a key role in future water treatment processes due to its effectiveness in removal of micropollutants. However, the loss of membrane flux due to fouling is one of the main impediments in the development of membrane processes for use in drinking water treatment. Currently there is a wholly inadequate mechanistic understanding of the role of biofilm on the fouling of nanofiltration membranes.
Applying techniques including confocal microscopy, force spectroscopy, and infrared spectroscopy using an experimental programme informed by a technique known as scale-down together with mathematical modelling, it is confidently expected that significant advances will be gained in the mechanistic understanding of nanofiltration biofouling.
The specific objectives are 1. How is the rate of formation and extent of such biofilms influenced by the biological response to the local microenvironment? 2 Elucidate the effect of extracellular polysaccharide substances on physical properties, composition and structure of these biofilms. 3: Investigate mechanisms to enhance biofilm removal by a physical detachment process complemented by techniques that alter biofilm material properties.
A more fundamental insight into the mechanisms of nanofiltration operation will help in further development of this treatment method in future water treatment processes.
Max ERC Funding
1 468 987 €
Duration
Start date: 2011-10-01, End date: 2016-09-30
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 ARMOS
Project Advanced multifunctional Reactors for green Mobility and Solar fuels
Researcher (PI) Athanasios Konstandopoulos
Host Institution (HI) ETHNIKO KENTRO EREVNAS KAI TECHNOLOGIKIS ANAPTYXIS
Call Details Advanced Grant (AdG), PE8, ERC-2010-AdG_20100224
Summary Green Mobility requires an integrated approach to the chain fuel/engine/emissions. The present project aims at ground breaking advances in the area of Green Mobility by (a) enabling the production of affordable, carbon-neutral, clean, solar fuels using exclusively renewable/recyclable raw materials, namely solar energy, water and captured Carbon Dioxide from combustion power plants (b) developing a highly compact, multifunctional reactor, able to eliminate gaseous and particulate emissions from the exhaust of engines operated on such clean fuels.
The overall research approach will be based on material science, engineering and simulation technology developed by the PI over the past 20 years in the area of Diesel Emission Control Reactors, which will be further extended and cross-fertilized in the area of Solar Thermochemical Reactors, an emerging discipline of high importance for sustainable development, where the PI’s research group has already made significant contributions, and received the 2006 European Commission’s Descartes Prize for the development of the first ever solar reactor, holding the potential to produce on a large scale, pure renewable Hydrogen from the thermochemical splitting of water, also known as the HYDROSOL technology.
Summary
Green Mobility requires an integrated approach to the chain fuel/engine/emissions. The present project aims at ground breaking advances in the area of Green Mobility by (a) enabling the production of affordable, carbon-neutral, clean, solar fuels using exclusively renewable/recyclable raw materials, namely solar energy, water and captured Carbon Dioxide from combustion power plants (b) developing a highly compact, multifunctional reactor, able to eliminate gaseous and particulate emissions from the exhaust of engines operated on such clean fuels.
The overall research approach will be based on material science, engineering and simulation technology developed by the PI over the past 20 years in the area of Diesel Emission Control Reactors, which will be further extended and cross-fertilized in the area of Solar Thermochemical Reactors, an emerging discipline of high importance for sustainable development, where the PI’s research group has already made significant contributions, and received the 2006 European Commission’s Descartes Prize for the development of the first ever solar reactor, holding the potential to produce on a large scale, pure renewable Hydrogen from the thermochemical splitting of water, also known as the HYDROSOL technology.
Max ERC Funding
1 750 000 €
Duration
Start date: 2011-02-01, End date: 2017-01-31
Project acronym ASSESS
Project Episodic Mass Loss in the Most Massive Stars: Key to Understanding the Explosive Early Universe
Researcher (PI) Alceste BONANOS
Host Institution (HI) NATIONAL OBSERVATORY OF ATHENS
Call Details Consolidator Grant (CoG), PE9, ERC-2017-COG
Summary Massive stars dominate their surroundings during their short lifetimes, while their explosive deaths impact the chemical evolution and spatial cohesion of their hosts. After birth, their evolution is largely dictated by their ability to remove layers of hydrogen from their envelopes. Multiple lines of evidence are pointing to violent, episodic mass-loss events being responsible for removing a large part of the massive stellar envelope, especially in low-metallicity galaxies. Episodic mass loss, however, is not understood theoretically, neither accounted for in state-of-the-art models of stellar evolution, which has far-reaching consequences for many areas of astronomy. We aim to determine whether episodic mass loss is a dominant process in the evolution of the most massive stars by conducting the first extensive, multi-wavelength survey of evolved massive stars in the nearby Universe. The project hinges on the fact that mass-losing stars form dust and are bright in the mid-infrared. We plan to (i) derive physical parameters of a large sample of dusty, evolved targets and estimate the amount of ejected mass, (ii) constrain evolutionary models, (iii) quantify the duration and frequency of episodic mass loss as a function of metallicity. The approach involves applying machine-learning algorithms to existing multi-band and time-series photometry of luminous sources in ~25 nearby galaxies. Dusty, luminous evolved massive stars will thus be automatically classified and follow-up spectroscopy will be obtained for selected targets. Atmospheric and SED modeling will yield parameters and estimates of time-dependent mass loss for ~1000 luminous stars. The emerging trend for the ubiquity of episodic mass loss, if confirmed, will be key to understanding the explosive early Universe and will have profound consequences for low-metallicity stars, reionization, and the chemical evolution of galaxies.
Summary
Massive stars dominate their surroundings during their short lifetimes, while their explosive deaths impact the chemical evolution and spatial cohesion of their hosts. After birth, their evolution is largely dictated by their ability to remove layers of hydrogen from their envelopes. Multiple lines of evidence are pointing to violent, episodic mass-loss events being responsible for removing a large part of the massive stellar envelope, especially in low-metallicity galaxies. Episodic mass loss, however, is not understood theoretically, neither accounted for in state-of-the-art models of stellar evolution, which has far-reaching consequences for many areas of astronomy. We aim to determine whether episodic mass loss is a dominant process in the evolution of the most massive stars by conducting the first extensive, multi-wavelength survey of evolved massive stars in the nearby Universe. The project hinges on the fact that mass-losing stars form dust and are bright in the mid-infrared. We plan to (i) derive physical parameters of a large sample of dusty, evolved targets and estimate the amount of ejected mass, (ii) constrain evolutionary models, (iii) quantify the duration and frequency of episodic mass loss as a function of metallicity. The approach involves applying machine-learning algorithms to existing multi-band and time-series photometry of luminous sources in ~25 nearby galaxies. Dusty, luminous evolved massive stars will thus be automatically classified and follow-up spectroscopy will be obtained for selected targets. Atmospheric and SED modeling will yield parameters and estimates of time-dependent mass loss for ~1000 luminous stars. The emerging trend for the ubiquity of episodic mass loss, if confirmed, will be key to understanding the explosive early Universe and will have profound consequences for low-metallicity stars, reionization, and the chemical evolution of galaxies.
Max ERC Funding
1 128 750 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym ASTROFLOW
Project The influence of stellar outflows on exoplanetary mass loss
Researcher (PI) Aline VIDOTTO
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), PE9, ERC-2018-COG
Summary ASTROFLOW aims to make ground-breaking progress in our physical understanding of exoplanetary mass loss, by quantifying the influence of stellar outflows on atmospheric escape of close-in exoplanets. Escape plays a key role in planetary evolution, population, and potential to develop life. Stellar irradiation and outflows affect planetary mass loss: irradiation heats planetary atmospheres, which inflate and more likely escape; outflows cause pressure confinement around otherwise freely escaping atmospheres. This external pressure can increase, reduce or even suppress escape rates; its effects on exoplanetary mass loss remain largely unexplored due to the complexity of such interactions. I will fill this knowledge gap by developing a novel modelling framework of atmospheric escape that will, for the first time, consider the effects of realistic stellar outflows on exoplanetary mass loss. My expertise in stellar wind theory and 3D magnetohydrodynamic simulations is crucial for producing the next-generation models of planetary escape. My framework will consist of state-of-the-art, time-dependent, 3D simulations of stellar outflows (Method 1), which will be coupled to novel 3D simulations of atmospheric escape (Method 2). My models will account for the major underlying physical processes of mass loss. With this, I will determine the response of planetary mass loss to realistic stellar particle, magnetic and radiation environments and will characterise the physical conditions of the escaping material. I will compute how its extinction varies during transit and compare synthetic line profiles to atmospheric escape observations from, eg, Hubble and our NASA cubesat CUTE. Strong synergy with upcoming observations (JWST, TESS, SPIRou, CARMENES) also exists. Determining the lifetime of planetary atmospheres is essential to understanding populations of exoplanets. ASTROFLOW’s work will be the foundation for future research of how exoplanets evolve under mass-loss processes.
Summary
ASTROFLOW aims to make ground-breaking progress in our physical understanding of exoplanetary mass loss, by quantifying the influence of stellar outflows on atmospheric escape of close-in exoplanets. Escape plays a key role in planetary evolution, population, and potential to develop life. Stellar irradiation and outflows affect planetary mass loss: irradiation heats planetary atmospheres, which inflate and more likely escape; outflows cause pressure confinement around otherwise freely escaping atmospheres. This external pressure can increase, reduce or even suppress escape rates; its effects on exoplanetary mass loss remain largely unexplored due to the complexity of such interactions. I will fill this knowledge gap by developing a novel modelling framework of atmospheric escape that will, for the first time, consider the effects of realistic stellar outflows on exoplanetary mass loss. My expertise in stellar wind theory and 3D magnetohydrodynamic simulations is crucial for producing the next-generation models of planetary escape. My framework will consist of state-of-the-art, time-dependent, 3D simulations of stellar outflows (Method 1), which will be coupled to novel 3D simulations of atmospheric escape (Method 2). My models will account for the major underlying physical processes of mass loss. With this, I will determine the response of planetary mass loss to realistic stellar particle, magnetic and radiation environments and will characterise the physical conditions of the escaping material. I will compute how its extinction varies during transit and compare synthetic line profiles to atmospheric escape observations from, eg, Hubble and our NASA cubesat CUTE. Strong synergy with upcoming observations (JWST, TESS, SPIRou, CARMENES) also exists. Determining the lifetime of planetary atmospheres is essential to understanding populations of exoplanets. ASTROFLOW’s work will be the foundation for future research of how exoplanets evolve under mass-loss processes.
Max ERC Funding
1 999 956 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym ATMOPACS
Project Atmospheric Organic Particulate Matter, Air Quality and Climate Change Studies
Researcher (PI) Spyridon Pandis
Host Institution (HI) FOUNDATION FOR RESEARCH AND TECHNOLOGY HELLAS
Call Details Advanced Grant (AdG), PE10, ERC-2010-AdG_20100224
Summary Despite its importance for human health and climate change organic aerosol (OA) remains one of the least understood aspects of atmospheric chemistry. We propose to develop an innovative new framework for the description of OA in chemical transport and climate models that will be able to overcome the challenges posed by the chemical complexity of OA while capturing its essential features.
The objectives of ATMOPACS are: (i) The development of a new unified framework for the description of OA based on its two most important parameters: volatility and oxygen content. (ii) The development of measurement techniques for the volatility distribution and oxygen content distribution of OA. This will allow the experimental characterization of OA in this new “coordinate system”. (iii) The study of the major OA processes (partitioning, chemical aging, hygroscopicity, CCN formation, nucleation) in this new framework combining lab and field measurements. (iv) The development and evaluation of the next generation of regional and global CTMs using the above framework. (v) The quantification of the importance of the various sources and formation pathways of OA in Europe and the world, of the sensitivity of OA to emission control strategies, and its role in the direct and indirect effects of aerosols on climate.
The proposed work involves a combination of laboratory measurements, field measurements including novel “atmospheric perturbation experiments”, OA model development, and modelling in urban, regional, and global scales. Therefore, it will span the system scales starting from the nanoscale to the global. The modelling tools that will be developed will be made available to all other research groups.
Summary
Despite its importance for human health and climate change organic aerosol (OA) remains one of the least understood aspects of atmospheric chemistry. We propose to develop an innovative new framework for the description of OA in chemical transport and climate models that will be able to overcome the challenges posed by the chemical complexity of OA while capturing its essential features.
The objectives of ATMOPACS are: (i) The development of a new unified framework for the description of OA based on its two most important parameters: volatility and oxygen content. (ii) The development of measurement techniques for the volatility distribution and oxygen content distribution of OA. This will allow the experimental characterization of OA in this new “coordinate system”. (iii) The study of the major OA processes (partitioning, chemical aging, hygroscopicity, CCN formation, nucleation) in this new framework combining lab and field measurements. (iv) The development and evaluation of the next generation of regional and global CTMs using the above framework. (v) The quantification of the importance of the various sources and formation pathways of OA in Europe and the world, of the sensitivity of OA to emission control strategies, and its role in the direct and indirect effects of aerosols on climate.
The proposed work involves a combination of laboratory measurements, field measurements including novel “atmospheric perturbation experiments”, OA model development, and modelling in urban, regional, and global scales. Therefore, it will span the system scales starting from the nanoscale to the global. The modelling tools that will be developed will be made available to all other research groups.
Max ERC Funding
2 496 000 €
Duration
Start date: 2011-01-01, End date: 2015-12-31
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 BioWater
Project Development of new chemical imaging techniques to understand the function of water in biocompatibility, biodegradation and biofouling
Researcher (PI) Aoife Ann Gowen
Host Institution (HI) UNIVERSITY COLLEGE DUBLIN, NATIONAL UNIVERSITY OF IRELAND, DUBLIN
Call Details Starting Grant (StG), PE8, ERC-2013-StG
Summary Water is the first molecule to come into contact with biomaterials in biological systems and thus essential to the processes of biodegradation, biocompatibility and biofouling. Despite this fact, little is currently known about how biomaterials interact with water. This knowledge is crucial for the development and optimisation of novel functional biomaterials for human health (e.g. biosensing devices, erodible biomaterials, drug release carriers, wound dressings). BioWater will develop near and mid infrared chemical imaging (NIR-MIR-CI) techniques to investigate the fundamental interaction between biomaterials and water in order to understand the key processes of biodegradation, biocompatibility and biofouling. This ambitious yet achievable project will focus on two major categories of biomaterials relevant to human health: extracellular collagens and synthetic biopolymers. Initially, interactions between these biomaterials and water will be investigated; subsequently interactions with more complicated matrices (e.g. protein solutions and cellular systems) will be studied. CI data will be correlated with standard surface characterization, biocompatibility and biodegradation measurements. Molecular dynamic simulations will complement this work to identify the most probable molecular structures of water at different biomaterial interfaces.
Advanced understanding of the role of water in biocompatibility, biofouling and biodegradation processes will facilitate the optimization of biomaterials tailored to specific cellular environments with a broad range of therapeutic applications (e.g. drug eluting stents, tissue engineering, wound healing). The new NIR-MIR-CI/chemometric methodologies developed in BioWater will allow for the rapid characterization and monitoring of novel biomaterials at pre-clinical stages, improving process control by overcoming the laborious and time consuming large-scale sampling methods currently required in biomaterials development.
Summary
Water is the first molecule to come into contact with biomaterials in biological systems and thus essential to the processes of biodegradation, biocompatibility and biofouling. Despite this fact, little is currently known about how biomaterials interact with water. This knowledge is crucial for the development and optimisation of novel functional biomaterials for human health (e.g. biosensing devices, erodible biomaterials, drug release carriers, wound dressings). BioWater will develop near and mid infrared chemical imaging (NIR-MIR-CI) techniques to investigate the fundamental interaction between biomaterials and water in order to understand the key processes of biodegradation, biocompatibility and biofouling. This ambitious yet achievable project will focus on two major categories of biomaterials relevant to human health: extracellular collagens and synthetic biopolymers. Initially, interactions between these biomaterials and water will be investigated; subsequently interactions with more complicated matrices (e.g. protein solutions and cellular systems) will be studied. CI data will be correlated with standard surface characterization, biocompatibility and biodegradation measurements. Molecular dynamic simulations will complement this work to identify the most probable molecular structures of water at different biomaterial interfaces.
Advanced understanding of the role of water in biocompatibility, biofouling and biodegradation processes will facilitate the optimization of biomaterials tailored to specific cellular environments with a broad range of therapeutic applications (e.g. drug eluting stents, tissue engineering, wound healing). The new NIR-MIR-CI/chemometric methodologies developed in BioWater will allow for the rapid characterization and monitoring of novel biomaterials at pre-clinical stages, improving process control by overcoming the laborious and time consuming large-scale sampling methods currently required in biomaterials development.
Max ERC Funding
1 487 682 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym BONDS
Project Bilayered ON-Demand Scaffolds: On-Demand Delivery from induced Pluripotent Stem Cell Derived Scaffolds for Diabetic Foot Ulcers
Researcher (PI) Cathal KEARNEY
Host Institution (HI) ROYAL COLLEGE OF SURGEONS IN IRELAND
Call Details Starting Grant (StG), PE8, ERC-2017-STG
Summary This program’s goal is to develop a scaffold using a new biomaterial source that is functionalised with on-demand delivery of genes for coordinated healing of diabetic foot ulcers (DFUs). DFUs are chronic wounds that are often recalcitrant to treatment, which devastatingly results in lower leg amputation. This project builds on the PI’s experience growing matrix from induced-pluripotent stem cell derived (iPS)-fibroblasts and in developing on-demand drug delivery technologies. The aim of this project is to first develop a SiPS: a scaffold from iPS-fibroblast grown matrix, which has never been tested as a source material for scaffolds. iPS-fibroblasts grow a more pro-repair and angiogenic matrix than (non-iPS) adult fibroblasts. The SiPS structure will be bilayered to mimic native skin: dermis made mostly by fibroblasts and epidermis made by keratinocytes. The dermal layer will consist of a porous scaffold with optimised pore size and mechanical properties and the epidermal layer will be film-like, optimised for keratinisation.
Second, the SiPS will be functionalised with delivery of plasmid-DNA (platelet derived growth factor gene, pPDGF) to direct angiogenesis on-demand. As DFUs undergo uncoordinated healing, timed pPDGF delivery will guide them through angiogenesis and healing. To achieve this, alginate microparticles, designed to respond to ultrasound by releasing pPDGF, will be interspersed throughout the SiPS. This BONDS will be tested in an in vivo pre-clinical DFU model to confirm its ability to heal wounds by providing cells with the appropriate biomimetic scaffold environment and timed directions for healing. With >100 million current diabetics expected to get a DFU, the BONDS would have a powerful clinical impact.
This research program combines a disruptive technology, the SiPS, with a new platform for on-demand delivery of pDNA to heal DFUs. The PI will build his lab around these innovative platforms, adapting them for treatment of diverse complex wounds.
Summary
This program’s goal is to develop a scaffold using a new biomaterial source that is functionalised with on-demand delivery of genes for coordinated healing of diabetic foot ulcers (DFUs). DFUs are chronic wounds that are often recalcitrant to treatment, which devastatingly results in lower leg amputation. This project builds on the PI’s experience growing matrix from induced-pluripotent stem cell derived (iPS)-fibroblasts and in developing on-demand drug delivery technologies. The aim of this project is to first develop a SiPS: a scaffold from iPS-fibroblast grown matrix, which has never been tested as a source material for scaffolds. iPS-fibroblasts grow a more pro-repair and angiogenic matrix than (non-iPS) adult fibroblasts. The SiPS structure will be bilayered to mimic native skin: dermis made mostly by fibroblasts and epidermis made by keratinocytes. The dermal layer will consist of a porous scaffold with optimised pore size and mechanical properties and the epidermal layer will be film-like, optimised for keratinisation.
Second, the SiPS will be functionalised with delivery of plasmid-DNA (platelet derived growth factor gene, pPDGF) to direct angiogenesis on-demand. As DFUs undergo uncoordinated healing, timed pPDGF delivery will guide them through angiogenesis and healing. To achieve this, alginate microparticles, designed to respond to ultrasound by releasing pPDGF, will be interspersed throughout the SiPS. This BONDS will be tested in an in vivo pre-clinical DFU model to confirm its ability to heal wounds by providing cells with the appropriate biomimetic scaffold environment and timed directions for healing. With >100 million current diabetics expected to get a DFU, the BONDS would have a powerful clinical impact.
This research program combines a disruptive technology, the SiPS, with a new platform for on-demand delivery of pDNA to heal DFUs. The PI will build his lab around these innovative platforms, adapting them for treatment of diverse complex wounds.
Max ERC Funding
1 372 135 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym BONEMECHBIO
Project Frontier research in bone mechanobiology during normal physiology, disease and for tissue regeneration
Researcher (PI) Laoise Maria Cunningham
Host Institution (HI) NATIONAL UNIVERSITY OF IRELAND GALWAY
Call Details Starting Grant (StG), PE8, ERC-2010-StG_20091028
Summary While previous studies have investigated cell-signalling pathways that facilitate mechanotransduction and have provided a wealth of data, to date, in vivo mechanobiology is not fully understood. In the research study proposed the applicant will embark upon frontier research to delineate these specific aspects of bone mechanotransduction during normal physiology, disease and for tissue regeneration purposes. If these quantities were better understood the proposed research program will deliver significant advances in the understanding of the mechanical regulation of bone remodelling during normal physiology and osteoporosis, and will enhance approaches for regeneration of bone tissue for treatment of bone pathologies. The primary objective is to delineate the normal mechanosensory and signalling mechanisms of bone cells. The secondary objective is to determine whether the regulatory role of bone cells is inhibited or impaired during bone diseases such as osteoporosis. The final objective of this project is to develop an in vitro mechanical loading device that can enhance bone tissue regeneration and thereby advance current treatment approaches for bone pathologies. To address these objectives, five hypotheses have been defined, each of which will underpin the research of five work packages. A combination of experimental studies, using animal models and in vitro cell culture, and computational modelling will be taken to test each of these hypotheses. Answering these hypotheses will bring us closer to an understanding of the origins of bone mechanobiology and diseases such as osteoporosis. Furthermore, the results of these studies will facilitate development of novel approaches to enhance bone regeneration in vitro.
Summary
While previous studies have investigated cell-signalling pathways that facilitate mechanotransduction and have provided a wealth of data, to date, in vivo mechanobiology is not fully understood. In the research study proposed the applicant will embark upon frontier research to delineate these specific aspects of bone mechanotransduction during normal physiology, disease and for tissue regeneration purposes. If these quantities were better understood the proposed research program will deliver significant advances in the understanding of the mechanical regulation of bone remodelling during normal physiology and osteoporosis, and will enhance approaches for regeneration of bone tissue for treatment of bone pathologies. The primary objective is to delineate the normal mechanosensory and signalling mechanisms of bone cells. The secondary objective is to determine whether the regulatory role of bone cells is inhibited or impaired during bone diseases such as osteoporosis. The final objective of this project is to develop an in vitro mechanical loading device that can enhance bone tissue regeneration and thereby advance current treatment approaches for bone pathologies. To address these objectives, five hypotheses have been defined, each of which will underpin the research of five work packages. A combination of experimental studies, using animal models and in vitro cell culture, and computational modelling will be taken to test each of these hypotheses. Answering these hypotheses will bring us closer to an understanding of the origins of bone mechanobiology and diseases such as osteoporosis. Furthermore, the results of these studies will facilitate development of novel approaches to enhance bone regeneration in vitro.
Max ERC Funding
1 499 911 €
Duration
Start date: 2011-02-01, End date: 2016-01-31
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 CARBENZYMES
Project Probing the relevance of carbene binding motifs in enzyme reactivity
Researcher (PI) Martin Albrecht
Host Institution (HI) UNIVERSITY COLLEGE DUBLIN, NATIONAL UNIVERSITY OF IRELAND, DUBLIN
Call Details Starting Grant (StG), PE4, ERC-2007-StG
Summary Histidine (His) is an ubiquitous ligand in the active site of metalloenzymes that is assumed by default to bind the metal center through one of its nitrogen atoms. However, protonation of His, which is likely to occur in locally slightly acidic environment, gives imidazolium sites that can bind a metal in a carbene-type structure as found in N-heterocyclic carbene complexes. Such carbene bonding has a dramatic effect on the properties of the metal center and may provide a rational for the mode of action of metalloenzymes that are still lacking a solid understanding. Up to now, the possibility of carbene bonding has been completely overlooked. Hence, any evidence for such His coordination via carbon will induce a shift of paradigm in classical peptide chemistry and will be directly included in basic textbooks. Moreover, this unprecedented bonding mode will provide access to unique and hitherto unknown reactivity patterns for artificial enzyme mimics. Undoubtedly, such a break-through will set a new stage in modern metalloenzyme research. A multicentered approach is proposed to identify for the first time carbene bonding in enzymes. This approach unconventionally combines the current frontiers of organometallic and biochemical knowledge and hence crosses traditional boarders. Specifically, we aim at probing carbene bonding of His by identifying reactivity patterns that are selective for metal-carbenes but not for metal-imine complexes. This will allow for efficient screening of large classes of metalloenzymes. In parallel, active site models will be constructed in which the His ligand is substituted by a heterocyclic carbene as a rigidly C-bonding His analog. For this purpose chemical synthesis will be considered as well as enzyme mutagenesis and subsequent carbene coordination. While such new bioorganometallic entities will be highly attractive to probe the influence of C-bound His on the metal site, they also provide conceputally new types of versatile catalysts.
Summary
Histidine (His) is an ubiquitous ligand in the active site of metalloenzymes that is assumed by default to bind the metal center through one of its nitrogen atoms. However, protonation of His, which is likely to occur in locally slightly acidic environment, gives imidazolium sites that can bind a metal in a carbene-type structure as found in N-heterocyclic carbene complexes. Such carbene bonding has a dramatic effect on the properties of the metal center and may provide a rational for the mode of action of metalloenzymes that are still lacking a solid understanding. Up to now, the possibility of carbene bonding has been completely overlooked. Hence, any evidence for such His coordination via carbon will induce a shift of paradigm in classical peptide chemistry and will be directly included in basic textbooks. Moreover, this unprecedented bonding mode will provide access to unique and hitherto unknown reactivity patterns for artificial enzyme mimics. Undoubtedly, such a break-through will set a new stage in modern metalloenzyme research. A multicentered approach is proposed to identify for the first time carbene bonding in enzymes. This approach unconventionally combines the current frontiers of organometallic and biochemical knowledge and hence crosses traditional boarders. Specifically, we aim at probing carbene bonding of His by identifying reactivity patterns that are selective for metal-carbenes but not for metal-imine complexes. This will allow for efficient screening of large classes of metalloenzymes. In parallel, active site models will be constructed in which the His ligand is substituted by a heterocyclic carbene as a rigidly C-bonding His analog. For this purpose chemical synthesis will be considered as well as enzyme mutagenesis and subsequent carbene coordination. While such new bioorganometallic entities will be highly attractive to probe the influence of C-bound His on the metal site, they also provide conceputally new types of versatile catalysts.
Max ERC Funding
1 249 808 €
Duration
Start date: 2008-07-01, End date: 2013-06-30
Project acronym CAUSALPATH
Project Next Generation Causal Analysis: Inspired by the Induction of Biological Pathways from Cytometry Data
Researcher (PI) Ioannis Tsamardinos
Host Institution (HI) PANEPISTIMIO KRITIS
Call Details Consolidator Grant (CoG), PE6, ERC-2013-CoG
Summary Discovering the causal mechanisms of a complex system of interacting components is necessary in order to control it. Computational Causal Discovery (CD) is a field that offers the potential to discover causal relations under certain conditions from observational data alone or with a limited number of interventions/manipulations.
An important, challenging biological problem that may take decades of experimental work is the induction of biological cellular pathways; pathways are informal causal models indispensable in biological research and drug design. Recent exciting advances in flow/mass cytometry biotechnology allow the generation of large-sample datasets containing measurements on single cells, thus setting the problem of pathway learning suitable for CD methods.
CAUSALPATH builds upon and further advances recent breakthrough developments in CD methods to enable the induction of biological pathways from cytometry and other omics data. As a testbed problem we focus on the differentiation of human T-cells; these are involved in autoimmune and inflammatory diseases, as well as cancer and thus, are targets of new drug development for a range of chronic diseases. The biological problem acts as our campus for general novel formalisms, practical algorithms, and useful tools development, pointing to fundamental CD problems: presence of feedback cycles, presence of latent confounding variables, CD from time-course data, Integrative Causal Analysis (INCA) of heterogeneous datasets and others.
Three features complement CAUSALPATH’s approach: (A) methods development will co-evolve with biological wet-lab experiments periodically testing the algorithmic postulates, (B) Open-source tools will be developed for the non-expert, and (C) Commercial exploitation of the results will be sought out.
CAUSALPATH brings together an interdisciplinary team, committed to this vision. It builds upon the PI’s group recent important results on INCA algorithms.
Summary
Discovering the causal mechanisms of a complex system of interacting components is necessary in order to control it. Computational Causal Discovery (CD) is a field that offers the potential to discover causal relations under certain conditions from observational data alone or with a limited number of interventions/manipulations.
An important, challenging biological problem that may take decades of experimental work is the induction of biological cellular pathways; pathways are informal causal models indispensable in biological research and drug design. Recent exciting advances in flow/mass cytometry biotechnology allow the generation of large-sample datasets containing measurements on single cells, thus setting the problem of pathway learning suitable for CD methods.
CAUSALPATH builds upon and further advances recent breakthrough developments in CD methods to enable the induction of biological pathways from cytometry and other omics data. As a testbed problem we focus on the differentiation of human T-cells; these are involved in autoimmune and inflammatory diseases, as well as cancer and thus, are targets of new drug development for a range of chronic diseases. The biological problem acts as our campus for general novel formalisms, practical algorithms, and useful tools development, pointing to fundamental CD problems: presence of feedback cycles, presence of latent confounding variables, CD from time-course data, Integrative Causal Analysis (INCA) of heterogeneous datasets and others.
Three features complement CAUSALPATH’s approach: (A) methods development will co-evolve with biological wet-lab experiments periodically testing the algorithmic postulates, (B) Open-source tools will be developed for the non-expert, and (C) Commercial exploitation of the results will be sought out.
CAUSALPATH brings together an interdisciplinary team, committed to this vision. It builds upon the PI’s group recent important results on INCA algorithms.
Max ERC Funding
1 724 000 €
Duration
Start date: 2015-01-01, End date: 2019-12-31
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 CiliaMechanoBio
Project Primary Cilium-Mediated Mesenchymal Stem Cell Mechanobiology in Bone
Researcher (PI) David Hoey
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), PE8, ERC-2013-StG
Summary Every 30 seconds a person suffers an osteoporosis-related bone fracture in the EU, resulting in significant morbidity, mortality, and health-care costs estimated at €36billion annually. Current therapeutics target bone resorbing osteoclasts, but these are associated with severe side effects. Osteoporosis arises when mesenchymal stem cells (MSC) fail to produce sufficient numbers of bone forming osteoblasts. A key regulator of MSC behaviour is physical loading, yet the mechanisms by which MSCs sense and respond to changes in their mechanical environment are virtually unknown. Primary cilia are nearly ubiquitous ‘antennae-like’ cellular organelles that have very recently emerged as extracellular mechano/chemo-sensors and thus, are strong candidates to play a role in regulating MSC responses in bone. Therefore, the objective of this research program is to determine the role of the primary cilium and associated molecular components in the osteogenic differentiation and recruitment of human MSCs in loading-induced bone adaptation. This will be achieved through ground-breaking in vitro and in vivo techniques developed by the applicant. The knowledge generated in this proposal will represent a profound advance in our understanding of stem cell mechanobiology. In particular, the identification of the cilium and associated molecules as central to stem cell behaviour will lead to the direct manipulation of MSCs via novel cilia-targeted therapeutics that mimic the regenerative influence of loading at a molecular level. These novel therapeutics would therefore target bone formation, providing an alternative path to treatment, resulting in an improved supply of bone forming cells, preventing osteoporosis. Furthermore, these novel therapeutics will be incorporated into biomaterials, generating bioactive osteoinductive scaffolds. These advances will not only improve quality of life for the patient but will significantly reduce the financial burden of bone loss diseases in the EU.
Summary
Every 30 seconds a person suffers an osteoporosis-related bone fracture in the EU, resulting in significant morbidity, mortality, and health-care costs estimated at €36billion annually. Current therapeutics target bone resorbing osteoclasts, but these are associated with severe side effects. Osteoporosis arises when mesenchymal stem cells (MSC) fail to produce sufficient numbers of bone forming osteoblasts. A key regulator of MSC behaviour is physical loading, yet the mechanisms by which MSCs sense and respond to changes in their mechanical environment are virtually unknown. Primary cilia are nearly ubiquitous ‘antennae-like’ cellular organelles that have very recently emerged as extracellular mechano/chemo-sensors and thus, are strong candidates to play a role in regulating MSC responses in bone. Therefore, the objective of this research program is to determine the role of the primary cilium and associated molecular components in the osteogenic differentiation and recruitment of human MSCs in loading-induced bone adaptation. This will be achieved through ground-breaking in vitro and in vivo techniques developed by the applicant. The knowledge generated in this proposal will represent a profound advance in our understanding of stem cell mechanobiology. In particular, the identification of the cilium and associated molecules as central to stem cell behaviour will lead to the direct manipulation of MSCs via novel cilia-targeted therapeutics that mimic the regenerative influence of loading at a molecular level. These novel therapeutics would therefore target bone formation, providing an alternative path to treatment, resulting in an improved supply of bone forming cells, preventing osteoporosis. Furthermore, these novel therapeutics will be incorporated into biomaterials, generating bioactive osteoinductive scaffolds. These advances will not only improve quality of life for the patient but will significantly reduce the financial burden of bone loss diseases in the EU.
Max ERC Funding
1 455 068 €
Duration
Start date: 2013-11-01, End date: 2018-10-31
Project acronym CODAMODA
Project Controlling Data Movement in the Digital Age
Researcher (PI) Aggelos Kiayias
Host Institution (HI) ETHNIKO KAI KAPODISTRIAKO PANEPISTIMIO ATHINON
Call Details Starting Grant (StG), PE6, ERC-2010-StG_20091028
Summary Nowadays human intellectual product is increasingly produced and disseminated solely in digital form. The capability of digital data for effortless reproduction and transfer has lead to a true revolution that impacts every aspect of human creativity. Nevertheless, as with every technological revolution, this digital media revolution comes with a dark side that, if left unaddressed, it will limit its impact and may counter its potential advantages. In particular, the way we produce and disseminate digital content today does not lend itself to controlling the way data move and change. It turns out that the power of being digital can be a double-edged sword: the ease of production, dissemination and editing also implies the ease of misappropriation, plagiarism and improper modification.
To counter the above problems, the proposed research activity will focus on the development of a new generation of enabling cryptographic technologies that have the power to facilitate the appropriate controls for data movement. Using the techniques developed in this project it will be feasible to build digital content distribution systems where content producers will have the full possible control on the dissemination of their intellectual product, while at the same time the rights of the end-users in terms of privacy and fair use can be preserved. The PI is uniquely qualified to carry out the proposed research activity as he has extensive prior experience in making innovations in the area of digital content distribution as well as in the management of research projects. As part of the project activities, the PI will establish the CODAMODA laboratory in the University of Athens and will seek opportunities for technology transfer and interdisciplinary work with the legal science community.
Summary
Nowadays human intellectual product is increasingly produced and disseminated solely in digital form. The capability of digital data for effortless reproduction and transfer has lead to a true revolution that impacts every aspect of human creativity. Nevertheless, as with every technological revolution, this digital media revolution comes with a dark side that, if left unaddressed, it will limit its impact and may counter its potential advantages. In particular, the way we produce and disseminate digital content today does not lend itself to controlling the way data move and change. It turns out that the power of being digital can be a double-edged sword: the ease of production, dissemination and editing also implies the ease of misappropriation, plagiarism and improper modification.
To counter the above problems, the proposed research activity will focus on the development of a new generation of enabling cryptographic technologies that have the power to facilitate the appropriate controls for data movement. Using the techniques developed in this project it will be feasible to build digital content distribution systems where content producers will have the full possible control on the dissemination of their intellectual product, while at the same time the rights of the end-users in terms of privacy and fair use can be preserved. The PI is uniquely qualified to carry out the proposed research activity as he has extensive prior experience in making innovations in the area of digital content distribution as well as in the management of research projects. As part of the project activities, the PI will establish the CODAMODA laboratory in the University of Athens and will seek opportunities for technology transfer and interdisciplinary work with the legal science community.
Max ERC Funding
1 212 960 €
Duration
Start date: 2011-04-01, End date: 2017-03-31
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 COLLREGEN
Project Collagen scaffolds for bone regeneration: applied biomaterials, bioreactor and stem cell technology
Researcher (PI) Fergal Joseph O'brien
Host Institution (HI) ROYAL COLLEGE OF SURGEONS IN IRELAND
Call Details Starting Grant (StG), PE8, ERC-2009-StG
Summary Regenerative medicine aims to regenerate damaged tissues by developing functional cell, tissue, and organ substitutes to repair, replace or enhance biological function in damaged tissues. The focus of this research programme is to develop bone graft substitute biomaterials and laboratory-engineered bone tissue for implantation in damaged sites. At a simplistic level, biological tissues consist of cells, signalling mechanisms and extracellular matrix. Regenerative medicine/tissue engineering technologies are based on this biological triad and involve the successful interaction between three components: the scaffold that holds the cells together to create the tissues physical form, the cells that create the tissue, and the biological signalling mechanisms (such as growth factors or bioreactors) that direct the cells to express the desired tissue phenotype. The research proposed in this project includes specific projects in all three areas. The programme will be centred on the collagen-based biomaterials developed in the applicant s laboratory and will incorporate cutting edge stem cell technologies, growth factor delivery, gene therapy and bioreactor technology which will translate to in vivo tissue repair. This translational research programme will be divided into four specific themes: (i) development of novel osteoinductive and angiogenic smart scaffolds for bone tissue regeneration, (ii) scaffold and stem cell therapies for bone tissue regeneration, (iii) bone tissue engineering using a flow perfusion bioreactor and (iv) in vivo bone repair using engineered bone and smart scaffolds.
Summary
Regenerative medicine aims to regenerate damaged tissues by developing functional cell, tissue, and organ substitutes to repair, replace or enhance biological function in damaged tissues. The focus of this research programme is to develop bone graft substitute biomaterials and laboratory-engineered bone tissue for implantation in damaged sites. At a simplistic level, biological tissues consist of cells, signalling mechanisms and extracellular matrix. Regenerative medicine/tissue engineering technologies are based on this biological triad and involve the successful interaction between three components: the scaffold that holds the cells together to create the tissues physical form, the cells that create the tissue, and the biological signalling mechanisms (such as growth factors or bioreactors) that direct the cells to express the desired tissue phenotype. The research proposed in this project includes specific projects in all three areas. The programme will be centred on the collagen-based biomaterials developed in the applicant s laboratory and will incorporate cutting edge stem cell technologies, growth factor delivery, gene therapy and bioreactor technology which will translate to in vivo tissue repair. This translational research programme will be divided into four specific themes: (i) development of novel osteoinductive and angiogenic smart scaffolds for bone tissue regeneration, (ii) scaffold and stem cell therapies for bone tissue regeneration, (iii) bone tissue engineering using a flow perfusion bioreactor and (iv) in vivo bone repair using engineered bone and smart scaffolds.
Max ERC Funding
1 999 530 €
Duration
Start date: 2009-11-01, End date: 2015-09-30
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 CutLoops
Project Loop amplitudes in quantum field theory
Researcher (PI) Ruth Britto
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), PE2, ERC-2014-CoG
Summary The traditional formulation of relativistic quantum theory is ill-equipped to handle the range of difficult computations needed to describe particle collisions at the Large Hadron Collider (LHC) within a suitable time frame. Yet, recent work shows that probability amplitudes in quantum gauge field theories, such as those describing the Standard Model and its extensions, take surprisingly simple forms. The simplicity indicates deep structure in gauge theory that has already led to dramatic computational improvements, but remains to be fully understood. For precision calculations and investigations of the deep structure of gauge theory, a comprehensive method for computing multi-loop amplitudes systematically and efficiently must be found.
The goal of this proposal is to construct a new and complete approach to computing amplitudes from a detailed understanding of their singularities, based on prior successes of so-called on-shell methods combined with the latest developments in the mathematics of Feynman integrals. Scattering processes relevant to the LHC and to formal investigations of quantum field theory will be computed within the new framework.
Summary
The traditional formulation of relativistic quantum theory is ill-equipped to handle the range of difficult computations needed to describe particle collisions at the Large Hadron Collider (LHC) within a suitable time frame. Yet, recent work shows that probability amplitudes in quantum gauge field theories, such as those describing the Standard Model and its extensions, take surprisingly simple forms. The simplicity indicates deep structure in gauge theory that has already led to dramatic computational improvements, but remains to be fully understood. For precision calculations and investigations of the deep structure of gauge theory, a comprehensive method for computing multi-loop amplitudes systematically and efficiently must be found.
The goal of this proposal is to construct a new and complete approach to computing amplitudes from a detailed understanding of their singularities, based on prior successes of so-called on-shell methods combined with the latest developments in the mathematics of Feynman integrals. Scattering processes relevant to the LHC and to formal investigations of quantum field theory will be computed within the new framework.
Max ERC Funding
1 954 065 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym D-TECT
Project Does dust triboelectrification affect our climate?
Researcher (PI) Vasileios AMOIRIDIS
Host Institution (HI) NATIONAL OBSERVATORY OF ATHENS
Call Details Consolidator Grant (CoG), PE10, ERC-2016-COG
Summary The recent IPCC report identifies mineral dust and the associated uncertainties in climate projections as key topics for future research. Dust size distribution in climate models controls the dust-radiation-cloud interactions and is a major contributor to these uncertainties. Observations show that the coarse mode of dust can be sustained during long-range transport, while current understanding fails in explaining why the lifetime of large airborne dust particles is longer than expected from gravitational settling theories. This discrepancy between observations and theory suggests that other processes counterbalance the effect of gravity along transport. D-TECT envisages filling this knowledge gap by studying the contribution of the triboelectrification (contact electrification) on particle removal processes. Our hypothesis is that triboelectric charging generates adequate electric fields to hold large dust particles up in the atmosphere. D-TECT aims to (i) parameterize the physical mechanisms responsible for dust triboelectrification; (ii) assess the impact of electrification on dust settling; (iii) quantify the climatic impacts of the process, particularly the effect on the dust size evolution during transport, on dry deposition and on CCN/IN reservoirs, and the effect of the electric field on particle orientation and on radiative transfer. The approach involves the development of a novel specialized high-power lidar system to detect and characterize aerosol particle orientation and a large-scale field experiment in the Mediterranean Basin using unprecedented ground-based remote sensing and airborne in-situ observation synergies. Considering aerosol-electricity interactions, the observations will be used to improve theoretical understanding and simulations of dust lifecycle. The project will provide new fundamental understanding, able to open new horizons for weather and climate science, including biogeochemistry, volcanic ash and extraterrestrial dust research.
Summary
The recent IPCC report identifies mineral dust and the associated uncertainties in climate projections as key topics for future research. Dust size distribution in climate models controls the dust-radiation-cloud interactions and is a major contributor to these uncertainties. Observations show that the coarse mode of dust can be sustained during long-range transport, while current understanding fails in explaining why the lifetime of large airborne dust particles is longer than expected from gravitational settling theories. This discrepancy between observations and theory suggests that other processes counterbalance the effect of gravity along transport. D-TECT envisages filling this knowledge gap by studying the contribution of the triboelectrification (contact electrification) on particle removal processes. Our hypothesis is that triboelectric charging generates adequate electric fields to hold large dust particles up in the atmosphere. D-TECT aims to (i) parameterize the physical mechanisms responsible for dust triboelectrification; (ii) assess the impact of electrification on dust settling; (iii) quantify the climatic impacts of the process, particularly the effect on the dust size evolution during transport, on dry deposition and on CCN/IN reservoirs, and the effect of the electric field on particle orientation and on radiative transfer. The approach involves the development of a novel specialized high-power lidar system to detect and characterize aerosol particle orientation and a large-scale field experiment in the Mediterranean Basin using unprecedented ground-based remote sensing and airborne in-situ observation synergies. Considering aerosol-electricity interactions, the observations will be used to improve theoretical understanding and simulations of dust lifecycle. The project will provide new fundamental understanding, able to open new horizons for weather and climate science, including biogeochemistry, volcanic ash and extraterrestrial dust research.
Max ERC Funding
1 968 000 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym DANCER
Project DAtacommunications based on NanophotoniC Resonators
Researcher (PI) John William Whelan-Curtin
Host Institution (HI) CORK INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), PE7, ERC-2013-StG
Summary A key challenge for the 21st century is, therefore to provide billions of people with the means to access, move and manipulate, what has become, huge volumes of information. The environmental and economic implications becoming serious, making energy efficient data communications key to the operation of today’s society.
In this project, the Principal Investigator will develop a new framework for optical interconnects and provide a common platform that spans Fibre-to-the-home to chip-to-chip links, even as far as global on-chip interconnects. The project is based on the efficient coupling of the Photonic Crystal resonators with the outside world. These provide the ultimate confinement of light in both space and time allowing orders of magnitude improvements in performance relative to the state of the art, yet in a simpler simple system- the innovator’s dream. New versions of the key components of optical links- light sources, modulators and photo-detectors- will be realised in this new framework providing a new paradigm for energy efficient communication.
Summary
A key challenge for the 21st century is, therefore to provide billions of people with the means to access, move and manipulate, what has become, huge volumes of information. The environmental and economic implications becoming serious, making energy efficient data communications key to the operation of today’s society.
In this project, the Principal Investigator will develop a new framework for optical interconnects and provide a common platform that spans Fibre-to-the-home to chip-to-chip links, even as far as global on-chip interconnects. The project is based on the efficient coupling of the Photonic Crystal resonators with the outside world. These provide the ultimate confinement of light in both space and time allowing orders of magnitude improvements in performance relative to the state of the art, yet in a simpler simple system- the innovator’s dream. New versions of the key components of optical links- light sources, modulators and photo-detectors- will be realised in this new framework providing a new paradigm for energy efficient communication.
Max ERC Funding
1 495 450 €
Duration
Start date: 2013-12-01, End date: 2019-05-31
Project acronym DARE
Project Soil Foundation Structure Systems Beyond Conventional Seismic Failure Thresholds: Application to New or Existing Structures and Monuments
Researcher (PI) George Gazetas
Host Institution (HI) NATIONAL TECHNICAL UNIVERSITY OF ATHENS - NTUA
Call Details Advanced Grant (AdG), PE8, ERC-2008-AdG
Summary The main goal of the proposed research is to investigate the possibility of allowing below-ground support systems to respond to strong seismic shaking by going beyond a number of thresholds that would conventionally imply failure and are today forbidden by codes. Such thresholds include : (a) sliding at the soil-foundation interface ; (b) separation and uplifting of a shallow foundation from the soils ; (c) mobilization of bearing capacity failure mechanism for shallow foundations ; (d) structural yielding of pile foundations ; (e) combination of some of the above. Whereas under static loading conditions a slight exceedance of such thresholds leads to failure, the oscillatory nature of seismic shaking will allow such exceedances for a short period of time, with perhaps no detrimental or irreparable consequences. The latter take the form of permanent foundation displacements, rotations, or injuries , which the designer will aspire to confine within rational limits. The motivation and the need for this research has come from : (i) observations of actual behaviour in a variety of earthquakes ; conspicuous examples : the permanent tilting , overturning, and often survival of numerous buildings on extremely soft soil in Adapazari during the Kocaeli 1999 earthquake ; (ii) the foundation design of a number of critical structures (e.g., major bridge pier, air control tower, tall monuments, elevated water tanks,) against large seismic actions ; the disproportionately large overturning moment and/or base shear force of such slender structures can hardly be faced with today s conventional foundation methods, (iii) the need to seismically retrofit and rehabilitate older structures and historical monuments; (iv) structural yielding of pile foundations is now detectable (thanks to technological advances), thus eliminating one of the reasons for avoiding it.
Summary
The main goal of the proposed research is to investigate the possibility of allowing below-ground support systems to respond to strong seismic shaking by going beyond a number of thresholds that would conventionally imply failure and are today forbidden by codes. Such thresholds include : (a) sliding at the soil-foundation interface ; (b) separation and uplifting of a shallow foundation from the soils ; (c) mobilization of bearing capacity failure mechanism for shallow foundations ; (d) structural yielding of pile foundations ; (e) combination of some of the above. Whereas under static loading conditions a slight exceedance of such thresholds leads to failure, the oscillatory nature of seismic shaking will allow such exceedances for a short period of time, with perhaps no detrimental or irreparable consequences. The latter take the form of permanent foundation displacements, rotations, or injuries , which the designer will aspire to confine within rational limits. The motivation and the need for this research has come from : (i) observations of actual behaviour in a variety of earthquakes ; conspicuous examples : the permanent tilting , overturning, and often survival of numerous buildings on extremely soft soil in Adapazari during the Kocaeli 1999 earthquake ; (ii) the foundation design of a number of critical structures (e.g., major bridge pier, air control tower, tall monuments, elevated water tanks,) against large seismic actions ; the disproportionately large overturning moment and/or base shear force of such slender structures can hardly be faced with today s conventional foundation methods, (iii) the need to seismically retrofit and rehabilitate older structures and historical monuments; (iv) structural yielding of pile foundations is now detectable (thanks to technological advances), thus eliminating one of the reasons for avoiding it.
Max ERC Funding
2 399 992 €
Duration
Start date: 2008-12-01, End date: 2013-10-31
Project acronym DBSModel
Project Multiscale Modelling of the Neuromuscular System for Closed Loop Deep Brain Stimulation
Researcher (PI) Madeleine Mary Lowery
Host Institution (HI) UNIVERSITY COLLEGE DUBLIN, NATIONAL UNIVERSITY OF IRELAND, DUBLIN
Call Details Consolidator Grant (CoG), PE7, ERC-2014-CoG
Summary Deep brain stimulation (DBS) is an effective therapy for treating the symptoms of Parkinson’s disease (PD). Despite its success, the mechanisms of DBS are not understood and there is a need to improve DBS to improve long-term stimulation in a wider patient population, limit side-effects, and extend battery life. Currently DBS operates in ‘open-loop’, with stimulus parameters empirically set. Closed-loop DBS, which adjusts parameters based on the state of the system, has the potential to overcome current limitations to increase therapeutic efficacy while reducing side-effects, costs and energy. Several key questions need to be addressed before closed loop DBS can be implemented clinically.
This research will develop a new multiscale model of the neuromuscular system for closed-loop DBS. The model will simulate neural sensing and stimulation on a scale not previously considered, encompassing the electric field around the electrode, the effect on individual neurons and neural networks, and generation of muscle force. This will involve integration across multiple temporal and spatial scales, in a complex system with incomplete knowledge of system variables. Experiments will be conducted to validate the model, and identify new biomarkers of neural activity that can used with signals from the brain to enable continuous symptom monitoring. The model will be used to design a new control strategy for closed-loop DBS that can accommodate the nonlinear nature of the system, and short- and long-term changes in system behavior.
Though challenging, this research will provide new insights into the changes that take place in PD and the mechanisms by which DBS exerts its therapeutic influence. This knowledge will be used to design a new strategy for closed-loop DBS, ready for testing in patients, with the potential to significantly improve patient outcomes in PD and fundamentally change the way in which implanted devices utilise electrical stimulation to modulate neural activity.
Summary
Deep brain stimulation (DBS) is an effective therapy for treating the symptoms of Parkinson’s disease (PD). Despite its success, the mechanisms of DBS are not understood and there is a need to improve DBS to improve long-term stimulation in a wider patient population, limit side-effects, and extend battery life. Currently DBS operates in ‘open-loop’, with stimulus parameters empirically set. Closed-loop DBS, which adjusts parameters based on the state of the system, has the potential to overcome current limitations to increase therapeutic efficacy while reducing side-effects, costs and energy. Several key questions need to be addressed before closed loop DBS can be implemented clinically.
This research will develop a new multiscale model of the neuromuscular system for closed-loop DBS. The model will simulate neural sensing and stimulation on a scale not previously considered, encompassing the electric field around the electrode, the effect on individual neurons and neural networks, and generation of muscle force. This will involve integration across multiple temporal and spatial scales, in a complex system with incomplete knowledge of system variables. Experiments will be conducted to validate the model, and identify new biomarkers of neural activity that can used with signals from the brain to enable continuous symptom monitoring. The model will be used to design a new control strategy for closed-loop DBS that can accommodate the nonlinear nature of the system, and short- and long-term changes in system behavior.
Though challenging, this research will provide new insights into the changes that take place in PD and the mechanisms by which DBS exerts its therapeutic influence. This knowledge will be used to design a new strategy for closed-loop DBS, ready for testing in patients, with the potential to significantly improve patient outcomes in PD and fundamentally change the way in which implanted devices utilise electrical stimulation to modulate neural activity.
Max ERC Funding
1 999 474 €
Duration
Start date: 2015-08-01, End date: 2020-07-31
Project acronym DC_Nutrient
Project Investigating nutrients as key determinants of DC-induced CD8 T cell responses
Researcher (PI) David FINLAY
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), LS6, ERC-2017-COG
Summary A new immunoregulatory axis has emerged in recent years demonstrating that cellular metabolism is crucial in controlling immune responses. This regulatory axis is acutely sensitive to nutrients that fuel metabolic pathways and support nutrient sensitive signalling pathways. My recent research demonstrates that nutrients are dynamically controlled and are not equally available to all immune cells. The data shows that activated T cells, clustered around a dendritic cell (DC), can consume the available nutrients, leaving the DC nutrient deprived in vitro. This local regulation of the DC nutrient microenvironment by neighbouring cells has profound effects on DC function and T cell responses. Nutrient deprived DC have altered signalling (decreased mTORC1 activity), increased pro-inflammatory functions (IL12 and costimulatory molecule expression) and induce enhanced T cell responses (proliferation, IFNγ production). However, proving this, particularly in vivo, is a major challenge as the tools to investigate nutrient dynamics within complex microenvironments have not yet been developed. This research programme will generate innovative new technologies to measure the local distribution of glucose, glutamine and leucine (all of which control mTORC1 signalling) to be visualised and quantified. These technologies will pioneer a new era of in vivo nutrient analysis. Nutrient deprivation of antigen presenting DC will then be investigated (using our new technologies) in response to various stimuli within the inflammatory lymph node and correlated to CD8 T cell responses. We will generate state-of-the-art transgenic mice to specifically knock-down nutrient transporters for glucose, glutamine, or leucine in DC to definitively prove that the availability of these nutrients to antigen presenting DC is a key mechanism for controlling CD8 T cells responses. This would be a paradigm shifting discovery that would open new horizons for the study of nutrient-regulated immune responses.
Summary
A new immunoregulatory axis has emerged in recent years demonstrating that cellular metabolism is crucial in controlling immune responses. This regulatory axis is acutely sensitive to nutrients that fuel metabolic pathways and support nutrient sensitive signalling pathways. My recent research demonstrates that nutrients are dynamically controlled and are not equally available to all immune cells. The data shows that activated T cells, clustered around a dendritic cell (DC), can consume the available nutrients, leaving the DC nutrient deprived in vitro. This local regulation of the DC nutrient microenvironment by neighbouring cells has profound effects on DC function and T cell responses. Nutrient deprived DC have altered signalling (decreased mTORC1 activity), increased pro-inflammatory functions (IL12 and costimulatory molecule expression) and induce enhanced T cell responses (proliferation, IFNγ production). However, proving this, particularly in vivo, is a major challenge as the tools to investigate nutrient dynamics within complex microenvironments have not yet been developed. This research programme will generate innovative new technologies to measure the local distribution of glucose, glutamine and leucine (all of which control mTORC1 signalling) to be visualised and quantified. These technologies will pioneer a new era of in vivo nutrient analysis. Nutrient deprivation of antigen presenting DC will then be investigated (using our new technologies) in response to various stimuli within the inflammatory lymph node and correlated to CD8 T cell responses. We will generate state-of-the-art transgenic mice to specifically knock-down nutrient transporters for glucose, glutamine, or leucine in DC to definitively prove that the availability of these nutrients to antigen presenting DC is a key mechanism for controlling CD8 T cells responses. This would be a paradigm shifting discovery that would open new horizons for the study of nutrient-regulated immune responses.
Max ERC Funding
1 995 861 €
Duration
Start date: 2018-05-01, End date: 2023-04-30
Project acronym DeFiNER
Project Nucleotide Excision Repair: Decoding its Functional Role in Mammals
Researcher (PI) Georgios Garinis
Host Institution (HI) IDRYMA TECHNOLOGIAS KAI EREVNAS
Call Details Consolidator Grant (CoG), LS4, ERC-2014-CoG
Summary Genome maintenance, chromatin remodelling and transcription are tightly linked biological processes that are currently poorly understood and vastly unexplored. Nucleotide excision repair (NER) is a major DNA repair pathway that mammalian cells employ to maintain their genome intact and faithfully transmit it into their progeny. Besides cancer and aging, however, defects in NER give rise to developmental disorders whose clinical heterogeneity and varying severity can only insufficiently be explained by the DNA repair defect. Recent work reveals that NER factors play a role, in addition to DNA repair, in transcription and the three-dimensional organization of our genome. Indeed, NER factors are now known to function in the regulation of gene expression, the transcriptional reprogramming of pluripotent stem cells and the fine-tuning of growth hormones during mammalian development. In this regard, the non-random organization of our genome, chromatin and the process of transcription itself are expected to play paramount roles in how NER factors coordinate, prioritize and execute their distinct tasks during development and disease progression. At present, however, no solid evidence exists as to how NER is functionally involved in such complex processes, what are the NER-associated protein complexes and underlying gene networks or how NER factors operate within the complex chromatin architecture. This is primarily due to our difficulties in dissecting the diverse functional contributions of NER proteins in an intact organism. Here, we propose to use a unique series of knock-in, transgenic and NER progeroid mice to decode the functional role of NER in mammals, thus paving the way for understanding how genome maintenance pathways are connected to developmental defects and disease mechanisms in vivo.
Summary
Genome maintenance, chromatin remodelling and transcription are tightly linked biological processes that are currently poorly understood and vastly unexplored. Nucleotide excision repair (NER) is a major DNA repair pathway that mammalian cells employ to maintain their genome intact and faithfully transmit it into their progeny. Besides cancer and aging, however, defects in NER give rise to developmental disorders whose clinical heterogeneity and varying severity can only insufficiently be explained by the DNA repair defect. Recent work reveals that NER factors play a role, in addition to DNA repair, in transcription and the three-dimensional organization of our genome. Indeed, NER factors are now known to function in the regulation of gene expression, the transcriptional reprogramming of pluripotent stem cells and the fine-tuning of growth hormones during mammalian development. In this regard, the non-random organization of our genome, chromatin and the process of transcription itself are expected to play paramount roles in how NER factors coordinate, prioritize and execute their distinct tasks during development and disease progression. At present, however, no solid evidence exists as to how NER is functionally involved in such complex processes, what are the NER-associated protein complexes and underlying gene networks or how NER factors operate within the complex chromatin architecture. This is primarily due to our difficulties in dissecting the diverse functional contributions of NER proteins in an intact organism. Here, we propose to use a unique series of knock-in, transgenic and NER progeroid mice to decode the functional role of NER in mammals, thus paving the way for understanding how genome maintenance pathways are connected to developmental defects and disease mechanisms in vivo.
Max ERC Funding
1 995 000 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym dEMORY
Project Dissecting the Role of Dendrites in Memory
Researcher (PI) Panayiota Poirazi
Host Institution (HI) FOUNDATION FOR RESEARCH AND TECHNOLOGY HELLAS
Call Details Starting Grant (StG), LS5, ERC-2012-StG_20111109
Summary Understanding the rules and mechanisms underlying memory formation, storage and retrieval is a grand challenge in neuroscience. In light of cumulating evidence regarding non-linear dendritic events (dendritic-spikes, branch strength potentiation, temporal sequence detection etc) together with activity-dependent rewiring of the connection matrix, the classical notion of information storage via Hebbian-like changes in synaptic connections is inadequate. While more recent plasticity theories consider non-linear dendritic properties, a unifying theory of how dendrites are utilized to achieve memory coding, storing and/or retrieval is cruelly missing. Using computational models, we will simulate memory processes in three key brain regions: the hippocampus, the amygdala and the prefrontal cortex. Models will incorporate biologically constrained dendrites and state-of-the-art plasticity rules and will span different levels of abstraction, ranging from detailed biophysical single neurons and circuits to integrate-and-fire networks and abstract theoretical models. Our main goal is to dissect the role of dendrites in information processing and storage across the three different regions by systematically altering their anatomical, biophysical and plasticity properties. Findings will further our understanding of the fundamental computations supported by these structures and how these computations, reinforced by plasticity mechanisms, sub-serve memory formation and associated dysfunctions, thus opening new avenues for hypothesis driven experimentation and development of novel treatments for memory-related diseases. Identification of dendrites as the key processing units across brain regions and complexity levels will lay the foundations for a new era in computational and experimental neuroscience and serve as the basis for groundbreaking advances in the robotics and artificial intelligence fields while also having a large impact on the machine learning community.
Summary
Understanding the rules and mechanisms underlying memory formation, storage and retrieval is a grand challenge in neuroscience. In light of cumulating evidence regarding non-linear dendritic events (dendritic-spikes, branch strength potentiation, temporal sequence detection etc) together with activity-dependent rewiring of the connection matrix, the classical notion of information storage via Hebbian-like changes in synaptic connections is inadequate. While more recent plasticity theories consider non-linear dendritic properties, a unifying theory of how dendrites are utilized to achieve memory coding, storing and/or retrieval is cruelly missing. Using computational models, we will simulate memory processes in three key brain regions: the hippocampus, the amygdala and the prefrontal cortex. Models will incorporate biologically constrained dendrites and state-of-the-art plasticity rules and will span different levels of abstraction, ranging from detailed biophysical single neurons and circuits to integrate-and-fire networks and abstract theoretical models. Our main goal is to dissect the role of dendrites in information processing and storage across the three different regions by systematically altering their anatomical, biophysical and plasticity properties. Findings will further our understanding of the fundamental computations supported by these structures and how these computations, reinforced by plasticity mechanisms, sub-serve memory formation and associated dysfunctions, thus opening new avenues for hypothesis driven experimentation and development of novel treatments for memory-related diseases. Identification of dendrites as the key processing units across brain regions and complexity levels will lay the foundations for a new era in computational and experimental neuroscience and serve as the basis for groundbreaking advances in the robotics and artificial intelligence fields while also having a large impact on the machine learning community.
Max ERC Funding
1 398 000 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
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 DYNACOM
Project From Genome Integrity to Genome Plasticity:
Dynamic Complexes Controlling Once per Cell Cycle Replication
Researcher (PI) Zoi Lygerou
Host Institution (HI) PANEPISTIMIO PATRON
Call Details Starting Grant (StG), LS3, ERC-2011-StG_20101109
Summary Accurate genome duplication is controlled by multi-subunit protein complexes which associate with chromatin and dictate when and where replication should take place. Dynamic changes in these complexes lie at the heart of their ability to ensure the maintenance of genomic integrity. Defects in origin bound complexes lead to re-replication of the genome across evolution, have been linked to DNA-replication stress and may predispose for gene amplification events. Such genomic aberrations are central to malignant transformation.
We wish to understand how once per cell cycle replication is normally controlled within the context of the living cell and how defects in this control may result in loss of genome integrity and provide genome plasticity. To this end, live cell imaging in human cells in culture will be combined with genetic studies in fission yeast and modelling and in silico analysis.
The proposed research aims to:
1. Decipher the regulatory mechanisms which act in time and space to ensure once per cell cycle replication within living cells and how they may be affected by system aberrations, using functional live cell imaging.
2. Test whether aberrations in the licensing system may provide a selective advantage, through amplification of multiple genomic loci. To this end, a natural selection experiment will be set up in fission yeast .
3. Investigate how rereplication takes place along the genome in single cells. Is there heterogeneity amongst a population, leading to a plethora of different genotypes? In silico analysis of full genome DNA rereplication will be combined to single cell analysis in fission yeast.
4. Assess the relevance of our findings for gene amplification events in cancer. Does ectopic expression of human Cdt1/Cdc6 in cancer cells enhance drug resistance through gene amplification?
Our findings are expected to offer novel insight into mechanisms underlying cancer development and progression.
Summary
Accurate genome duplication is controlled by multi-subunit protein complexes which associate with chromatin and dictate when and where replication should take place. Dynamic changes in these complexes lie at the heart of their ability to ensure the maintenance of genomic integrity. Defects in origin bound complexes lead to re-replication of the genome across evolution, have been linked to DNA-replication stress and may predispose for gene amplification events. Such genomic aberrations are central to malignant transformation.
We wish to understand how once per cell cycle replication is normally controlled within the context of the living cell and how defects in this control may result in loss of genome integrity and provide genome plasticity. To this end, live cell imaging in human cells in culture will be combined with genetic studies in fission yeast and modelling and in silico analysis.
The proposed research aims to:
1. Decipher the regulatory mechanisms which act in time and space to ensure once per cell cycle replication within living cells and how they may be affected by system aberrations, using functional live cell imaging.
2. Test whether aberrations in the licensing system may provide a selective advantage, through amplification of multiple genomic loci. To this end, a natural selection experiment will be set up in fission yeast .
3. Investigate how rereplication takes place along the genome in single cells. Is there heterogeneity amongst a population, leading to a plethora of different genotypes? In silico analysis of full genome DNA rereplication will be combined to single cell analysis in fission yeast.
4. Assess the relevance of our findings for gene amplification events in cancer. Does ectopic expression of human Cdt1/Cdc6 in cancer cells enhance drug resistance through gene amplification?
Our findings are expected to offer novel insight into mechanisms underlying cancer development and progression.
Max ERC Funding
1 531 000 €
Duration
Start date: 2012-02-01, End date: 2017-01-31
Project acronym EASY
Project Ejection Accretion Structures in YSOs (EASY)
Researcher (PI) Thomas RAY
Host Institution (HI) DUBLIN INSTITUTE FOR ADVANCED STUDIES
Call Details Advanced Grant (AdG), PE9, ERC-2016-ADG
Summary For a number of reasons, in particular their proximity and the abundant range of diagnostics to determine their characteristics, outflows from young stellar objects (YSOs) offer us the best opportunity of discovering how astrophysical jets are generated and the nature of the link between outflows and their accretion disks. Models predict that the jet is initially launched from within 0.1 to a few au of the star and focused on scales at most ten times larger. Thus, even for the nearest star formation region, we need high spatial resolution to image the “central engine” and test current models.
With these ideas in mind, and the availability of a whole new set of observational and computational resources, it is proposed to investigate the origin of YSO jets, and the jet/accretion zone link, using a number of highly novel approaches to test magneto-hydrodynamic (MHD) models including:
(a) Near-infrared interferometry to determine the spatial distribution and kinematics of the outflow as it is launched as a way of discriminating between competing models.
(b) A multi-epoch study of the strength and configuration of the magnetic field of the parent star to see whether model values and geometries agree with observations and the nature of its variability.
(c) Examining, through high spatial resolution radio observations, how the ionized component of these jets are collimated very close to the source and how shocks in the flow can give rise to low energy cosmic rays.
(d) Use the James Webb Space Telescope (JWST) and, in particular, the Mid-Infrared Instrument (MIRI) and Near-Infrared Spectrograph (NIRSpec) to investigate with high spatial resolution atomic jets from protostars that are still acquiring most of their mass. In addition, we will study how accretion is affected by metallicity by studying young solar-like stars in the low metallicity Magellanic Clouds.
In all cases the required observational campaigns have been approved.
Summary
For a number of reasons, in particular their proximity and the abundant range of diagnostics to determine their characteristics, outflows from young stellar objects (YSOs) offer us the best opportunity of discovering how astrophysical jets are generated and the nature of the link between outflows and their accretion disks. Models predict that the jet is initially launched from within 0.1 to a few au of the star and focused on scales at most ten times larger. Thus, even for the nearest star formation region, we need high spatial resolution to image the “central engine” and test current models.
With these ideas in mind, and the availability of a whole new set of observational and computational resources, it is proposed to investigate the origin of YSO jets, and the jet/accretion zone link, using a number of highly novel approaches to test magneto-hydrodynamic (MHD) models including:
(a) Near-infrared interferometry to determine the spatial distribution and kinematics of the outflow as it is launched as a way of discriminating between competing models.
(b) A multi-epoch study of the strength and configuration of the magnetic field of the parent star to see whether model values and geometries agree with observations and the nature of its variability.
(c) Examining, through high spatial resolution radio observations, how the ionized component of these jets are collimated very close to the source and how shocks in the flow can give rise to low energy cosmic rays.
(d) Use the James Webb Space Telescope (JWST) and, in particular, the Mid-Infrared Instrument (MIRI) and Near-Infrared Spectrograph (NIRSpec) to investigate with high spatial resolution atomic jets from protostars that are still acquiring most of their mass. In addition, we will study how accretion is affected by metallicity by studying young solar-like stars in the low metallicity Magellanic Clouds.
In all cases the required observational campaigns have been approved.
Max ERC Funding
1 853 090 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
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 EyeRegen
Project Engineering a scaffold based therapy for corneal regeneration
Researcher (PI) Mark Joseph Ahearne
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), PE8, ERC-2014-STG
Summary Corneal blindness resulting from disease, physical injury or chemical burns affects millions worldwide and has a considerable economic and social impact on the lives of people across Europe. In many cases corneal transplants can restore vision however the shortage of donor corneas suitable for transplantation has necessitated the development of alternative treatments. The aim of this project is to develop a new approach to corneal tissue regeneration. Previous approaches at engineering corneal tissue have required access to donor cells and lengthy culture periods in an attempt to grow tissue in vitro prior to implantation with only limited success and at great expense. Our approach will differ fundamentally from these in that we will design artificial corneal scaffolds that do not require donated cells or in vitro culture but instead will recruit the patient’s own cells to regenerate the cornea post-implantation. These biomaterial scaffolds will incorporate specific chemical and physical cues with the deliberate aim of attracting cells and inducing tissue formation. Studies will be undertaken to examine how different chemical, biochemical, physical and mechanical cues can be used to control the behaviour of corneal epithelial, stromal and endothelial cells. Once the optimal combination of these cues has been determined, this information will be incorporated into the design of the scaffold. Recent advances in manufacturing and material processing technology will enable us to develop scaffolds with organized nanometric architectures and that incorporate controlled growth factor release mechanisms. Techniques such as 3D bio-printing and nanofiber electrospinning will be used to fabricate scaffolds. The ability of the scaffold to attract cells and promote matrix remodelling will be examined by developing an in vitro bioreactor system capable of mimicking the ocular environment and by performing in vivo tests using a live animal model.
Summary
Corneal blindness resulting from disease, physical injury or chemical burns affects millions worldwide and has a considerable economic and social impact on the lives of people across Europe. In many cases corneal transplants can restore vision however the shortage of donor corneas suitable for transplantation has necessitated the development of alternative treatments. The aim of this project is to develop a new approach to corneal tissue regeneration. Previous approaches at engineering corneal tissue have required access to donor cells and lengthy culture periods in an attempt to grow tissue in vitro prior to implantation with only limited success and at great expense. Our approach will differ fundamentally from these in that we will design artificial corneal scaffolds that do not require donated cells or in vitro culture but instead will recruit the patient’s own cells to regenerate the cornea post-implantation. These biomaterial scaffolds will incorporate specific chemical and physical cues with the deliberate aim of attracting cells and inducing tissue formation. Studies will be undertaken to examine how different chemical, biochemical, physical and mechanical cues can be used to control the behaviour of corneal epithelial, stromal and endothelial cells. Once the optimal combination of these cues has been determined, this information will be incorporated into the design of the scaffold. Recent advances in manufacturing and material processing technology will enable us to develop scaffolds with organized nanometric architectures and that incorporate controlled growth factor release mechanisms. Techniques such as 3D bio-printing and nanofiber electrospinning will be used to fabricate scaffolds. The ability of the scaffold to attract cells and promote matrix remodelling will be examined by developing an in vitro bioreactor system capable of mimicking the ocular environment and by performing in vivo tests using a live animal model.
Max ERC Funding
1 498 734 €
Duration
Start date: 2015-07-01, End date: 2020-06-30
Project acronym FastBio
Project A genomics and systems biology approach to explore the molecular signature and functional consequences of long-term, structured fasting in humans
Researcher (PI) Antigoni DIMA
Host Institution (HI) BIOMEDICAL SCIENCES RESEARCH CENTER ALEXANDER FLEMING
Call Details Starting Grant (StG), LS2, ERC-2016-STG
Summary Dietary intake has an enormous impact on aspects of human health, yet scientific consensus about how what we eat affects our biology remains elusive. To address the complex biological impact of diet, I propose to apply an unconventional, ‘humans-as-model-organisms’ approach to compare the molecular and functional effects of a highly structured dietary regime, specified by the Eastern Orthodox Christian Church (EOCC), to the unstructured diet followed by the general population. Individuals who follow the EOCC regime abstain from meat, dairy products and eggs for 180-200 days annually, in a temporally-structured manner initiated in childhood. I aim to explore the biological signatures of structured vs. unstructured diet by addressing three objectives. First I will investigate the effects of the two regimes, and of genetic variation, on higher-level phenotypes including anthropometric, physiological and biomarker traits. Second, I will carry out a comprehensive set of omics assays (metabolomics, transcriptomics, epigenomics and investigation of the gut microbiome), will associate omics phenotypes with genetic variation, and will integrate data across biological levels to uncover complex molecular signatures. Third, I will interrogate the functional consequences of dietary regimes at the cellular level through primary cell culture. Acute and long-term effects of dietary intake will be explored for all objectives through a two timepoint sampling strategy. This proposal therefore comprises a unique opportunity to study a specific perturbation (EOCC structured diet) introduced to a steady-state system (unstructured diet followed by the general population) in a ground-breaking human systems biology type of study. This approach brings together expertise from genomics, computational biology, statistics, medicine and epidemiology. It will lead to novel insights regarding the potent signalling nature of nutrients and is likely to yield results of high translational value.
Summary
Dietary intake has an enormous impact on aspects of human health, yet scientific consensus about how what we eat affects our biology remains elusive. To address the complex biological impact of diet, I propose to apply an unconventional, ‘humans-as-model-organisms’ approach to compare the molecular and functional effects of a highly structured dietary regime, specified by the Eastern Orthodox Christian Church (EOCC), to the unstructured diet followed by the general population. Individuals who follow the EOCC regime abstain from meat, dairy products and eggs for 180-200 days annually, in a temporally-structured manner initiated in childhood. I aim to explore the biological signatures of structured vs. unstructured diet by addressing three objectives. First I will investigate the effects of the two regimes, and of genetic variation, on higher-level phenotypes including anthropometric, physiological and biomarker traits. Second, I will carry out a comprehensive set of omics assays (metabolomics, transcriptomics, epigenomics and investigation of the gut microbiome), will associate omics phenotypes with genetic variation, and will integrate data across biological levels to uncover complex molecular signatures. Third, I will interrogate the functional consequences of dietary regimes at the cellular level through primary cell culture. Acute and long-term effects of dietary intake will be explored for all objectives through a two timepoint sampling strategy. This proposal therefore comprises a unique opportunity to study a specific perturbation (EOCC structured diet) introduced to a steady-state system (unstructured diet followed by the general population) in a ground-breaking human systems biology type of study. This approach brings together expertise from genomics, computational biology, statistics, medicine and epidemiology. It will lead to novel insights regarding the potent signalling nature of nutrients and is likely to yield results of high translational value.
Max ERC Funding
1 500 000 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym FAT NKT
Project Targeting iNKT cell and adipocyte crosstalk for control of metabolism and body weight
Researcher (PI) Lydia Lynch
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), LS6, ERC-2015-STG
Summary Obesity has reached epidemic proportions globally. At least 2.8 million people die each year as a result of being overweight or obese, the biggest burden being obesity-related diseases. It is now clear that inflammation is an underlying cause or contributor to many of these diseases, including type 2 diabetes, atherosclerosis, and cancer. Recognition that the immune system can regulate metabolic pathways has prompted a new way of thinking about diabetes and weight management. Despite much recent progress, most immunometabolic pathways, and how to target them, are currently unknown. One such pathway is the cross-talk between invariant natural killer (iNKT) cells and neighboring adipocytes. iNKT cells are the innate lipid-sensing arm of the immune system. Since our discovery that mammalian adipose tissue is enriched for iNKT cells, we have identified a critical role for iNKT cells in regulating adipose inflammation and body weight. The goal of this project is to use a multi-disciplinary approach to identify key signals and molecules used by iNKT cells to induce metabolic control and weight loss in obesity. Using immunological assays and multi-photon intravital microscopy, cells and pathways that control the unique regulatory functions of adipose iNKT cells will be identified and characterised. Novel lipid antigens in adipose tissue will be identified using a biochemical approach, perhaps explaining iNKT cell conservation in adipose depots, and providing safe tools for iNKT cell manipulation in vivo. Finally, using proteomics and whole body metabolic analysis in vivo, novel ‘weight-loss inducing’ factors produced by adipose iNKT cells will be identified. This ambitious and high impact project has the potential to yield major insights into immunometabolic interactions at steady state and in obesity. The ability to activate or induce adipose iNKT cells holds remarkable potential as an entirely new therapeutic direction for treating obesity and type 2 diabetes.
Summary
Obesity has reached epidemic proportions globally. At least 2.8 million people die each year as a result of being overweight or obese, the biggest burden being obesity-related diseases. It is now clear that inflammation is an underlying cause or contributor to many of these diseases, including type 2 diabetes, atherosclerosis, and cancer. Recognition that the immune system can regulate metabolic pathways has prompted a new way of thinking about diabetes and weight management. Despite much recent progress, most immunometabolic pathways, and how to target them, are currently unknown. One such pathway is the cross-talk between invariant natural killer (iNKT) cells and neighboring adipocytes. iNKT cells are the innate lipid-sensing arm of the immune system. Since our discovery that mammalian adipose tissue is enriched for iNKT cells, we have identified a critical role for iNKT cells in regulating adipose inflammation and body weight. The goal of this project is to use a multi-disciplinary approach to identify key signals and molecules used by iNKT cells to induce metabolic control and weight loss in obesity. Using immunological assays and multi-photon intravital microscopy, cells and pathways that control the unique regulatory functions of adipose iNKT cells will be identified and characterised. Novel lipid antigens in adipose tissue will be identified using a biochemical approach, perhaps explaining iNKT cell conservation in adipose depots, and providing safe tools for iNKT cell manipulation in vivo. Finally, using proteomics and whole body metabolic analysis in vivo, novel ‘weight-loss inducing’ factors produced by adipose iNKT cells will be identified. This ambitious and high impact project has the potential to yield major insights into immunometabolic interactions at steady state and in obesity. The ability to activate or induce adipose iNKT cells holds remarkable potential as an entirely new therapeutic direction for treating obesity and type 2 diabetes.
Max ERC Funding
1 804 052 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym FibreRemodel
Project Frontier research in arterial fibre remodelling for vascular disease diagnosis and tissue engineering
Researcher (PI) Caitriona Lally
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), PE8, ERC-2014-STG
Summary Each year cardiovascular diseases such as atherosclerosis and aneurysms cause 48% of all deaths in Europe. Arteries may be regarded as fibre-reinforced materials, with the stiffer collagen fibres present in the arterial wall bearing most of the load during pressurisation. Degenerative vascular diseases such as atherosclerosis and aneurysms alter the macroscopic mechanical properties of arterial tissue and therefore change the arterial wall composition and the quality and orientation of the underlying fibrous architecture. Information on the complex fibre architecture of arterial tissues is therefore at the core of understanding the aetiology of vascular diseases. The current proposal aims to use a combination of in vivo Diffusion Tensor Magnetic Resonance Imaging, with parallel in silico modelling, to non-invasively identify differences in the fibre architecture of human carotid arteries which can be directly linked with carotid artery disease and hence used to diagnose vulnerable plaque rupture risk.
Knowledge of arterial fibre patterns, and how these fibres alter in response to their mechanical environment, also provides a means of understanding remodelling of tissue engineered vessels. Therefore, in the second phase of this project, this novel imaging framework will be used to determine fibre patterns of decellularised arterial constructs in vitro with a view to directing mesenchymal stem cell growth and differentiation and creating a biologically and mechanically compatible tissue engineered vessel. In silico mechanobiological models will also be used to help identify the optimum loading environment for the vessels to encourage cell repopulation but prevent excessive intimal hyperplasia.
This combination of novel in vivo, in vitro and in silico work has the potential to revolutionise approaches to early diagnosis of vascular diseases and vascular tissue engineering strategies.
Summary
Each year cardiovascular diseases such as atherosclerosis and aneurysms cause 48% of all deaths in Europe. Arteries may be regarded as fibre-reinforced materials, with the stiffer collagen fibres present in the arterial wall bearing most of the load during pressurisation. Degenerative vascular diseases such as atherosclerosis and aneurysms alter the macroscopic mechanical properties of arterial tissue and therefore change the arterial wall composition and the quality and orientation of the underlying fibrous architecture. Information on the complex fibre architecture of arterial tissues is therefore at the core of understanding the aetiology of vascular diseases. The current proposal aims to use a combination of in vivo Diffusion Tensor Magnetic Resonance Imaging, with parallel in silico modelling, to non-invasively identify differences in the fibre architecture of human carotid arteries which can be directly linked with carotid artery disease and hence used to diagnose vulnerable plaque rupture risk.
Knowledge of arterial fibre patterns, and how these fibres alter in response to their mechanical environment, also provides a means of understanding remodelling of tissue engineered vessels. Therefore, in the second phase of this project, this novel imaging framework will be used to determine fibre patterns of decellularised arterial constructs in vitro with a view to directing mesenchymal stem cell growth and differentiation and creating a biologically and mechanically compatible tissue engineered vessel. In silico mechanobiological models will also be used to help identify the optimum loading environment for the vessels to encourage cell repopulation but prevent excessive intimal hyperplasia.
This combination of novel in vivo, in vitro and in silico work has the potential to revolutionise approaches to early diagnosis of vascular diseases and vascular tissue engineering strategies.
Max ERC Funding
1 521 875 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym HA-NFKB-VILI
Project Hypercapnic Acidosis and NF-kB in Ventilator Induced Lung Injury: Developing strategies to minimize lung injury and facilitate repair
Researcher (PI) John Laffey
Host Institution (HI) NATIONAL UNIVERSITY OF IRELAND GALWAY
Call Details Starting Grant (StG), LS6, ERC-2007-StG
Summary Acute Respiratory Distress Syndrome and Acute Lung Injury [ALI/ARDS] are devastating diseases, causing over 20,000 deaths annually in the US. Mechanical ventilation may worsen ALI/ARDS, a process termed Ventilator Induced Lung Injury [VILI]. Hypercapnic acidosis (HA) is a central component of lung ventilatory strategies to minimize VILI, and is a potent biologic agent, exerting a myriad of effects on diverse biologic pathways. Deliberately induced HA is protective in multiple lung injury models. However, HA may inhibit the host response to bacterial sepsis. Furthermore, HA may retard the repair process and slow recovery following ALI/ARDS. Hence, the diverse biologic actions of HA may result in net beneficial – or deleterious – effects depending on the specific context. An alternative approach is to manipulate a single key effector pathway, central to the protective effects of HA, which would also be effective in patients in whom hypercapnia is contra-indicated. Hypercapnia attenuates NF-kB activation, and may exert its effects – both beneficial and deleterious – via this mechanism. NF-kB is a pivotal regulator of the pro-inflammatory response, but is also a key epithelial cytoprotectant. Selective modulation of the NF-kB pathway, at the pulmonary epithelial surface, may accentuate the beneficial effects of HA on injury but minimize the potential for delayed tissue repair. We will investigate the contribution of NF-kB to the effects of HA, and characterize the direct effects modulation of NF-kB, in both in vitro and preclinical models of lung injury and repair. We will utilize pulmonary gene therapy, which facilitates delivery of high quantities of the therapeutic agent directly to the injury site, to maximize the potential for therapeutic benefit. These studies will provide novel insights into: key pathways contributing to lung injury and to repair; the role of HA and NF-kB in these processes; and the potential of pulmonary gene therapy in ALI/ARDS.
Summary
Acute Respiratory Distress Syndrome and Acute Lung Injury [ALI/ARDS] are devastating diseases, causing over 20,000 deaths annually in the US. Mechanical ventilation may worsen ALI/ARDS, a process termed Ventilator Induced Lung Injury [VILI]. Hypercapnic acidosis (HA) is a central component of lung ventilatory strategies to minimize VILI, and is a potent biologic agent, exerting a myriad of effects on diverse biologic pathways. Deliberately induced HA is protective in multiple lung injury models. However, HA may inhibit the host response to bacterial sepsis. Furthermore, HA may retard the repair process and slow recovery following ALI/ARDS. Hence, the diverse biologic actions of HA may result in net beneficial – or deleterious – effects depending on the specific context. An alternative approach is to manipulate a single key effector pathway, central to the protective effects of HA, which would also be effective in patients in whom hypercapnia is contra-indicated. Hypercapnia attenuates NF-kB activation, and may exert its effects – both beneficial and deleterious – via this mechanism. NF-kB is a pivotal regulator of the pro-inflammatory response, but is also a key epithelial cytoprotectant. Selective modulation of the NF-kB pathway, at the pulmonary epithelial surface, may accentuate the beneficial effects of HA on injury but minimize the potential for delayed tissue repair. We will investigate the contribution of NF-kB to the effects of HA, and characterize the direct effects modulation of NF-kB, in both in vitro and preclinical models of lung injury and repair. We will utilize pulmonary gene therapy, which facilitates delivery of high quantities of the therapeutic agent directly to the injury site, to maximize the potential for therapeutic benefit. These studies will provide novel insights into: key pathways contributing to lung injury and to repair; the role of HA and NF-kB in these processes; and the potential of pulmonary gene therapy in ALI/ARDS.
Max ERC Funding
1 052 556 €
Duration
Start date: 2009-01-01, End date: 2013-12-31
Project acronym HIGHWAVE
Project Breaking of highly energetic waves
Researcher (PI) Frederic DIAS
Host Institution (HI) UNIVERSITY COLLEGE DUBLIN, NATIONAL UNIVERSITY OF IRELAND, DUBLIN
Call Details Advanced Grant (AdG), PE8, ERC-2018-ADG
Summary HIGHWAVE is an interdisciplinary project at the frontiers of coastal/ocean engineering, earth system science, statistics and fluid mechanics that will explore fundamental open questions in wave breaking. Why do waves break, how do they dissipate energy and why is this important? A central element of the work builds on recent international developments in the field of wave breaking and wave run-up led by the PI that have provided the first universal criterion for predicting the onset of breaking of water waves in uniform water depths from deep to intermediate. This work has also shown that the run-up of nonlinear waves impinging on a vertical wall can exceed up to 12 times the far-field amplitude of the incoming waves. These results have now opened up the possibility for more accurate operational wave models. They have practical and economic benefits in determining structural loads on ships and coastal/offshore infrastructure, evaluating seabed response to extreme waves, and optimizing operational strategies for maritime and marine renewable energy enterprises. This is a tremendous advance comparable to the introduction of wave prediction during World War II, and the PI aims to be at the forefront of this research effort to take research in wave breaking into fundamentally new directions. The objectives of the project are: (i) to develop an innovative approach to include accurate wave breaking physics into coupled sea state and ocean weather forecasting models; (ii) to obtain improved criteria for the design of ships and coastal/offshore infrastructure; (iii) to quantify erosion by powerful breaking waves, and (iv) to develop new concepts in wave measurement with improved characterization of wave breaking using real-time instrumentation. This highly interdisciplinary project will involve an ambitious and unconventional combination of computational simulation/theory, laboratory experiments, and field measurements of sea waves, closely informed by application needs.
Summary
HIGHWAVE is an interdisciplinary project at the frontiers of coastal/ocean engineering, earth system science, statistics and fluid mechanics that will explore fundamental open questions in wave breaking. Why do waves break, how do they dissipate energy and why is this important? A central element of the work builds on recent international developments in the field of wave breaking and wave run-up led by the PI that have provided the first universal criterion for predicting the onset of breaking of water waves in uniform water depths from deep to intermediate. This work has also shown that the run-up of nonlinear waves impinging on a vertical wall can exceed up to 12 times the far-field amplitude of the incoming waves. These results have now opened up the possibility for more accurate operational wave models. They have practical and economic benefits in determining structural loads on ships and coastal/offshore infrastructure, evaluating seabed response to extreme waves, and optimizing operational strategies for maritime and marine renewable energy enterprises. This is a tremendous advance comparable to the introduction of wave prediction during World War II, and the PI aims to be at the forefront of this research effort to take research in wave breaking into fundamentally new directions. The objectives of the project are: (i) to develop an innovative approach to include accurate wave breaking physics into coupled sea state and ocean weather forecasting models; (ii) to obtain improved criteria for the design of ships and coastal/offshore infrastructure; (iii) to quantify erosion by powerful breaking waves, and (iv) to develop new concepts in wave measurement with improved characterization of wave breaking using real-time instrumentation. This highly interdisciplinary project will involve an ambitious and unconventional combination of computational simulation/theory, laboratory experiments, and field measurements of sea waves, closely informed by application needs.
Max ERC Funding
2 499 946 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym HIVBIOCHIP
Project A POINT-OF-CARE BIOCHIP FOR HIV MONITORING IN THE DEVELOPING WORLD
Researcher (PI) Nikolaos Chronis
Host Institution (HI) "NATIONAL CENTER FOR SCIENTIFIC RESEARCH ""DEMOKRITOS"""
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary HIV/AIDS is one of the most destructive pandemics in human history, responsible for more than 25 million deaths. More than 30 million people live with limited or no access to therapeutic treatments, mainly due to the high cost of highly active antiretroviral therapies (HAART) and current diagnostic tests as well as due to the lack of basic infrastructure (e.g. lack of electricity, no trained personnel) that can support these tests. The need for innovative, inexpensive diagnostic instrumentation technology that can be used in resource-limited settings is immediate.
While programs that offer free HAART are being implemented in resource-limited settings, no diagnostic tests are available for evaluating the efficacy of HAART provided for the reasons mentioned above. Efficient management of HAART requires monitoring the course of HIV infection over time. The World Health Organization (WHO) recommends the CD4 T-cell count test for monitoring the clinical status of HIV individuals in resource-limited settings.
We propose to develop a portable, inexpensive, MEMS (MicroElectroMechanical Systems)-based, imaging system for counting the absolute number of CD4 cells from 1 l of whole blood. We use the term ‘imaging system’ to denote the different approach we follow for counting CD4 cells: rather the reading one by one singles cells (as it is done with flow cytometry), our system can image simultaneously thousands of individual cells, pre-assembled on the surface of a biochip. Although the proposed imaging system can replace current expensive cell counting instrumentation, our goal is to develop a system that can reach the end-user wherever limited infrastructure is present and no access to a hospital or clinic is possible. Such technology will not only enable to monitor the efficacy of an individual’s HAART in the developing world, but it will make more medicines available by identifying patients who need a treatment from patients who do not need it.
Summary
HIV/AIDS is one of the most destructive pandemics in human history, responsible for more than 25 million deaths. More than 30 million people live with limited or no access to therapeutic treatments, mainly due to the high cost of highly active antiretroviral therapies (HAART) and current diagnostic tests as well as due to the lack of basic infrastructure (e.g. lack of electricity, no trained personnel) that can support these tests. The need for innovative, inexpensive diagnostic instrumentation technology that can be used in resource-limited settings is immediate.
While programs that offer free HAART are being implemented in resource-limited settings, no diagnostic tests are available for evaluating the efficacy of HAART provided for the reasons mentioned above. Efficient management of HAART requires monitoring the course of HIV infection over time. The World Health Organization (WHO) recommends the CD4 T-cell count test for monitoring the clinical status of HIV individuals in resource-limited settings.
We propose to develop a portable, inexpensive, MEMS (MicroElectroMechanical Systems)-based, imaging system for counting the absolute number of CD4 cells from 1 l of whole blood. We use the term ‘imaging system’ to denote the different approach we follow for counting CD4 cells: rather the reading one by one singles cells (as it is done with flow cytometry), our system can image simultaneously thousands of individual cells, pre-assembled on the surface of a biochip. Although the proposed imaging system can replace current expensive cell counting instrumentation, our goal is to develop a system that can reach the end-user wherever limited infrastructure is present and no access to a hospital or clinic is possible. Such technology will not only enable to monitor the efficacy of an individual’s HAART in the developing world, but it will make more medicines available by identifying patients who need a treatment from patients who do not need it.
Max ERC Funding
1 986 000 €
Duration
Start date: 2012-06-01, End date: 2017-05-31