Project acronym 20SComplexity
Project An integrative approach to uncover the multilevel regulation of 20S proteasome degradation
Researcher (PI) Michal Sharon
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS1, ERC-2014-STG
Summary For many years, the ubiquitin-26S proteasome degradation pathway was considered the primary route for proteasomal degradation. However, it is now becoming clear that proteins can also be targeted for degradation by a ubiquitin-independent mechanism mediated by the core 20S proteasome itself. Although initially believed to be limited to rare exceptions, degradation by the 20S proteasome is now understood to have a wide range of substrates, many of which are key regulatory proteins. Despite its importance, little is known about the mechanisms that control 20S proteasomal degradation, unlike the extensive knowledge acquired over the years concerning degradation by the 26S proteasome. Our overall aim is to reveal the multiple regulatory levels that coordinate the 20S proteasome degradation route.
To achieve this goal we will carry out a comprehensive research program characterizing three distinct levels of 20S proteasome regulation:
Intra-molecular regulation- Revealing the intrinsic molecular switch that activates the latent 20S proteasome.
Inter-molecular regulation- Identifying novel proteins that bind the 20S proteasome to regulate its activity and characterizing their mechanism of function.
Cellular regulatory networks- Unraveling the cellular cues and multiple pathways that influence 20S proteasome activity using a novel systematic and unbiased screening approach.
Our experimental strategy involves the combination of biochemical approaches with native mass spectrometry, cross-linking and fluorescence measurements, complemented by cell biology analyses and high-throughput screening. Such a multidisciplinary approach, integrating in vitro and in vivo findings, will likely provide the much needed knowledge on the 20S proteasome degradation route. When completed, we anticipate that this work will be part of a new paradigm – no longer perceiving the 20S proteasome mediated degradation as a simple and passive event but rather a tightly regulated and coordinated process.
Summary
For many years, the ubiquitin-26S proteasome degradation pathway was considered the primary route for proteasomal degradation. However, it is now becoming clear that proteins can also be targeted for degradation by a ubiquitin-independent mechanism mediated by the core 20S proteasome itself. Although initially believed to be limited to rare exceptions, degradation by the 20S proteasome is now understood to have a wide range of substrates, many of which are key regulatory proteins. Despite its importance, little is known about the mechanisms that control 20S proteasomal degradation, unlike the extensive knowledge acquired over the years concerning degradation by the 26S proteasome. Our overall aim is to reveal the multiple regulatory levels that coordinate the 20S proteasome degradation route.
To achieve this goal we will carry out a comprehensive research program characterizing three distinct levels of 20S proteasome regulation:
Intra-molecular regulation- Revealing the intrinsic molecular switch that activates the latent 20S proteasome.
Inter-molecular regulation- Identifying novel proteins that bind the 20S proteasome to regulate its activity and characterizing their mechanism of function.
Cellular regulatory networks- Unraveling the cellular cues and multiple pathways that influence 20S proteasome activity using a novel systematic and unbiased screening approach.
Our experimental strategy involves the combination of biochemical approaches with native mass spectrometry, cross-linking and fluorescence measurements, complemented by cell biology analyses and high-throughput screening. Such a multidisciplinary approach, integrating in vitro and in vivo findings, will likely provide the much needed knowledge on the 20S proteasome degradation route. When completed, we anticipate that this work will be part of a new paradigm – no longer perceiving the 20S proteasome mediated degradation as a simple and passive event but rather a tightly regulated and coordinated process.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-04-01, End date: 2020-03-31
Project acronym 2DHIBSA
Project Nanoscopic and Hierachical Materials via Living Crystallization-Driven Self-Assembly
Researcher (PI) Ian MANNERS
Host Institution (HI) UNIVERSITY OF BRISTOL
Call Details Advanced Grant (AdG), PE5, ERC-2017-ADG
Summary A key synthetic challenge of widespread interest in chemical science involves the creation of well-defined 2D functional materials that exist on a length-scale of nanometers to microns. In this ambitious 5 year proposal we aim to tackle this issue by exploiting the unique opportunities made possible by recent developments with the living crystallization-driven self-assembly (CDSA) platform. Using this solution processing approach, amphiphilic block copolymers (BCPs) with crystallizable blocks, related amphiphiles, and polymers with charged end groups will be used to predictably construct monodisperse samples of tailored, functional soft matter-based 2D nanostructures with controlled shape, size, and spatially-defined chemistries. Many of the resulting nanostructures will also offer unprecedented opportunities as precursors to materials with hierarchical structures through further solution-based “bottom-up” assembly methods. In addition to fundamental studies, the proposed work also aims to make important impact in the cutting-edge fields of liquid crystals, interface stabilization, catalysis, supramolecular polymers, and hierarchical materials.
Summary
A key synthetic challenge of widespread interest in chemical science involves the creation of well-defined 2D functional materials that exist on a length-scale of nanometers to microns. In this ambitious 5 year proposal we aim to tackle this issue by exploiting the unique opportunities made possible by recent developments with the living crystallization-driven self-assembly (CDSA) platform. Using this solution processing approach, amphiphilic block copolymers (BCPs) with crystallizable blocks, related amphiphiles, and polymers with charged end groups will be used to predictably construct monodisperse samples of tailored, functional soft matter-based 2D nanostructures with controlled shape, size, and spatially-defined chemistries. Many of the resulting nanostructures will also offer unprecedented opportunities as precursors to materials with hierarchical structures through further solution-based “bottom-up” assembly methods. In addition to fundamental studies, the proposed work also aims to make important impact in the cutting-edge fields of liquid crystals, interface stabilization, catalysis, supramolecular polymers, and hierarchical materials.
Max ERC Funding
2 499 597 €
Duration
Start date: 2018-05-01, End date: 2023-04-30
Project acronym AAMDDR
Project DNA damage response and genome stability: The role of ATM, ATR and the Mre11 complex
Researcher (PI) Vincenzo Costanzo
Host Institution (HI) CANCER RESEARCH UK LBG
Call Details Starting Grant (StG), LS1, ERC-2007-StG
Summary Chromosomal DNA is continuously subjected to exogenous and endogenous damaging insults. In the presence of DNA damage cells activate a multi-faceted checkpoint response that delays cell cycle progression and promotes DNA repair. Failures in this response lead to genomic instability, the main feature of cancer cells. Several cancer-prone human syndromes including the Ataxia teleangiectasia (A-T), the A-T Like Disorder (ATLD) and the Seckel Syndrome reflect defects in the specific genes of the DNA damage response such as ATM, MRE11 and ATR. DNA damage response pathways are poorly understood at biochemical level in vertebrate organisms. We have established a cell-free system based on Xenopus laevis egg extract to study molecular events underlying DNA damage response. This is the first in vitro system that recapitulates different aspects of the DNA damage response in vertebrates. Using this system we propose to study the biochemistry of the ATM, ATR and the Mre11 complex dependent DNA damage response. In particular we will: 1) Dissect the signal transduction pathway that senses DNA damage and promotes cell cycle arrest and DNA damage repair; 2) Analyze at molecular level the role of ATM, ATR, Mre11 in chromosomal DNA replication and mitosis during normal and stressful conditions; 3) Identify substrates of the ATM and ATR dependent DNA damage response using an innovative screening procedure.
Summary
Chromosomal DNA is continuously subjected to exogenous and endogenous damaging insults. In the presence of DNA damage cells activate a multi-faceted checkpoint response that delays cell cycle progression and promotes DNA repair. Failures in this response lead to genomic instability, the main feature of cancer cells. Several cancer-prone human syndromes including the Ataxia teleangiectasia (A-T), the A-T Like Disorder (ATLD) and the Seckel Syndrome reflect defects in the specific genes of the DNA damage response such as ATM, MRE11 and ATR. DNA damage response pathways are poorly understood at biochemical level in vertebrate organisms. We have established a cell-free system based on Xenopus laevis egg extract to study molecular events underlying DNA damage response. This is the first in vitro system that recapitulates different aspects of the DNA damage response in vertebrates. Using this system we propose to study the biochemistry of the ATM, ATR and the Mre11 complex dependent DNA damage response. In particular we will: 1) Dissect the signal transduction pathway that senses DNA damage and promotes cell cycle arrest and DNA damage repair; 2) Analyze at molecular level the role of ATM, ATR, Mre11 in chromosomal DNA replication and mitosis during normal and stressful conditions; 3) Identify substrates of the ATM and ATR dependent DNA damage response using an innovative screening procedure.
Max ERC Funding
1 000 000 €
Duration
Start date: 2008-07-01, End date: 2013-06-30
Project acronym ABATSYNAPSE
Project Evolution of Alzheimer’s Disease: From dynamics of single synapses to memory loss
Researcher (PI) Inna Slutsky
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS5, ERC-2011-StG_20101109
Summary A persistent challenge in unravelling mechanisms that regulate memory function is how to bridge the gap between inter-molecular dynamics of single proteins, activity of individual synapses and emerging properties of neuronal circuits. The prototype condition of disintegrating neuronal circuits is Alzheimer’s Disease (AD). Since the early time of Alois Alzheimer at the turn of the 20th century, scientists have been searching for a molecular entity that is in the roots of the cognitive deficits. Although diverse lines of evidence suggest that the amyloid-beta peptide (Abeta) plays a central role in synaptic dysfunctions of AD, several key questions remain unresolved. First, endogenous Abeta peptides are secreted by neurons throughout life, but their physiological functions are largely unknown. Second, experience-dependent physiological mechanisms that initiate the changes in Abeta composition in sporadic, the most frequent form of AD, are unidentified. And finally, molecular mechanisms that trigger Abeta-induced synaptic failure and memory decline remain elusive.
To target these questions, I propose to develop an integrative approach to correlate structure and function at the level of single synapses in hippocampal circuits. State-of-the-art techniques will enable the simultaneous real-time visualization of inter-molecular dynamics within signalling complexes and functional synaptic modifications. Utilizing FRET spectroscopy, high-resolution optical imaging, electrophysiology, molecular biology and biochemistry we will determine the casual relationship between ongoing neuronal activity, temporo-spatial dynamics and molecular composition of Abeta, structural rearrangements within the Abeta signalling complexes and plasticity of single synapses and whole networks. The proposed research will elucidate fundamental principles of neuronal circuits function and identify critical steps that initiate primary synaptic dysfunctions at the very early stages of sporadic AD.
Summary
A persistent challenge in unravelling mechanisms that regulate memory function is how to bridge the gap between inter-molecular dynamics of single proteins, activity of individual synapses and emerging properties of neuronal circuits. The prototype condition of disintegrating neuronal circuits is Alzheimer’s Disease (AD). Since the early time of Alois Alzheimer at the turn of the 20th century, scientists have been searching for a molecular entity that is in the roots of the cognitive deficits. Although diverse lines of evidence suggest that the amyloid-beta peptide (Abeta) plays a central role in synaptic dysfunctions of AD, several key questions remain unresolved. First, endogenous Abeta peptides are secreted by neurons throughout life, but their physiological functions are largely unknown. Second, experience-dependent physiological mechanisms that initiate the changes in Abeta composition in sporadic, the most frequent form of AD, are unidentified. And finally, molecular mechanisms that trigger Abeta-induced synaptic failure and memory decline remain elusive.
To target these questions, I propose to develop an integrative approach to correlate structure and function at the level of single synapses in hippocampal circuits. State-of-the-art techniques will enable the simultaneous real-time visualization of inter-molecular dynamics within signalling complexes and functional synaptic modifications. Utilizing FRET spectroscopy, high-resolution optical imaging, electrophysiology, molecular biology and biochemistry we will determine the casual relationship between ongoing neuronal activity, temporo-spatial dynamics and molecular composition of Abeta, structural rearrangements within the Abeta signalling complexes and plasticity of single synapses and whole networks. The proposed research will elucidate fundamental principles of neuronal circuits function and identify critical steps that initiate primary synaptic dysfunctions at the very early stages of sporadic AD.
Max ERC Funding
2 000 000 €
Duration
Start date: 2011-12-01, End date: 2017-09-30
Project acronym ABDESIGN
Project Computational design of novel protein function in antibodies
Researcher (PI) Sarel-Jacob Fleishman
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS1, ERC-2013-StG
Summary We propose to elucidate the structural design principles of naturally occurring antibody complementarity-determining regions (CDRs) and to computationally design novel antibody functions. Antibodies represent the most versatile known system for molecular recognition. Research has yielded many insights into antibody design principles and promising biotechnological and pharmaceutical applications. Still, our understanding of how CDRs encode specific loop conformations lags far behind our understanding of structure-function relationships in non-immunological scaffolds. Thus, design of antibodies from first principles has not been demonstrated. We propose a computational-experimental strategy to address this challenge. We will: (a) characterize the design principles and sequence elements that rigidify antibody CDRs. Natural antibody loops will be subjected to computational modeling, crystallography, and a combined in vitro evolution and deep-sequencing approach to isolate sequence features that rigidify loop backbones; (b) develop a novel computational-design strategy, which uses the >1000 solved structures of antibodies deposited in structure databases to realistically model CDRs and design them to recognize proteins that have not been co-crystallized with antibodies. For example, we will design novel antibodies targeting insulin, for which clinically useful diagnostics are needed. By accessing much larger sequence/structure spaces than are available to natural immune-system repertoires and experimental methods, computational antibody design could produce higher-specificity and higher-affinity binders, even to challenging targets; and (c) develop new strategies to program conformational change in CDRs, generating, e.g., the first allosteric antibodies. These will allow targeting, in principle, of any molecule, potentially revolutionizing how antibodies are generated for research and medicine, providing new insights on the design principles of protein functional sites.
Summary
We propose to elucidate the structural design principles of naturally occurring antibody complementarity-determining regions (CDRs) and to computationally design novel antibody functions. Antibodies represent the most versatile known system for molecular recognition. Research has yielded many insights into antibody design principles and promising biotechnological and pharmaceutical applications. Still, our understanding of how CDRs encode specific loop conformations lags far behind our understanding of structure-function relationships in non-immunological scaffolds. Thus, design of antibodies from first principles has not been demonstrated. We propose a computational-experimental strategy to address this challenge. We will: (a) characterize the design principles and sequence elements that rigidify antibody CDRs. Natural antibody loops will be subjected to computational modeling, crystallography, and a combined in vitro evolution and deep-sequencing approach to isolate sequence features that rigidify loop backbones; (b) develop a novel computational-design strategy, which uses the >1000 solved structures of antibodies deposited in structure databases to realistically model CDRs and design them to recognize proteins that have not been co-crystallized with antibodies. For example, we will design novel antibodies targeting insulin, for which clinically useful diagnostics are needed. By accessing much larger sequence/structure spaces than are available to natural immune-system repertoires and experimental methods, computational antibody design could produce higher-specificity and higher-affinity binders, even to challenging targets; and (c) develop new strategies to program conformational change in CDRs, generating, e.g., the first allosteric antibodies. These will allow targeting, in principle, of any molecule, potentially revolutionizing how antibodies are generated for research and medicine, providing new insights on the design principles of protein functional sites.
Max ERC Funding
1 499 930 €
Duration
Start date: 2013-09-01, End date: 2018-08-31
Project acronym ACMO
Project Systematic dissection of molecular machines and neural circuits coordinating C. elegans aggregation behaviour
Researcher (PI) Mario De Bono
Host Institution (HI) MEDICAL RESEARCH COUNCIL
Call Details Advanced Grant (AdG), LS5, ERC-2010-AdG_20100317
Summary Elucidating how neural circuits coordinate behaviour, and how molecules underpin the properties of individual neurons are major goals of neuroscience. Optogenetics and neural imaging combined with the powerful genetics and well-described nervous system of C. elegans offer special opportunities to address these questions. Previously, we identified a series of sensory neurons that modulate aggregation of C. elegans. These include neurons that respond to O2, CO2, noxious cues, satiety state, and pheromones. We propose to take our analysis to the next level by dissecting how, in mechanistic molecular terms, these distributed inputs modify the activity of populations of interneurons and motoneurons to coordinate group formation. Our strategy is to develop new, highly parallel approaches to replace the traditional piecemeal analysis.
We propose to:
1) Harness next generation sequencing (NGS) to forward genetics, rapidly to identify a molecular ¿parts list¿ for aggregation. Much of the genetics has been done: we have identified almost 200 mutations that inhibit or enhance aggregation but otherwise show no overt phenotype. A pilot study of 50 of these mutations suggests they identify dozens of genes not previously implicated in aggregation. NGS will allow us to molecularly identify these genes in a few months, providing multiple entry points to study molecular and circuitry mechanisms for behaviour.
2) Develop new methods to image the activity of populations of neurons in immobilized and freely moving animals, using genetically encoded indicators such as the calcium sensor cameleon and the voltage indicator mermaid.
This will be the first time a complex behaviour has been dissected in this way. We expect to identify novel conserved molecular and circuitry mechanisms.
Summary
Elucidating how neural circuits coordinate behaviour, and how molecules underpin the properties of individual neurons are major goals of neuroscience. Optogenetics and neural imaging combined with the powerful genetics and well-described nervous system of C. elegans offer special opportunities to address these questions. Previously, we identified a series of sensory neurons that modulate aggregation of C. elegans. These include neurons that respond to O2, CO2, noxious cues, satiety state, and pheromones. We propose to take our analysis to the next level by dissecting how, in mechanistic molecular terms, these distributed inputs modify the activity of populations of interneurons and motoneurons to coordinate group formation. Our strategy is to develop new, highly parallel approaches to replace the traditional piecemeal analysis.
We propose to:
1) Harness next generation sequencing (NGS) to forward genetics, rapidly to identify a molecular ¿parts list¿ for aggregation. Much of the genetics has been done: we have identified almost 200 mutations that inhibit or enhance aggregation but otherwise show no overt phenotype. A pilot study of 50 of these mutations suggests they identify dozens of genes not previously implicated in aggregation. NGS will allow us to molecularly identify these genes in a few months, providing multiple entry points to study molecular and circuitry mechanisms for behaviour.
2) Develop new methods to image the activity of populations of neurons in immobilized and freely moving animals, using genetically encoded indicators such as the calcium sensor cameleon and the voltage indicator mermaid.
This will be the first time a complex behaviour has been dissected in this way. We expect to identify novel conserved molecular and circuitry mechanisms.
Max ERC Funding
2 439 996 €
Duration
Start date: 2011-04-01, End date: 2017-03-31
Project acronym ACTINONSRF
Project MAL: an actin-regulated SRF transcriptional coactivator
Researcher (PI) Richard Treisman
Host Institution (HI) THE FRANCIS CRICK INSTITUTE LIMITED
Call Details Advanced Grant (AdG), LS1, ERC-2010-AdG_20100317
Summary MAL: an actin-regulated SRF transcriptional coactivator
Recent years have seen a revitalised interest in the role of actin in nuclear processes, but the molecular mechanisms involved remain largely unexplored. We will elucidate the molecular basis for the actin-based control of the SRF transcriptional coactivator, MAL. SRF controls transcription through two families of coactivators, the actin-binding MRTFs (MAL, Mkl2), which couple its activity to cytoskeletal dynamics, and the ERK-regulated TCFs (Elk-1, SAP-1, Net). MAL subcellular localisation and transcriptional activity responds to signal-induced changes in G-actin concentration, which are sensed by its actin-binding N-terminal RPEL domain. Members of a second family of RPEL proteins, the Phactrs, also exhibit actin-regulated nucleocytoplasmic shuttling. The proposal addresses the following novel features of actin biology:
¿ Actin as a transcriptional regulator
¿ Actin as a signalling molecule
¿ Actin-binding proteins as targets for regulation by actin, rather than regulators of actin function
We will analyse the sequences and proteins involved in actin-regulated nucleocytoplasmic shuttling, using structural biology and biochemistry to analyse its control by changes in actin-RPEL domain interactions. We will characterise the dynamics of shuttling, and develop reporters for changes in actin-MAL interaction for analysis of pathway activation in vivo. We will identify genes controlling MAL itself, and the balance between the nuclear and cytoplasmic actin pools. The mechanism by which actin represses transcriptional activation by MAL in the nucleus, and its relation to MAL phosphorylation, will be elucidated. Finally, we will map MRTF and TCF cofactor recruitment to SRF targets on a genome-wide scale, and identify the steps in transcription controlled by actin-MAL interaction.
Summary
MAL: an actin-regulated SRF transcriptional coactivator
Recent years have seen a revitalised interest in the role of actin in nuclear processes, but the molecular mechanisms involved remain largely unexplored. We will elucidate the molecular basis for the actin-based control of the SRF transcriptional coactivator, MAL. SRF controls transcription through two families of coactivators, the actin-binding MRTFs (MAL, Mkl2), which couple its activity to cytoskeletal dynamics, and the ERK-regulated TCFs (Elk-1, SAP-1, Net). MAL subcellular localisation and transcriptional activity responds to signal-induced changes in G-actin concentration, which are sensed by its actin-binding N-terminal RPEL domain. Members of a second family of RPEL proteins, the Phactrs, also exhibit actin-regulated nucleocytoplasmic shuttling. The proposal addresses the following novel features of actin biology:
¿ Actin as a transcriptional regulator
¿ Actin as a signalling molecule
¿ Actin-binding proteins as targets for regulation by actin, rather than regulators of actin function
We will analyse the sequences and proteins involved in actin-regulated nucleocytoplasmic shuttling, using structural biology and biochemistry to analyse its control by changes in actin-RPEL domain interactions. We will characterise the dynamics of shuttling, and develop reporters for changes in actin-MAL interaction for analysis of pathway activation in vivo. We will identify genes controlling MAL itself, and the balance between the nuclear and cytoplasmic actin pools. The mechanism by which actin represses transcriptional activation by MAL in the nucleus, and its relation to MAL phosphorylation, will be elucidated. Finally, we will map MRTF and TCF cofactor recruitment to SRF targets on a genome-wide scale, and identify the steps in transcription controlled by actin-MAL interaction.
Max ERC Funding
1 889 995 €
Duration
Start date: 2011-10-01, End date: 2017-09-30
Project acronym ACTIVE_NEUROGENESIS
Project Activity-dependent signaling in radial glial cells and their neuronal progeny
Researcher (PI) Colin Akerman
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Starting Grant (StG), LS5, ERC-2009-StG
Summary A significant advance in the field of development has been the appreciation that radial glial cells are progenitors and give birth to neurons in the brain. In order to advance this exciting area of biology, we need approaches that combine structural and functional studies of these cells. This is reflected by the emerging realisation that dynamic interactions involving radial glia may be critical for the regulation of their proliferative behaviour. It has been observed that radial glia experience transient elevations in intracellular Ca2+ but the nature of these signals, and the information that they convey, is not known. The inability to observe these cells in vivo and over the course of their development has also meant that basic questions remain unexplored. For instance, how does the behaviour of a radial glial cell at one point in development, influence the final identity of its progeny? I propose to build a research team that will capitalise upon methods we have developed for observing individual radial glia and their progeny in an intact vertebrate nervous system. The visual system of Xenopus Laevis tadpoles offers non-invasive optical access to the brain, making time-lapse imaging of single cells feasible over minutes and weeks. The system s anatomy lends itself to techniques that measure the activity of the cells in a functional sensory network. We will use this to examine signalling mechanisms in radial glia and how a radial glial cell s experience influences its proliferative behaviour and the types of neuron it generates. We will also examine the interactions that continue between a radial glial cell and its daughter neurons. Finally, we will explore the relationships that exist within neuronal progeny derived from a single radial glial cell.
Summary
A significant advance in the field of development has been the appreciation that radial glial cells are progenitors and give birth to neurons in the brain. In order to advance this exciting area of biology, we need approaches that combine structural and functional studies of these cells. This is reflected by the emerging realisation that dynamic interactions involving radial glia may be critical for the regulation of their proliferative behaviour. It has been observed that radial glia experience transient elevations in intracellular Ca2+ but the nature of these signals, and the information that they convey, is not known. The inability to observe these cells in vivo and over the course of their development has also meant that basic questions remain unexplored. For instance, how does the behaviour of a radial glial cell at one point in development, influence the final identity of its progeny? I propose to build a research team that will capitalise upon methods we have developed for observing individual radial glia and their progeny in an intact vertebrate nervous system. The visual system of Xenopus Laevis tadpoles offers non-invasive optical access to the brain, making time-lapse imaging of single cells feasible over minutes and weeks. The system s anatomy lends itself to techniques that measure the activity of the cells in a functional sensory network. We will use this to examine signalling mechanisms in radial glia and how a radial glial cell s experience influences its proliferative behaviour and the types of neuron it generates. We will also examine the interactions that continue between a radial glial cell and its daughter neurons. Finally, we will explore the relationships that exist within neuronal progeny derived from a single radial glial cell.
Max ERC Funding
1 284 808 €
Duration
Start date: 2010-02-01, End date: 2015-01-31
Project acronym ACTSELECTCONTEXT
Project Action Selection under Contextual Uncertainty: the Role of Learning and Effective Connectivity in the Human Brain
Researcher (PI) Sven Bestmann
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Starting Grant (StG), LS5, ERC-2010-StG_20091118
Summary In a changing world, one hallmark feature of human behaviour is the ability to learn about the statistics of the environment and use this prior information for action selection. Knowing about a forthcoming event allows for adjusting our actions pre-emptively, which can optimize survival.
This proposal studies how the human brain learns about the uncertainty in the environment, and how this leads to flexible and efficient action selection.
I hypothesise that the accumulation of evidence for future movements through learning reflects a fundamental organisational principle for action control. This explains widely distributed perceptual-, learning-, decision-, and movement-related signals in the human brain. However, little is known about the concerted interplay between brain regions in terms of effective connectivity which is required for flexible behaviour.
My proposal seeks to shed light on this unresolved issue. To this end, I will use i) a multi-disciplinary neuroimaging approach, together with model-based analyses and Bayesian model comparison, adapted to human reaching behaviour as occurring in daily life; and ii) two novel approaches for testing effective connectivity: dynamic causal modelling (DCM) and concurrent transcranial magnetic stimulation-functional magnetic resonance imaging.
My prediction is that action selection relies on effective connectivity changes, which are a function of the prior information that the brain has to learn about.
If true, this will provide novel insight into the human ability to select actions, based on learning about the uncertainty which is inherent in contextual information. This is relevant for understanding action selection during development and ageing, and for pathologies of action such as Parkinson s disease or stroke.
Summary
In a changing world, one hallmark feature of human behaviour is the ability to learn about the statistics of the environment and use this prior information for action selection. Knowing about a forthcoming event allows for adjusting our actions pre-emptively, which can optimize survival.
This proposal studies how the human brain learns about the uncertainty in the environment, and how this leads to flexible and efficient action selection.
I hypothesise that the accumulation of evidence for future movements through learning reflects a fundamental organisational principle for action control. This explains widely distributed perceptual-, learning-, decision-, and movement-related signals in the human brain. However, little is known about the concerted interplay between brain regions in terms of effective connectivity which is required for flexible behaviour.
My proposal seeks to shed light on this unresolved issue. To this end, I will use i) a multi-disciplinary neuroimaging approach, together with model-based analyses and Bayesian model comparison, adapted to human reaching behaviour as occurring in daily life; and ii) two novel approaches for testing effective connectivity: dynamic causal modelling (DCM) and concurrent transcranial magnetic stimulation-functional magnetic resonance imaging.
My prediction is that action selection relies on effective connectivity changes, which are a function of the prior information that the brain has to learn about.
If true, this will provide novel insight into the human ability to select actions, based on learning about the uncertainty which is inherent in contextual information. This is relevant for understanding action selection during development and ageing, and for pathologies of action such as Parkinson s disease or stroke.
Max ERC Funding
1 341 805 €
Duration
Start date: 2011-06-01, End date: 2016-05-31
Project acronym ADOR
Project Assembly-disassembly-organisation-reassembly of microporous materials
Researcher (PI) Russell MORRIS
Host Institution (HI) THE UNIVERSITY COURT OF THE UNIVERSITY OF ST ANDREWS
Call Details Advanced Grant (AdG), PE5, ERC-2017-ADG
Summary Microporous materials are an important class of solid; the two main members of this family are zeolites and metal-organic frameworks (MOFs). Zeolites are industrial solids whose applications range from catalysis, through ion exchange and adsorption technologies to medicine. MOFs are some of the most exciting new materials to have been developed over the last two decades, and they are just beginning to be applied commercially.
Over recent years the applicant’s group has developed new synthetic strategies to prepare microporous materials, called the Assembly-Disassembly-Organisation-Reassembly (ADOR) process. In significant preliminary work the ADOR process has shown to be an extremely important new synthetic methodology that differs fundamentally from traditional solvothermal methods.
In this project I will look to overturn the conventional thinking in materials science by developing methodologies that can target both zeolites and MOF materials that are difficult to prepare using traditional methods – the so-called ‘unfeasible’ materials. The importance of such a new methodology is that it will open up routes to materials that have different properties (both chemical and topological) to those we currently have. Since zeolites and MOFs have so many actual and potential uses, the preparation of materials with different properties has a high chance of leading to new technologies in the medium/long term. To complete the major objective I will look to complete four closely linked activities covering the development of design strategies for zeolites and MOFs (activities 1 & 2), mechanistic studies to understand the process at the molecular level using in situ characterisation techniques (activity 3) and an exploration of potential applied science for the prepared materials (activity 4).
Summary
Microporous materials are an important class of solid; the two main members of this family are zeolites and metal-organic frameworks (MOFs). Zeolites are industrial solids whose applications range from catalysis, through ion exchange and adsorption technologies to medicine. MOFs are some of the most exciting new materials to have been developed over the last two decades, and they are just beginning to be applied commercially.
Over recent years the applicant’s group has developed new synthetic strategies to prepare microporous materials, called the Assembly-Disassembly-Organisation-Reassembly (ADOR) process. In significant preliminary work the ADOR process has shown to be an extremely important new synthetic methodology that differs fundamentally from traditional solvothermal methods.
In this project I will look to overturn the conventional thinking in materials science by developing methodologies that can target both zeolites and MOF materials that are difficult to prepare using traditional methods – the so-called ‘unfeasible’ materials. The importance of such a new methodology is that it will open up routes to materials that have different properties (both chemical and topological) to those we currently have. Since zeolites and MOFs have so many actual and potential uses, the preparation of materials with different properties has a high chance of leading to new technologies in the medium/long term. To complete the major objective I will look to complete four closely linked activities covering the development of design strategies for zeolites and MOFs (activities 1 & 2), mechanistic studies to understand the process at the molecular level using in situ characterisation techniques (activity 3) and an exploration of potential applied science for the prepared materials (activity 4).
Max ERC Funding
2 489 220 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym AlCat
Project Bond activation and catalysis with low-valent aluminium
Researcher (PI) Michael James COWLEY
Host Institution (HI) THE UNIVERSITY OF EDINBURGH
Call Details Starting Grant (StG), PE5, ERC-2016-STG
Summary This project will develop the principles required to enable bond-modifying redox catalysis based on aluminium by preparing and studying new Al(I) compounds capable of reversible oxidative addition.
Catalytic processes are involved in the synthesis of 75 % of all industrially produced chemicals, but most catalysts involved are based on precious metals such as rhodium, palladium or platinum. These metals are expensive and their supply limited and unstable; there is a significant need to develop the chemistry of non-precious metals as alternatives. On toxicity and abundance alone, aluminium is an attractive candidate. Furthermore, recent work, including in our group, has demonstrated that Al(I) compounds can perform a key step in catalytic cycles - the oxidative addition of E-H bonds.
In order to realise the significant potential of Al(I) for transition-metal style catalysis we urgently need to:
- establish the principles governing oxidative addition and reductive elimination reactivity in aluminium systems.
- know how the reactivity of Al(I) compounds can be controlled by varying properties of ligand frameworks.
- understand the onward reactivity of oxidative addition products of Al(I) to enable applications in catalysis.
In this project we will:
- Study mechanisms of oxidative addition and reductive elimination of a range of synthetically relevant bonds at Al(I) centres, establishing the principles governing this fundamental reactivity.
- Develop new ligand frameworks to support of Al(I) centres and evaluate the effect of the ligand on oxidative addition/reductive elimination at Al centres.
- Investigate methods for Al-mediated functionalisation of organic compounds by exploring the reactivity of E-H oxidative addition products with unsaturated organic compounds.
Summary
This project will develop the principles required to enable bond-modifying redox catalysis based on aluminium by preparing and studying new Al(I) compounds capable of reversible oxidative addition.
Catalytic processes are involved in the synthesis of 75 % of all industrially produced chemicals, but most catalysts involved are based on precious metals such as rhodium, palladium or platinum. These metals are expensive and their supply limited and unstable; there is a significant need to develop the chemistry of non-precious metals as alternatives. On toxicity and abundance alone, aluminium is an attractive candidate. Furthermore, recent work, including in our group, has demonstrated that Al(I) compounds can perform a key step in catalytic cycles - the oxidative addition of E-H bonds.
In order to realise the significant potential of Al(I) for transition-metal style catalysis we urgently need to:
- establish the principles governing oxidative addition and reductive elimination reactivity in aluminium systems.
- know how the reactivity of Al(I) compounds can be controlled by varying properties of ligand frameworks.
- understand the onward reactivity of oxidative addition products of Al(I) to enable applications in catalysis.
In this project we will:
- Study mechanisms of oxidative addition and reductive elimination of a range of synthetically relevant bonds at Al(I) centres, establishing the principles governing this fundamental reactivity.
- Develop new ligand frameworks to support of Al(I) centres and evaluate the effect of the ligand on oxidative addition/reductive elimination at Al centres.
- Investigate methods for Al-mediated functionalisation of organic compounds by exploring the reactivity of E-H oxidative addition products with unsaturated organic compounds.
Max ERC Funding
1 493 679 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym ALIGN
Project Ab-initio computational modelling of photovoltaic interfaces
Researcher (PI) Feliciano Giustino
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Starting Grant (StG), PE5, ERC-2009-StG
Summary The aim of the ALIGN project is to understand, predict, and optimize the photovoltaic energy conversion in third-generation solar cells, starting from an atomic-scale quantum-mechanical modelling of the photovoltaic interface. The quest for photovoltaic materials suitable for low-cost synthesis, large-area production, and functional architecture has driven substantial research efforts towards third-generation photovoltaic devices such as plastic solar cells, organic-inorganic cells, and photo-electrochemical cells. The physical and chemical processes involved in the harvesting of sunlight, the transport of electrical charge, and the build-up of the photo-voltage in these devices are fundamentally different from those encountered in traditional semiconductor heterojunction solar cells. A detailed atomic-scale quantum-mechanical description of such processes will lay down the basis for a rational approach to the modelling, optimization, and design of new photovoltaic materials. The short name of the proposal hints at one of the key materials parameters in the area of photovoltaic interfaces: the alignment of the quantum energy levels between the light-absorbing material and the electron acceptor. The level alignment drives the separation of the electron-hole pairs formed upon absorption of sunlight, and determines the open circuit voltage of the solar cell. The energy level alignment not only represents a key parameter for the design of photovoltaic devices, but also constitutes one of the grand challenges of modern computational materials science. Within this project we will develop and apply new ground-breaking computational methods to understand, predict, and optimize the energy level alignment and other design parameters of third-generation photovoltaic devices.
Summary
The aim of the ALIGN project is to understand, predict, and optimize the photovoltaic energy conversion in third-generation solar cells, starting from an atomic-scale quantum-mechanical modelling of the photovoltaic interface. The quest for photovoltaic materials suitable for low-cost synthesis, large-area production, and functional architecture has driven substantial research efforts towards third-generation photovoltaic devices such as plastic solar cells, organic-inorganic cells, and photo-electrochemical cells. The physical and chemical processes involved in the harvesting of sunlight, the transport of electrical charge, and the build-up of the photo-voltage in these devices are fundamentally different from those encountered in traditional semiconductor heterojunction solar cells. A detailed atomic-scale quantum-mechanical description of such processes will lay down the basis for a rational approach to the modelling, optimization, and design of new photovoltaic materials. The short name of the proposal hints at one of the key materials parameters in the area of photovoltaic interfaces: the alignment of the quantum energy levels between the light-absorbing material and the electron acceptor. The level alignment drives the separation of the electron-hole pairs formed upon absorption of sunlight, and determines the open circuit voltage of the solar cell. The energy level alignment not only represents a key parameter for the design of photovoltaic devices, but also constitutes one of the grand challenges of modern computational materials science. Within this project we will develop and apply new ground-breaking computational methods to understand, predict, and optimize the energy level alignment and other design parameters of third-generation photovoltaic devices.
Max ERC Funding
1 000 000 €
Duration
Start date: 2010-03-01, End date: 2016-02-29
Project acronym ALZSYN
Project Imaging synaptic contributors to dementia
Researcher (PI) Tara Spires-Jones
Host Institution (HI) THE UNIVERSITY OF EDINBURGH
Call Details Consolidator Grant (CoG), LS5, ERC-2015-CoG
Summary Alzheimer's disease, the most common cause of dementia in older people, is a devastating condition that is becoming a public health crisis as our population ages. Despite great progress recently in Alzheimer’s disease research, we have no disease modifying drugs and a decade with a 99.6% failure rate of clinical trials attempting to treat the disease. This project aims to develop relevant therapeutic targets to restore brain function in Alzheimer’s disease by integrating human and model studies of synapses. It is widely accepted in the field that alterations in amyloid beta initiate the disease process. However the cascade leading from changes in amyloid to widespread tau pathology and neurodegeneration remain unclear. Synapse loss is the strongest pathological correlate of dementia in Alzheimer’s, and mounting evidence suggests that synapse degeneration plays a key role in causing cognitive decline. Here I propose to test the hypothesis that the amyloid cascade begins at the synapse leading to tau pathology, synapse dysfunction and loss, and ultimately neural circuit collapse causing cognitive impairment. The team will use cutting-edge multiphoton and array tomography imaging techniques to test mechanisms downstream of amyloid beta at synapses, and determine whether intervening in the cascade allows recovery of synapse structure and function. Importantly, I will combine studies in robust models of familial Alzheimer’s disease with studies in postmortem human brain to confirm relevance of our mechanistic studies to human disease. Finally, human stem cell derived neurons will be used to test mechanisms and potential therapeutics in neurons expressing the human proteome. Together, these experiments are ground-breaking since they have the potential to further our understanding of how synapses are lost in Alzheimer’s disease and to identify targets for effective therapeutic intervention, which is a critical unmet need in today’s health care system.
Summary
Alzheimer's disease, the most common cause of dementia in older people, is a devastating condition that is becoming a public health crisis as our population ages. Despite great progress recently in Alzheimer’s disease research, we have no disease modifying drugs and a decade with a 99.6% failure rate of clinical trials attempting to treat the disease. This project aims to develop relevant therapeutic targets to restore brain function in Alzheimer’s disease by integrating human and model studies of synapses. It is widely accepted in the field that alterations in amyloid beta initiate the disease process. However the cascade leading from changes in amyloid to widespread tau pathology and neurodegeneration remain unclear. Synapse loss is the strongest pathological correlate of dementia in Alzheimer’s, and mounting evidence suggests that synapse degeneration plays a key role in causing cognitive decline. Here I propose to test the hypothesis that the amyloid cascade begins at the synapse leading to tau pathology, synapse dysfunction and loss, and ultimately neural circuit collapse causing cognitive impairment. The team will use cutting-edge multiphoton and array tomography imaging techniques to test mechanisms downstream of amyloid beta at synapses, and determine whether intervening in the cascade allows recovery of synapse structure and function. Importantly, I will combine studies in robust models of familial Alzheimer’s disease with studies in postmortem human brain to confirm relevance of our mechanistic studies to human disease. Finally, human stem cell derived neurons will be used to test mechanisms and potential therapeutics in neurons expressing the human proteome. Together, these experiments are ground-breaking since they have the potential to further our understanding of how synapses are lost in Alzheimer’s disease and to identify targets for effective therapeutic intervention, which is a critical unmet need in today’s health care system.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-11-01, End date: 2021-10-31
Project acronym AMPRO
Project Advanced Electronic Materials and Devices through Novel Processing Paradigms
Researcher (PI) Thomas Anthopoulos
Host Institution (HI) IMPERIAL COLLEGE OF SCIENCE TECHNOLOGY AND MEDICINE
Call Details Starting Grant (StG), PE5, ERC-2011-StG_20101014
Summary "I propose a structured multidisciplinary research programme that seeks to combine advanced materials, such as metal oxides and organics, with novel fabrication methods to develop devices for application in: (1) large area electronics, (2) integrated nanoelectronics and (3) sensors. At the heart of this programme lies the development of novel oxide semiconductors. These will be synthesised from solution using precursors. Chemical doping via physical blending will be explored for the tuning of the electronic properties of these compounds. This simple approach will enable the rapid development of a library of materials far beyond those accessible by traditional methods. Oxides will then be combined with inorganic/organic dielectrics to demonstrate low power transistors. Ultimate target for application area (1) is the development of transistors with hole/electron mobilities exceeding 20/200 cm^2/Vs respectively. For application area (2) I will combine the precursor formulations with advanced scanning thermochemical nanolithography. A heated atomic force microscope tip will be used for the local chemical conversion of the precursor to oxide with sub-50 nm resolution. This will enable patterning of nanostructures with desirable shape and size. Sequential patterning of semi/conductive layers combined with SAM dielectrics would enable fabrication of nano-sized devices and circuits. For application area (3), research effort will focus on novel hybrid phototransistors. Use of different light absorbing organic dyes functionalised onto the oxide channel will be explored as a mean for developing high sensitivity phototransistors and full colour sensing arrays. Organic dyes will also be combined with nano-sized transistors to demonstrate integrated nano-scale optoelectronics. The unique combination of bottom-up and top-down strategies adopted in this project will lead to the development of novel high performance devices with a host of existing and new applications."
Summary
"I propose a structured multidisciplinary research programme that seeks to combine advanced materials, such as metal oxides and organics, with novel fabrication methods to develop devices for application in: (1) large area electronics, (2) integrated nanoelectronics and (3) sensors. At the heart of this programme lies the development of novel oxide semiconductors. These will be synthesised from solution using precursors. Chemical doping via physical blending will be explored for the tuning of the electronic properties of these compounds. This simple approach will enable the rapid development of a library of materials far beyond those accessible by traditional methods. Oxides will then be combined with inorganic/organic dielectrics to demonstrate low power transistors. Ultimate target for application area (1) is the development of transistors with hole/electron mobilities exceeding 20/200 cm^2/Vs respectively. For application area (2) I will combine the precursor formulations with advanced scanning thermochemical nanolithography. A heated atomic force microscope tip will be used for the local chemical conversion of the precursor to oxide with sub-50 nm resolution. This will enable patterning of nanostructures with desirable shape and size. Sequential patterning of semi/conductive layers combined with SAM dielectrics would enable fabrication of nano-sized devices and circuits. For application area (3), research effort will focus on novel hybrid phototransistors. Use of different light absorbing organic dyes functionalised onto the oxide channel will be explored as a mean for developing high sensitivity phototransistors and full colour sensing arrays. Organic dyes will also be combined with nano-sized transistors to demonstrate integrated nano-scale optoelectronics. The unique combination of bottom-up and top-down strategies adopted in this project will lead to the development of novel high performance devices with a host of existing and new applications."
Max ERC Funding
1 497 798 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym AMYTOX
Project Amyloid fibril cytotoxicity: new insights from novel approaches
Researcher (PI) Sheena Radford
Host Institution (HI) UNIVERSITY OF LEEDS
Call Details Advanced Grant (AdG), LS1, ERC-2012-ADG_20120314
Summary Despite the discovery of amyloidosis more than a century ago, the molecular and cellular mechanisms of these devastating human disorders remain obscure. In addition to their involvement in disease, amyloid fibrils perform physiological functions, whilst others have potentials as biomaterials. To realise their use in nanotechnology and to enable the development of amyloid therapies, there is an urgent need to understand the molecular pathways of amyloid assembly and to determine how amyloid fibrils interact with cells and cellular components. The challenges lie in the transient nature and low population of aggregating species and the panoply of amyloid fibril structures. This molecular complexity renders identification of the culprits of amyloid disease impossible to achieve using traditional methods.
Here I propose a series of exciting experiments that aim to cast new light on the molecular and cellular mechanisms of amyloidosis by exploiting approaches capable of imaging individual protein molecules or single protein fibrils in vitro and in living cells. The proposal builds on new data from our laboratory that have shown that amyloid fibrils (disease-associated, functional and created from de novo designed sequences) kill cells by a mechanism that depends on fibril length and on cellular uptake. Specifically, I will (i) use single molecule fluorescence and non-covalent mass spectrometry and to determine why short fibril samples disrupt biological membranes more than their longer counterparts and electron tomography to determine, for the first time, the structural properties of cytotoxic fibril ends; (ii) develop single molecule force spectroscopy to probe the interactions between amyloid precursors, fibrils and cellular membranes; and (iii) develop cell biological assays to discover the biological mechanism(s) of amyloid-induced cell death and high resolution imaging and electron tomography to visualise amyloid fibrils in the act of killing living cells.
Summary
Despite the discovery of amyloidosis more than a century ago, the molecular and cellular mechanisms of these devastating human disorders remain obscure. In addition to their involvement in disease, amyloid fibrils perform physiological functions, whilst others have potentials as biomaterials. To realise their use in nanotechnology and to enable the development of amyloid therapies, there is an urgent need to understand the molecular pathways of amyloid assembly and to determine how amyloid fibrils interact with cells and cellular components. The challenges lie in the transient nature and low population of aggregating species and the panoply of amyloid fibril structures. This molecular complexity renders identification of the culprits of amyloid disease impossible to achieve using traditional methods.
Here I propose a series of exciting experiments that aim to cast new light on the molecular and cellular mechanisms of amyloidosis by exploiting approaches capable of imaging individual protein molecules or single protein fibrils in vitro and in living cells. The proposal builds on new data from our laboratory that have shown that amyloid fibrils (disease-associated, functional and created from de novo designed sequences) kill cells by a mechanism that depends on fibril length and on cellular uptake. Specifically, I will (i) use single molecule fluorescence and non-covalent mass spectrometry and to determine why short fibril samples disrupt biological membranes more than their longer counterparts and electron tomography to determine, for the first time, the structural properties of cytotoxic fibril ends; (ii) develop single molecule force spectroscopy to probe the interactions between amyloid precursors, fibrils and cellular membranes; and (iii) develop cell biological assays to discover the biological mechanism(s) of amyloid-induced cell death and high resolution imaging and electron tomography to visualise amyloid fibrils in the act of killing living cells.
Max ERC Funding
2 498 465 €
Duration
Start date: 2013-05-01, End date: 2019-04-30
Project acronym ANXIETY MECHANISMS
Project Neurocognitive mechanisms of human anxiety: identifying and
targeting disrupted function
Researcher (PI) Sonia Jane Bishop
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Starting Grant (StG), LS5, ERC-2010-StG_20091118
Summary Within a 12 month period, 20% of adults will meet criteria for one or more clinical anxiety disorders (ADs). These disorders are hugely disruptive, placing an emotional burden on individuals and their families. While both cognitive behavioural therapy and pharmacological treatment are widely viewed as effective strategies for managing ADs, systematic review of the literature reveals that only 30–45% of patients demonstrate a marked response to treatment (anxiety levels being reduced into the nonaffected range). In addition, a significant proportion of initial responders relapse after treatment is discontinued. There is hence a real and marked need to improve upon current approaches to AD treatment.
One possible avenue for improving response rates is through optimizing initial treatment selection. Specifically, it is possible that certain individuals might respond better to cognitive interventions while others might respond better to pharmacological treatment. Recently it has been suggested that there may be two or more distinct biological pathways disrupted in anxiety. If this is the case, then specification of these pathways may be an important step in predicting which individuals are likely to respond to which treatment. Few studies have focused upon this issue and, in particular, upon identifying neural markers that might predict response to cognitive (as opposed to pharmacological) intervention. The proposed research aims to address this. Specifically, it tests the hypothesis that there are at least two mechanisms disrupted in ADs, one entailing amygdala hyper-responsivity to cues that signal threat, the other impoverished recruitment of frontal regions that support cognitive and emotional regulation.
Two series of functional magnetic resonance imaging experiments will be conducted. These will investigate differences in amygdala and frontal function during (a) attentional processing and (b) fear conditioning. Initial clinical experiments will investigate whether Generalised Anxiety Disorder and Specific Phobia involve differing degrees of disruption to frontal versus amygdala function during these tasks. This work will feed into training studies, the goal being to characterize AD patient subgroups that benefit from cognitive training.
Summary
Within a 12 month period, 20% of adults will meet criteria for one or more clinical anxiety disorders (ADs). These disorders are hugely disruptive, placing an emotional burden on individuals and their families. While both cognitive behavioural therapy and pharmacological treatment are widely viewed as effective strategies for managing ADs, systematic review of the literature reveals that only 30–45% of patients demonstrate a marked response to treatment (anxiety levels being reduced into the nonaffected range). In addition, a significant proportion of initial responders relapse after treatment is discontinued. There is hence a real and marked need to improve upon current approaches to AD treatment.
One possible avenue for improving response rates is through optimizing initial treatment selection. Specifically, it is possible that certain individuals might respond better to cognitive interventions while others might respond better to pharmacological treatment. Recently it has been suggested that there may be two or more distinct biological pathways disrupted in anxiety. If this is the case, then specification of these pathways may be an important step in predicting which individuals are likely to respond to which treatment. Few studies have focused upon this issue and, in particular, upon identifying neural markers that might predict response to cognitive (as opposed to pharmacological) intervention. The proposed research aims to address this. Specifically, it tests the hypothesis that there are at least two mechanisms disrupted in ADs, one entailing amygdala hyper-responsivity to cues that signal threat, the other impoverished recruitment of frontal regions that support cognitive and emotional regulation.
Two series of functional magnetic resonance imaging experiments will be conducted. These will investigate differences in amygdala and frontal function during (a) attentional processing and (b) fear conditioning. Initial clinical experiments will investigate whether Generalised Anxiety Disorder and Specific Phobia involve differing degrees of disruption to frontal versus amygdala function during these tasks. This work will feed into training studies, the goal being to characterize AD patient subgroups that benefit from cognitive training.
Max ERC Funding
1 708 407 €
Duration
Start date: 2011-04-01, End date: 2016-08-31
Project acronym ARYLATOR
Project New Catalytic Reactions and Exchange Pathways: Delivering Versatile and Reliable Arylation
Researcher (PI) Guy Charles Lloyd-Jones
Host Institution (HI) THE UNIVERSITY OF EDINBURGH
Call Details Advanced Grant (AdG), PE5, ERC-2013-ADG
Summary This proposal details the mechanism-based discovery of ground-breaking new catalyst systems for a broad range of arylation processes that will be of immediate and long-lasting utility to the pharmaceutical, agrochemical, and materials chemistry industries. These industries have become highly dependent on coupling technologies employing homogeneous late transition metal catalysis and this reliance will grow further, particularly if the substrate scope can be broadened, the economics, in terms of reagents and catalyst, made more favourable, the reliability at scale-up improved, and the generation of side-products, of particular importance for optical and electronic properties of materials, minimized or eliminated.
This proposal addresses these issues by conducting a detailed and comprehensive mechanistic investigation of direct arylation, so that a substantial expansion of the reaction scope can be achieved. At present, the regioselectivity can be very high, however catalyst turnover rates are moderate, and the arene is required to be in a fairly narrow window of activity. Specific aspects to be addressed in terms of mechanistic study are: catalyst speciation and pathways for deactivation; pathways for homocoupling; influence of anions and dummy ligands; protodemetalloidation pathways. Areas proposed for mechanism-informed development are: expansion of metalloid tolerance; expansion of arene scope; use of traceless activators and directors, new couplings via ligand exchange, the evolution of simpler / cheaper and more selective / active catalysts; expansion to oxidative double arylations (Ar-H + Ar’-H) with control, and without resort to super-stoichiometric bias.
The long-term legacy of these studies will be detailed insight for current and emerging systems, as well as readily extrapolated information for the design of new, more efficient catalyst systems in academia, and their scaleable application in industry
Summary
This proposal details the mechanism-based discovery of ground-breaking new catalyst systems for a broad range of arylation processes that will be of immediate and long-lasting utility to the pharmaceutical, agrochemical, and materials chemistry industries. These industries have become highly dependent on coupling technologies employing homogeneous late transition metal catalysis and this reliance will grow further, particularly if the substrate scope can be broadened, the economics, in terms of reagents and catalyst, made more favourable, the reliability at scale-up improved, and the generation of side-products, of particular importance for optical and electronic properties of materials, minimized or eliminated.
This proposal addresses these issues by conducting a detailed and comprehensive mechanistic investigation of direct arylation, so that a substantial expansion of the reaction scope can be achieved. At present, the regioselectivity can be very high, however catalyst turnover rates are moderate, and the arene is required to be in a fairly narrow window of activity. Specific aspects to be addressed in terms of mechanistic study are: catalyst speciation and pathways for deactivation; pathways for homocoupling; influence of anions and dummy ligands; protodemetalloidation pathways. Areas proposed for mechanism-informed development are: expansion of metalloid tolerance; expansion of arene scope; use of traceless activators and directors, new couplings via ligand exchange, the evolution of simpler / cheaper and more selective / active catalysts; expansion to oxidative double arylations (Ar-H + Ar’-H) with control, and without resort to super-stoichiometric bias.
The long-term legacy of these studies will be detailed insight for current and emerging systems, as well as readily extrapolated information for the design of new, more efficient catalyst systems in academia, and their scaleable application in industry
Max ERC Funding
2 114 223 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym ASPIRE
Project Aqueous Supramolecular Polymers and Peptide Conjugates in Reversible Systems
Researcher (PI) Oren Alexander Scherman
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Starting Grant (StG), PE5, ERC-2009-StG
Summary Supramolecular polymers are of major interest in the field of self assembly with a promising outlook in areas of viscosity modification, compartmentalized architectures, bio-conjugates and drug-delivery applications. They are dynamic macromolecular materials prepared by simple mixing of relatively small components bearing complementary or self-complementary recognition motifs. A major limitation in the field, however, has been access to synthetic systems capable of undergoing self assembly in an aqueous environment. This research proposal develops well-defined, self-organizing macromolecular structures that will overcome this limitation by focusing on systems that rely on several non-covalent interactions occurring in concert rather than on single interactions alone. The envisioned supramolecular polymers and bio-conjugates are designed as dynamic water-soluble smart materials, whose architectures can be controlled and exhibit reversibility upon exposure to external stimuli such as electrochemical, temperature or pH changes. Molecular recognition events occurring between functional handles on both synthetic and bio-polymers will be investigated in order to control the formation of desired functional architectures through stoichiometrically controlled complexation. Preparation of synthetic core motifs to assemble discrete peptide aggregates such as the dimeric through hexameric oligomers of amyloid-beta(40/42) will lead to structural elucidation and insight into several peptide misfolding pathologies like Alzheimer's or Parkinson's disease.
Summary
Supramolecular polymers are of major interest in the field of self assembly with a promising outlook in areas of viscosity modification, compartmentalized architectures, bio-conjugates and drug-delivery applications. They are dynamic macromolecular materials prepared by simple mixing of relatively small components bearing complementary or self-complementary recognition motifs. A major limitation in the field, however, has been access to synthetic systems capable of undergoing self assembly in an aqueous environment. This research proposal develops well-defined, self-organizing macromolecular structures that will overcome this limitation by focusing on systems that rely on several non-covalent interactions occurring in concert rather than on single interactions alone. The envisioned supramolecular polymers and bio-conjugates are designed as dynamic water-soluble smart materials, whose architectures can be controlled and exhibit reversibility upon exposure to external stimuli such as electrochemical, temperature or pH changes. Molecular recognition events occurring between functional handles on both synthetic and bio-polymers will be investigated in order to control the formation of desired functional architectures through stoichiometrically controlled complexation. Preparation of synthetic core motifs to assemble discrete peptide aggregates such as the dimeric through hexameric oligomers of amyloid-beta(40/42) will lead to structural elucidation and insight into several peptide misfolding pathologies like Alzheimer's or Parkinson's disease.
Max ERC Funding
1 700 000 €
Duration
Start date: 2009-11-01, End date: 2015-10-31
Project acronym ATG9_SOLVES_IT
Project In vitro high resolution reconstitution of autophagosome nucleation and expansion catalyzed byATG9
Researcher (PI) Sharon TOOZE
Host Institution (HI) THE FRANCIS CRICK INSTITUTE LIMITED
Call Details Advanced Grant (AdG), LS1, ERC-2017-ADG
Summary Autophagy is a conserved, lysosomal-mediated pathway required for cell homeostasis and survival. It is controlled by the master regulators of energy (AMPK) and growth (TORC1) and mediated by the ATG (autophagy) proteins. Deregulation of autophagy is implicated in cancer, immunity, infection, aging and neurodegeneration. Autophagosomes form and expand using membranes from the secretory and endocytic pathways but how this occurs is not understood. ATG9, the only transmembrane ATG protein traffics through the cell in vesicles, and is essential for rapid initiation and expansion of the membranes which form the autophagosome. Crucially, how ATG9 functions is unknown. I will determine how ATG9 initiates the formation and expansion of the autophagosome by amino acid starvation through a molecular dissection of proteins resident in ATG9 vesicles which modulate the composition and property of the initiating membrane. I will employ high resolution light and electron microscopy to characterize the nucleation of the autophagosome, proximity-specific biotinylation and quantitative Mass Spectrometry to uncover the proteome required for the function of the ATG9, and optogenetic tools to acutely regulate signaling lipids. Lastly, with our tools and knowledge I will develop an in vitro reconstitution system to define at a molecular level how ATG9 vesicle proteins, membranes that interact with ATG9 vesicles, and other accessory ATG components nucleate and form an autophagosome. In vitro reconstitution of autophagosomes will be assayed biochemically, and by correlative light and cryo-EM and cryo-EM tomography, while functional reconstitution of autophagy will be tested by selective cargo recruitment. The development of a reconstituted system and identification proteins and lipids which are key components for autophagosome formation will provide a means to identify a new generation of targets for translational work leading to manipulation of autophagy for disease related therapies.
Summary
Autophagy is a conserved, lysosomal-mediated pathway required for cell homeostasis and survival. It is controlled by the master regulators of energy (AMPK) and growth (TORC1) and mediated by the ATG (autophagy) proteins. Deregulation of autophagy is implicated in cancer, immunity, infection, aging and neurodegeneration. Autophagosomes form and expand using membranes from the secretory and endocytic pathways but how this occurs is not understood. ATG9, the only transmembrane ATG protein traffics through the cell in vesicles, and is essential for rapid initiation and expansion of the membranes which form the autophagosome. Crucially, how ATG9 functions is unknown. I will determine how ATG9 initiates the formation and expansion of the autophagosome by amino acid starvation through a molecular dissection of proteins resident in ATG9 vesicles which modulate the composition and property of the initiating membrane. I will employ high resolution light and electron microscopy to characterize the nucleation of the autophagosome, proximity-specific biotinylation and quantitative Mass Spectrometry to uncover the proteome required for the function of the ATG9, and optogenetic tools to acutely regulate signaling lipids. Lastly, with our tools and knowledge I will develop an in vitro reconstitution system to define at a molecular level how ATG9 vesicle proteins, membranes that interact with ATG9 vesicles, and other accessory ATG components nucleate and form an autophagosome. In vitro reconstitution of autophagosomes will be assayed biochemically, and by correlative light and cryo-EM and cryo-EM tomography, while functional reconstitution of autophagy will be tested by selective cargo recruitment. The development of a reconstituted system and identification proteins and lipids which are key components for autophagosome formation will provide a means to identify a new generation of targets for translational work leading to manipulation of autophagy for disease related therapies.
Max ERC Funding
2 121 055 €
Duration
Start date: 2018-07-01, End date: 2023-06-30
Project acronym ATMINDDR
Project ATMINistrating ATM signalling: exploring the significance of ATM regulation by ATMIN
Researcher (PI) Axel Behrens
Host Institution (HI) THE FRANCIS CRICK INSTITUTE LIMITED
Call Details Starting Grant (StG), LS1, ERC-2011-StG_20101109
Summary ATM is the protein kinase that is mutated in the hereditary autosomal recessive disease ataxia telangiectasia (A-T). A-T patients display immune deficiencies, cancer predisposition and radiosensitivity. The molecular role of ATM is to respond to DNA damage by phosphorylating its substrates, thereby promoting repair of damage or arresting the cell cycle. Following the induction of double-strand breaks (DSBs), the NBS1 protein is required for activation of ATM. But ATM can also be activated in the absence of DNA damage. Treatment of cultured cells with hypotonic stress leads to the activation of ATM, presumably due to changes in chromatin structure. We have recently described a second ATM cofactor, ATMIN (ATM INteractor). ATMIN is dispensable for DSBs-induced ATM signalling, but ATM activation following hypotonic stress is mediated by ATMIN. While the biological role of ATM activation by DSBs and NBS1 is well established, the significance, if any, of ATM activation by ATMIN and changes in chromatin was up to now completely enigmatic.
ATM is required for class switch recombination (CSR) and the suppression of translocations in B cells. In order to determine whether ATMIN is required for any of the physiological functions of ATM, we generated a conditional knock-out mouse model for ATMIN. ATM signaling was dramatically reduced following osmotic stress in ATMIN-mutant B cells. ATMIN deficiency led to impaired CSR, and consequently ATMIN-mutant mice developed B cell lymphomas. Thus ablation of ATMIN resulted in a severe defect in ATM function. Our data strongly argue for the existence of a second NBS1-independent mode of ATM activation that is physiologically relevant. While a large amount of scientific effort has gone into characterising ATM signaling triggered by DSBs, essentially nothing is known about NBS1-independent ATM signaling. The experiments outlined in this proposal have the aim to identify and understand the molecular pathway of ATMIN-dependent ATM signaling.
Summary
ATM is the protein kinase that is mutated in the hereditary autosomal recessive disease ataxia telangiectasia (A-T). A-T patients display immune deficiencies, cancer predisposition and radiosensitivity. The molecular role of ATM is to respond to DNA damage by phosphorylating its substrates, thereby promoting repair of damage or arresting the cell cycle. Following the induction of double-strand breaks (DSBs), the NBS1 protein is required for activation of ATM. But ATM can also be activated in the absence of DNA damage. Treatment of cultured cells with hypotonic stress leads to the activation of ATM, presumably due to changes in chromatin structure. We have recently described a second ATM cofactor, ATMIN (ATM INteractor). ATMIN is dispensable for DSBs-induced ATM signalling, but ATM activation following hypotonic stress is mediated by ATMIN. While the biological role of ATM activation by DSBs and NBS1 is well established, the significance, if any, of ATM activation by ATMIN and changes in chromatin was up to now completely enigmatic.
ATM is required for class switch recombination (CSR) and the suppression of translocations in B cells. In order to determine whether ATMIN is required for any of the physiological functions of ATM, we generated a conditional knock-out mouse model for ATMIN. ATM signaling was dramatically reduced following osmotic stress in ATMIN-mutant B cells. ATMIN deficiency led to impaired CSR, and consequently ATMIN-mutant mice developed B cell lymphomas. Thus ablation of ATMIN resulted in a severe defect in ATM function. Our data strongly argue for the existence of a second NBS1-independent mode of ATM activation that is physiologically relevant. While a large amount of scientific effort has gone into characterising ATM signaling triggered by DSBs, essentially nothing is known about NBS1-independent ATM signaling. The experiments outlined in this proposal have the aim to identify and understand the molecular pathway of ATMIN-dependent ATM signaling.
Max ERC Funding
1 499 881 €
Duration
Start date: 2012-02-01, End date: 2018-01-31
Project acronym AtoFun
Project Atomic Scale Defects: Structure and Function
Researcher (PI) Felix HOFMANN
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Starting Grant (StG), PE5, ERC-2016-STG
Summary Atomic scale defects play a key role in determining the behaviour of all crystalline materials, profoundly modifying mechanical, thermal and electrical properties. Many current technological applications make do with phenomenological descriptions of these effects; yet myriad intriguing questions about the fundamental link between defect structure and material function remain.
Transmission electron microscopy revolutionised the study of atomic scale defects by enabling their direct imaging. The novel coherent X-ray diffraction techniques developed in this project promise a similar advancement, making it possible to probe the strain fields that govern defect interactions in 3D with high spatial resolution (<10 nm). They will allow us to clarify the effect of impurities and retained gas on dislocation strain fields, shedding light on opportunities to engineer dislocation properties. The exceptional strain sensitivity of coherent diffraction will enable us to explore the fundamental mechanisms governing the behaviour of ion-implantation-induced point defects that are invisible to TEM. While we concentrate on dislocations and point defects, the new techniques will apply to all crystalline materials where defects are important. Our characterisation of defect structure will be combined with laser transient grating measurements of thermal transport changes due to specific defect populations. This unique multifaceted perspective of defect behaviour will transform our ability to devise modelling approaches linking defect structure to material function.
Our proof-of-concept results highlight the feasibility of this ambitious research project. It opens up a vast range of exciting possibilities to gain a deep, fundamental understanding of atomic scale defects and their effect on material function. This is an essential prerequisite for exploiting and engineering defects to enhance material properties.
Summary
Atomic scale defects play a key role in determining the behaviour of all crystalline materials, profoundly modifying mechanical, thermal and electrical properties. Many current technological applications make do with phenomenological descriptions of these effects; yet myriad intriguing questions about the fundamental link between defect structure and material function remain.
Transmission electron microscopy revolutionised the study of atomic scale defects by enabling their direct imaging. The novel coherent X-ray diffraction techniques developed in this project promise a similar advancement, making it possible to probe the strain fields that govern defect interactions in 3D with high spatial resolution (<10 nm). They will allow us to clarify the effect of impurities and retained gas on dislocation strain fields, shedding light on opportunities to engineer dislocation properties. The exceptional strain sensitivity of coherent diffraction will enable us to explore the fundamental mechanisms governing the behaviour of ion-implantation-induced point defects that are invisible to TEM. While we concentrate on dislocations and point defects, the new techniques will apply to all crystalline materials where defects are important. Our characterisation of defect structure will be combined with laser transient grating measurements of thermal transport changes due to specific defect populations. This unique multifaceted perspective of defect behaviour will transform our ability to devise modelling approaches linking defect structure to material function.
Our proof-of-concept results highlight the feasibility of this ambitious research project. It opens up a vast range of exciting possibilities to gain a deep, fundamental understanding of atomic scale defects and their effect on material function. This is an essential prerequisite for exploiting and engineering defects to enhance material properties.
Max ERC Funding
1 610 231 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym Autocat
Project Autocatalysis: A bottom-up approach to understanding the origins of life
Researcher (PI) Stephen Patrick Fletcher
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Consolidator Grant (CoG), PE5, ERC-2015-CoG
Summary "The origin of life is not well understood, and is one of the great remaining questions in science. Autocatalytic chemical reactions have been extensively studied with the aim of providing insight into the principles underlying living systems. In biology, organisms can be thought of as imperfect self-replicators, which produce closely related species, allowing for selection and evolution. Autocatalysis is also an important part of many other biological processes.
This project aims to develop new autocatalytic reactions where two simple chemical building blocks come together to give a more complex product, and then the product aggregates to give primitive cell-like structures or "protocells" such as micelles or vesicles. The protocells allow the starting materials to mix more efficiently, speeding up the reaction in time and giving rise to complex behaviour of the protocells. These reactions will serve as models that I hope will contribute to understanding how cell-like systems can emerge from simpler chemicals and be relevant to how life started on earth.
This project will give the opportunity to study chemical systems that may be able to evolve in time, allow development of useful chemical models of important biological processes, and provide ‘bottom-up’ approaches to synthetic biology. This research will potential allow the study evolution in a new ways, develop technology useful to a number of scientific fields, and potentially shed light on the processes that allowed chemistry to become biology on the primitive Earth."
Summary
"The origin of life is not well understood, and is one of the great remaining questions in science. Autocatalytic chemical reactions have been extensively studied with the aim of providing insight into the principles underlying living systems. In biology, organisms can be thought of as imperfect self-replicators, which produce closely related species, allowing for selection and evolution. Autocatalysis is also an important part of many other biological processes.
This project aims to develop new autocatalytic reactions where two simple chemical building blocks come together to give a more complex product, and then the product aggregates to give primitive cell-like structures or "protocells" such as micelles or vesicles. The protocells allow the starting materials to mix more efficiently, speeding up the reaction in time and giving rise to complex behaviour of the protocells. These reactions will serve as models that I hope will contribute to understanding how cell-like systems can emerge from simpler chemicals and be relevant to how life started on earth.
This project will give the opportunity to study chemical systems that may be able to evolve in time, allow development of useful chemical models of important biological processes, and provide ‘bottom-up’ approaches to synthetic biology. This research will potential allow the study evolution in a new ways, develop technology useful to a number of scientific fields, and potentially shed light on the processes that allowed chemistry to become biology on the primitive Earth."
Max ERC Funding
2 278 073 €
Duration
Start date: 2016-05-01, End date: 2021-04-30
Project acronym AVIANEGG
Project Evolutionary genetics in a ‘classical’ avian study system by high throughput transcriptome sequencing and SNP genotyping
Researcher (PI) Jon Slate
Host Institution (HI) THE UNIVERSITY OF SHEFFIELD
Call Details Starting Grant (StG), LS5, ERC-2007-StG
Summary Long-term studies of free-living vertebrate populations have proved a rich resource for understanding evolutionary and ecological processes, because individuals’ life histories can be measured by tracking them from birth/hatching through to death. In recent years the ‘animal model’ has been applied to pedigreed long-term study populations with great success, dramatically advancing our understanding of quantitative genetic parameters such as heritabilities, genetic correlations and plasticities of traits that are relevant to microevolutionary responses to environmental change. Unfortunately, quantitative genetic approaches have one major drawback – they cannot identify the actual genes responsible for genetic variation. Therefore, it is impossible to link evolutionary responses to a changing environment to molecular genetic variation, making our picture of the process incomplete. Many of the best long-term studies have been conducted in passerine birds. Unfortunately genomics resources are only available for two model avian species, and are absent for bird species that are studied in the wild. I will fill this gap by exploiting recent advances in genomics technology to sequence the entire transcriptome of the longest running study of wild birds – the great tit population in Wytham Woods, Oxford. Having identified most of the sequence variation in the great tit transcriptome, I will then genotype all birds for whom phenotype records and blood samples are available This will be, by far, the largest phenotype-genotype dataset of any free-living vertebrate population. I will then use gene mapping techniques to identify genes and genomic regions responsible for variation in a number of key traits such as lifetime recruitment, clutch size and breeding/laying date. This will result in a greater understanding, at the molecular level, how microevolutionary change can arise (or be constrained).
Summary
Long-term studies of free-living vertebrate populations have proved a rich resource for understanding evolutionary and ecological processes, because individuals’ life histories can be measured by tracking them from birth/hatching through to death. In recent years the ‘animal model’ has been applied to pedigreed long-term study populations with great success, dramatically advancing our understanding of quantitative genetic parameters such as heritabilities, genetic correlations and plasticities of traits that are relevant to microevolutionary responses to environmental change. Unfortunately, quantitative genetic approaches have one major drawback – they cannot identify the actual genes responsible for genetic variation. Therefore, it is impossible to link evolutionary responses to a changing environment to molecular genetic variation, making our picture of the process incomplete. Many of the best long-term studies have been conducted in passerine birds. Unfortunately genomics resources are only available for two model avian species, and are absent for bird species that are studied in the wild. I will fill this gap by exploiting recent advances in genomics technology to sequence the entire transcriptome of the longest running study of wild birds – the great tit population in Wytham Woods, Oxford. Having identified most of the sequence variation in the great tit transcriptome, I will then genotype all birds for whom phenotype records and blood samples are available This will be, by far, the largest phenotype-genotype dataset of any free-living vertebrate population. I will then use gene mapping techniques to identify genes and genomic regions responsible for variation in a number of key traits such as lifetime recruitment, clutch size and breeding/laying date. This will result in a greater understanding, at the molecular level, how microevolutionary change can arise (or be constrained).
Max ERC Funding
1 560 770 €
Duration
Start date: 2008-10-01, End date: 2014-06-30
Project acronym AXONGROWTH
Project Systematic analysis of the molecular mechanisms underlying axon growth during development and following injury
Researcher (PI) Oren Schuldiner
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS5, ERC-2013-CoG
Summary Axon growth potential declines during development, contributing to the lack of effective regeneration in the adult central nervous system. What determines the intrinsic growth potential of neurites, and how such growth is regulated during development, disease and following injury is a fundamental question in neuroscience. Although multiple lines of evidence indicate that intrinsic growth capability is genetically encoded, its nature remains poorly defined. Neuronal remodeling of the Drosophila mushroom body offers a unique opportunity to study the mechanisms of various types of axon degeneration and growth. We have recently demonstrated that regrowth of axons following developmental pruning is not only distinct from initial outgrowth but also shares molecular similarities with regeneration following injury. In this proposal we combine state of the art tools from genomics, functional genetics and microscopy to perform a comprehensive study of the mechanisms underlying axon growth during development and following injury. First, we will combine genetic, biochemical and genomic studies to gain a mechanistic understanding of the developmental regrowth program. Next, we will perform extensive transcriptomic analyses and comparisons aimed at defining the genetic programs involved in initial axon growth, developmental regrowth, and regeneration following injury. Finally, we will harness the genetic power of Drosophila to perform a comprehensive functional analysis of genes and pathways, those previously known and new ones that we will discover, in various neurite growth paradigms. Importantly, these functional assays will be performed in the same organism, allowing us to use identical genetic mutations across our analyses. To this end, our identification of a new genetic program regulating developmental axon regrowth, together with emerging tools in genomics, places us in a unique position to gain a broad understanding of axon growth during development and following injury.
Summary
Axon growth potential declines during development, contributing to the lack of effective regeneration in the adult central nervous system. What determines the intrinsic growth potential of neurites, and how such growth is regulated during development, disease and following injury is a fundamental question in neuroscience. Although multiple lines of evidence indicate that intrinsic growth capability is genetically encoded, its nature remains poorly defined. Neuronal remodeling of the Drosophila mushroom body offers a unique opportunity to study the mechanisms of various types of axon degeneration and growth. We have recently demonstrated that regrowth of axons following developmental pruning is not only distinct from initial outgrowth but also shares molecular similarities with regeneration following injury. In this proposal we combine state of the art tools from genomics, functional genetics and microscopy to perform a comprehensive study of the mechanisms underlying axon growth during development and following injury. First, we will combine genetic, biochemical and genomic studies to gain a mechanistic understanding of the developmental regrowth program. Next, we will perform extensive transcriptomic analyses and comparisons aimed at defining the genetic programs involved in initial axon growth, developmental regrowth, and regeneration following injury. Finally, we will harness the genetic power of Drosophila to perform a comprehensive functional analysis of genes and pathways, those previously known and new ones that we will discover, in various neurite growth paradigms. Importantly, these functional assays will be performed in the same organism, allowing us to use identical genetic mutations across our analyses. To this end, our identification of a new genetic program regulating developmental axon regrowth, together with emerging tools in genomics, places us in a unique position to gain a broad understanding of axon growth during development and following injury.
Max ERC Funding
2 000 000 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym AXONSURVIVAL
Project Axon survival: the role of protein synthesis
Researcher (PI) Christine Elizabeth Holt
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Advanced Grant (AdG), LS5, ERC-2012-ADG_20120314
Summary Neurons make long-distance connections with synaptic targets via axons. These axons survive throughout the lifetime of an organism, often many years in mammals, yet how axons are maintained is not fully understood. Recently, we provided in vivo evidence that local mRNA translation in mature axons is required for their maintenance. This new finding, along with in vitro work from other groups, indicates that promoting axonal protein synthesis is a key mechanism by which trophic factors act to prevent axon degeneration. Here we propose a program of research to investigate the importance of ribosomal proteins (RPs) in axon maintenance and degeneration. The rationale for this is fourfold. First, recent genome-wide studies of axonal transcriptomes have revealed that protein synthesis (including RP mRNAs) is the highest functional category in several neuronal types. Second, some RPs have evolved extra-ribosomal functions that include signalling, such as 67LR which acts both as a cell surface receptor for laminin and as a RP. Third, mutations in different RPs in vertebrates cause unexpectedly specific defects, such as the loss of optic axons. Fourth, preliminary results show that RP mRNAs are translated in optic axons in response to trophic factors. Collectively these findings lead us to propose that locally synthesized RPs play a role in axon maintenance through either ribosomal or extra-ribosomal function. To pursue this proposal, we will perform unbiased screens and functional assays using an array of experimental approaches and animal models. By gaining an understanding of how local RP synthesis contributes to axon survival, our studies have the potential to provide novel insights into how components conventionally associated with a housekeeping role (translation) are linked to axon degeneration. Our findings could provide new directions for developing therapeutic tools for neurodegenerative disorders and may have an impact on more diverse areas of biology and disease.
Summary
Neurons make long-distance connections with synaptic targets via axons. These axons survive throughout the lifetime of an organism, often many years in mammals, yet how axons are maintained is not fully understood. Recently, we provided in vivo evidence that local mRNA translation in mature axons is required for their maintenance. This new finding, along with in vitro work from other groups, indicates that promoting axonal protein synthesis is a key mechanism by which trophic factors act to prevent axon degeneration. Here we propose a program of research to investigate the importance of ribosomal proteins (RPs) in axon maintenance and degeneration. The rationale for this is fourfold. First, recent genome-wide studies of axonal transcriptomes have revealed that protein synthesis (including RP mRNAs) is the highest functional category in several neuronal types. Second, some RPs have evolved extra-ribosomal functions that include signalling, such as 67LR which acts both as a cell surface receptor for laminin and as a RP. Third, mutations in different RPs in vertebrates cause unexpectedly specific defects, such as the loss of optic axons. Fourth, preliminary results show that RP mRNAs are translated in optic axons in response to trophic factors. Collectively these findings lead us to propose that locally synthesized RPs play a role in axon maintenance through either ribosomal or extra-ribosomal function. To pursue this proposal, we will perform unbiased screens and functional assays using an array of experimental approaches and animal models. By gaining an understanding of how local RP synthesis contributes to axon survival, our studies have the potential to provide novel insights into how components conventionally associated with a housekeeping role (translation) are linked to axon degeneration. Our findings could provide new directions for developing therapeutic tools for neurodegenerative disorders and may have an impact on more diverse areas of biology and disease.
Max ERC Funding
2 426 573 €
Duration
Start date: 2013-03-01, End date: 2018-09-30
Project acronym BIO-H-BORROW
Project Biocatalytic Amine Synthesis via Hydrogen Borrowing
Researcher (PI) Nicholas TURNER
Host Institution (HI) THE UNIVERSITY OF MANCHESTER
Call Details Advanced Grant (AdG), PE5, ERC-2016-ADG
Summary Amine containing compounds are ubiquitous in everyday life and find applications ranging from polymers to pharmaceuticals. The vast majority of amines are synthetic and manufactured on large scale which creates waste as well as requiring high temperatures and pressures. The increasing availability of biocatalysts, together with an understanding of how they can be used in organic synthesis (biocatalytic retrosynthesis), has stimulated chemists to consider new ways of making target molecules. In this context, the iterative construction of C-N bonds via biocatalytic hydrogen borrowing represents a powerful and unexplored way to synthesise a wide range of target amine molecules in an efficient manner. Hydrogen borrowing involves telescoping redox neutral reactions together using only catalytic amounts of hydrogen.
In this project we will engineer the three key target biocatalysts (reductive aminase, amine dehydrogenase, alcohol dehydrogenase) required for biocatalytic hydrogen borrowing such that they possess the required regio-, chemo- and stereo-selectivity for practical application. Recently discovered reductive aminases (RedAms) and amine dehydrogenases (AmDHs) will be engineered for enantioselective coupling of alcohols (1o, 2o) with ammonia/amines (1o, 2o, 3o) under redox neutral conditions. Alcohol dehydrogenases will be engineered for low enantioselectivity. Hydrogen borrowing requires mutually compatible cofactors shared by two enzymes and in some cases will require redesign of cofactor specificity. Thereafter we shall develop conditions for the combined use of these biocatalysts under hydrogen borrowing conditions (catalytic NADH, NADPH), to enable the conversion of simple and sustainable feedstocks (alcohols) into amines using ammonia as the nitrogen source.
The main deliverables of BIO-H-BORROW will be a set of novel engineered biocatalysts together with redox neutral cascades for the synthesis of amine products from inexpensive and renewable precursors.
Summary
Amine containing compounds are ubiquitous in everyday life and find applications ranging from polymers to pharmaceuticals. The vast majority of amines are synthetic and manufactured on large scale which creates waste as well as requiring high temperatures and pressures. The increasing availability of biocatalysts, together with an understanding of how they can be used in organic synthesis (biocatalytic retrosynthesis), has stimulated chemists to consider new ways of making target molecules. In this context, the iterative construction of C-N bonds via biocatalytic hydrogen borrowing represents a powerful and unexplored way to synthesise a wide range of target amine molecules in an efficient manner. Hydrogen borrowing involves telescoping redox neutral reactions together using only catalytic amounts of hydrogen.
In this project we will engineer the three key target biocatalysts (reductive aminase, amine dehydrogenase, alcohol dehydrogenase) required for biocatalytic hydrogen borrowing such that they possess the required regio-, chemo- and stereo-selectivity for practical application. Recently discovered reductive aminases (RedAms) and amine dehydrogenases (AmDHs) will be engineered for enantioselective coupling of alcohols (1o, 2o) with ammonia/amines (1o, 2o, 3o) under redox neutral conditions. Alcohol dehydrogenases will be engineered for low enantioselectivity. Hydrogen borrowing requires mutually compatible cofactors shared by two enzymes and in some cases will require redesign of cofactor specificity. Thereafter we shall develop conditions for the combined use of these biocatalysts under hydrogen borrowing conditions (catalytic NADH, NADPH), to enable the conversion of simple and sustainable feedstocks (alcohols) into amines using ammonia as the nitrogen source.
The main deliverables of BIO-H-BORROW will be a set of novel engineered biocatalysts together with redox neutral cascades for the synthesis of amine products from inexpensive and renewable precursors.
Max ERC Funding
2 337 548 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym BIOINCMED
Project Bioinorganic Chemistry for the Design of New Medicines
Researcher (PI) Peter John Sadler
Host Institution (HI) THE UNIVERSITY OF WARWICK
Call Details Advanced Grant (AdG), PE5, ERC-2009-AdG
Summary Bioinorganic chemistry is a rapidly expanding area of research, but the potential for the therapeutic application of metal complexes is highly underdeveloped. The basic principles required to guide the development of metal-containing therapeutic agents are lacking, despite the unique therapeutic opportunities which they offer. It is the goal of the proposed research to establish basic principles of medicinal coordination chemistry of metals that will allow the rational screening of future metallopharmaceuticals. We propose to utilize the power of inorganic chemistry to provide new knowledge of and new approaches for intervention in biological systems. This will be based on improved understanding of reactions of metal complexes under physiological conditions, on improving the specificity of their interactions, and gaining control over the potential toxicity of synthetic metal complexes. The research programme is highly interdisciplinary involving chemistry, physics, biology and pharmacology, with potential for the discovery of truly novel medicines, especially for the treatment of diseases and conditions which are currently intractable, such as cancer. The challenging and ambitious goals of the present work involve transition metal complexes with novel chemical and biochemical mechanisms of action. They will contain novel features which allow them (i) to be selectively activated by light in cells, or (ii) to be activated by a structural transition, or (ii) exhibit catalytic activity in cells. This ground-breaking research potentially has a very high impact and is based on recent discoveries in the applicant s laboratory. A feature of the programme is the use of state-of-the-art-and-beyond methodology to advance knowledge of medicinal metal coordination chemistry.
Summary
Bioinorganic chemistry is a rapidly expanding area of research, but the potential for the therapeutic application of metal complexes is highly underdeveloped. The basic principles required to guide the development of metal-containing therapeutic agents are lacking, despite the unique therapeutic opportunities which they offer. It is the goal of the proposed research to establish basic principles of medicinal coordination chemistry of metals that will allow the rational screening of future metallopharmaceuticals. We propose to utilize the power of inorganic chemistry to provide new knowledge of and new approaches for intervention in biological systems. This will be based on improved understanding of reactions of metal complexes under physiological conditions, on improving the specificity of their interactions, and gaining control over the potential toxicity of synthetic metal complexes. The research programme is highly interdisciplinary involving chemistry, physics, biology and pharmacology, with potential for the discovery of truly novel medicines, especially for the treatment of diseases and conditions which are currently intractable, such as cancer. The challenging and ambitious goals of the present work involve transition metal complexes with novel chemical and biochemical mechanisms of action. They will contain novel features which allow them (i) to be selectively activated by light in cells, or (ii) to be activated by a structural transition, or (ii) exhibit catalytic activity in cells. This ground-breaking research potentially has a very high impact and is based on recent discoveries in the applicant s laboratory. A feature of the programme is the use of state-of-the-art-and-beyond methodology to advance knowledge of medicinal metal coordination chemistry.
Max ERC Funding
1 565 397 €
Duration
Start date: 2010-07-01, End date: 2015-12-31
Project acronym BioMet
Project Selective Functionalization of Saturated Hydrocarbons
Researcher (PI) Ilan MAREK
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Advanced Grant (AdG), PE5, ERC-2017-ADG
Summary Despite that C–H functionalization represents a paradigm shift from the standard logic of organic synthesis, the selective activation of non-functionalized alkanes has puzzled chemists for centuries and is always referred to one of the remaining major challenges in chemical sciences. Alkanes are inert compounds representing the major constituents of natural gas and petroleum. Converting these cheap and widely available hydrocarbon feedstocks into added-value intermediates would tremendously affect the field of chemistry. For long saturated hydrocarbons, one must distinguish between non-equivalent but chemically very similar alkane substrate C−H bonds, and for functionalization at the terminus position, one must favor activation of the stronger, primary C−H bonds at the expense of weaker and numerous secondary C-H bonds. The goal of this work is to develop a general principle in organic synthesis for the preparation of a wide variety of more complex molecular architectures from saturated hydrocarbons. In our approach, the alkane will first be transformed into an alkene that will subsequently be engaged in a metal-catalyzed hydrometalation/migration sequence. The first step of the sequence, ideally represented by the removal of two hydrogen atoms, will be performed by the use of a mutated strain of Rhodococcus. The position and geometry of the formed double bond has no effect on the second step of the reaction as the metal-catalyzed hydrometalation/migration will isomerize the double bond along the carbon skeleton to selectively produce the primary organometallic species. Trapping the resulting organometallic derivatives with a large variety of electrophiles will provide the desired functionalized alkane. This work will lead to the invention of new, selective and efficient processes for the utilization of simple hydrocarbons and valorize the synthetic potential of raw hydrocarbon feedstock for the environmentally benign production of new compounds and new materials.
Summary
Despite that C–H functionalization represents a paradigm shift from the standard logic of organic synthesis, the selective activation of non-functionalized alkanes has puzzled chemists for centuries and is always referred to one of the remaining major challenges in chemical sciences. Alkanes are inert compounds representing the major constituents of natural gas and petroleum. Converting these cheap and widely available hydrocarbon feedstocks into added-value intermediates would tremendously affect the field of chemistry. For long saturated hydrocarbons, one must distinguish between non-equivalent but chemically very similar alkane substrate C−H bonds, and for functionalization at the terminus position, one must favor activation of the stronger, primary C−H bonds at the expense of weaker and numerous secondary C-H bonds. The goal of this work is to develop a general principle in organic synthesis for the preparation of a wide variety of more complex molecular architectures from saturated hydrocarbons. In our approach, the alkane will first be transformed into an alkene that will subsequently be engaged in a metal-catalyzed hydrometalation/migration sequence. The first step of the sequence, ideally represented by the removal of two hydrogen atoms, will be performed by the use of a mutated strain of Rhodococcus. The position and geometry of the formed double bond has no effect on the second step of the reaction as the metal-catalyzed hydrometalation/migration will isomerize the double bond along the carbon skeleton to selectively produce the primary organometallic species. Trapping the resulting organometallic derivatives with a large variety of electrophiles will provide the desired functionalized alkane. This work will lead to the invention of new, selective and efficient processes for the utilization of simple hydrocarbons and valorize the synthetic potential of raw hydrocarbon feedstock for the environmentally benign production of new compounds and new materials.
Max ERC Funding
2 499 375 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym BIOMOF
Project Biomineral-inspired growth and processing of metal-organic frameworks
Researcher (PI) Darren Bradshaw
Host Institution (HI) UNIVERSITY OF SOUTHAMPTON
Call Details Starting Grant (StG), PE5, ERC-2010-StG_20091028
Summary This ERC-StG proposal, BIOMOF, outlines a dual strategy for the growth and processing of porous metal-organic framework (MOF) materials, inspired by the interfacial interactions that characterise highly controlled biomineralisation processes. The aim is to prepare MOF (bio)-composite materials of hierarchical structure and multi-modal functionality to address key societal challenges in healthcare, catalysis and energy. In order for MOFs to reach their full potential, a transformative approach to their growth, and in particular their processability, is required since the insoluble macroscopic micron-sized crystals resulting from conventional syntheses are unsuitable for many applications. The BIOMOF project defines chemically flexible routes to MOFs under mild conditions, where the added value with respect to wide-ranging experimental procedures for the growth and processing of crystalline controllably nanoscale MOF materials with tunable structure and functionality that display significant porosity for wide-ranging applications is extremely high. Theme 1 exploits protein vesicles and abundant biopolymer matrices for the confined growth of soluble nanoscale MOFs for high-end biomedical applications such as cell imaging and targeted drug delivery, whereas theme 2 focuses on the cost-effective preparation of hierarchically porous MOF composites over several length scales, of relevance to bulk industrial applications such as sustainable catalysis, separations and gas-storage. This diverse yet complementary range of applications arising simply from the way the MOF is processed, coupled with the versatile structural and physical properties of MOFs themselves indicates strongly that the BIOMOF concept is a powerful convergent new approach to applied materials chemistry.
Summary
This ERC-StG proposal, BIOMOF, outlines a dual strategy for the growth and processing of porous metal-organic framework (MOF) materials, inspired by the interfacial interactions that characterise highly controlled biomineralisation processes. The aim is to prepare MOF (bio)-composite materials of hierarchical structure and multi-modal functionality to address key societal challenges in healthcare, catalysis and energy. In order for MOFs to reach their full potential, a transformative approach to their growth, and in particular their processability, is required since the insoluble macroscopic micron-sized crystals resulting from conventional syntheses are unsuitable for many applications. The BIOMOF project defines chemically flexible routes to MOFs under mild conditions, where the added value with respect to wide-ranging experimental procedures for the growth and processing of crystalline controllably nanoscale MOF materials with tunable structure and functionality that display significant porosity for wide-ranging applications is extremely high. Theme 1 exploits protein vesicles and abundant biopolymer matrices for the confined growth of soluble nanoscale MOFs for high-end biomedical applications such as cell imaging and targeted drug delivery, whereas theme 2 focuses on the cost-effective preparation of hierarchically porous MOF composites over several length scales, of relevance to bulk industrial applications such as sustainable catalysis, separations and gas-storage. This diverse yet complementary range of applications arising simply from the way the MOF is processed, coupled with the versatile structural and physical properties of MOFs themselves indicates strongly that the BIOMOF concept is a powerful convergent new approach to applied materials chemistry.
Max ERC Funding
1 492 970 €
Duration
Start date: 2010-11-01, End date: 2015-10-31
Project acronym BIONICS
Project Bio-Inspired Routes for Controlling the Structure and Properties of Materials: Reusing proven tricks on new materials
Researcher (PI) Boaz Pokroy
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), PE5, ERC-2013-StG
Summary "In the course of biomineralization, organisms produce a large variety of functional biogenic crystals that exhibit fascinating mechanical, optical, magnetic and other characteristics. More specifically, when living organisms grow crystals they can effectively control polymorph selection as well as the crystal morphology, shape, and even atomic structure. Materials existing in nature have extraordinary and specific functions, yet the materials employed in nature are quite different from those engineers would select.
I propose to emulate specific strategies used by organisms in forming structural biogenic crystals, and to apply these strategies biomimetically so as to form new structural materials with new properties and characteristics. This bio-inspired approach will involve the adoption of three specific biological strategies. We believe that this procedure will open up new ways to control the structure and properties of smart materials.
The three bio-inspired strategies that we will utilize are:
(i) to control the short-range order of amorphous materials, making it possible to predetermine the polymorph obtained when they transform from the amorphous to the succeeding crystalline phase;
(ii) to control the morphology of single crystals of various functional materials so that they can have intricate and curved surfaces and yet maintain their single-crystal nature;
(iii) to entrap organic molecules into single crystals of functional materials so as to tailor and manipulate their electronic structure.
The proposed research has significant potential for opening up new routes for the formation of novel functional materials. Specifically, it will make it possible for us
(1) to produce single, intricately shaped crystals without the need to etch, drill or polish;
(2) to control the short-range order of amorphous materials and hence the polymorph of the successive crystalline phase;
(3) to tune the band gap of semiconductors via incorporation of tailored bio-molecules."
Summary
"In the course of biomineralization, organisms produce a large variety of functional biogenic crystals that exhibit fascinating mechanical, optical, magnetic and other characteristics. More specifically, when living organisms grow crystals they can effectively control polymorph selection as well as the crystal morphology, shape, and even atomic structure. Materials existing in nature have extraordinary and specific functions, yet the materials employed in nature are quite different from those engineers would select.
I propose to emulate specific strategies used by organisms in forming structural biogenic crystals, and to apply these strategies biomimetically so as to form new structural materials with new properties and characteristics. This bio-inspired approach will involve the adoption of three specific biological strategies. We believe that this procedure will open up new ways to control the structure and properties of smart materials.
The three bio-inspired strategies that we will utilize are:
(i) to control the short-range order of amorphous materials, making it possible to predetermine the polymorph obtained when they transform from the amorphous to the succeeding crystalline phase;
(ii) to control the morphology of single crystals of various functional materials so that they can have intricate and curved surfaces and yet maintain their single-crystal nature;
(iii) to entrap organic molecules into single crystals of functional materials so as to tailor and manipulate their electronic structure.
The proposed research has significant potential for opening up new routes for the formation of novel functional materials. Specifically, it will make it possible for us
(1) to produce single, intricately shaped crystals without the need to etch, drill or polish;
(2) to control the short-range order of amorphous materials and hence the polymorph of the successive crystalline phase;
(3) to tune the band gap of semiconductors via incorporation of tailored bio-molecules."
Max ERC Funding
1 500 000 €
Duration
Start date: 2013-09-01, End date: 2018-08-31
Project acronym BOTTOM-UP_SYSCHEM
Project Systems Chemistry from Bottom Up: Switching, Gating and Oscillations in Non Enzymatic Peptide Networks
Researcher (PI) Gonen Ashkenasy
Host Institution (HI) BEN-GURION UNIVERSITY OF THE NEGEV
Call Details Starting Grant (StG), PE5, ERC-2010-StG_20091028
Summary The study of synthetic molecular networks is of fundamental importance for understanding the organizational principles of biological systems and may well be the key to unraveling the origins of life. In addition, such systems may be useful for parallel synthesis of molecules, implementation of catalysis via multi-step pathways, and as media for various applications in nano-medicine and nano-electronics. We have been involved recently in developing peptide-based replicating networks and revealed their dynamic characteristics. We argue here that the structural information embedded in the polypeptide chains is sufficiently rich to allow the construction of peptide 'Systems Chemistry', namely, to facilitate the use of replicating networks as cell-mimetics, featuring complex dynamic behavior. To bring this novel idea to reality, we plan to take a unique holistic approach by studying such networks both experimentally and via simulations, for elucidating basic-principles and towards applications in adjacent fields, such as molecular electronics. Towards realizing these aims, we will study three separate but inter-related objectives: (i) design and characterization of networks that react and rewire in response to external triggers, such as light, (ii) design of networks that operate via new dynamic rules of product formation that lead to oscillations, and (iii) exploitation of the molecular information gathered from the networks as means to control switching and gating in molecular electronic devices. We believe that achieving the project's objectives will be highly significant for the development of the arising field of Systems Chemistry, and in addition will provide valuable tools for studying related scientific fields, such as systems biology and molecular electronics.
Summary
The study of synthetic molecular networks is of fundamental importance for understanding the organizational principles of biological systems and may well be the key to unraveling the origins of life. In addition, such systems may be useful for parallel synthesis of molecules, implementation of catalysis via multi-step pathways, and as media for various applications in nano-medicine and nano-electronics. We have been involved recently in developing peptide-based replicating networks and revealed their dynamic characteristics. We argue here that the structural information embedded in the polypeptide chains is sufficiently rich to allow the construction of peptide 'Systems Chemistry', namely, to facilitate the use of replicating networks as cell-mimetics, featuring complex dynamic behavior. To bring this novel idea to reality, we plan to take a unique holistic approach by studying such networks both experimentally and via simulations, for elucidating basic-principles and towards applications in adjacent fields, such as molecular electronics. Towards realizing these aims, we will study three separate but inter-related objectives: (i) design and characterization of networks that react and rewire in response to external triggers, such as light, (ii) design of networks that operate via new dynamic rules of product formation that lead to oscillations, and (iii) exploitation of the molecular information gathered from the networks as means to control switching and gating in molecular electronic devices. We believe that achieving the project's objectives will be highly significant for the development of the arising field of Systems Chemistry, and in addition will provide valuable tools for studying related scientific fields, such as systems biology and molecular electronics.
Max ERC Funding
1 500 000 €
Duration
Start date: 2010-10-01, End date: 2015-09-30
Project acronym Brain circRNAs
Project Rounding the circle: Unravelling the biogenesis, function and mechanism of action of circRNAs in the Drosophila brain.
Researcher (PI) Sebastian Kadener
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Consolidator Grant (CoG), LS5, ERC-2014-CoG
Summary Tight regulation of RNA metabolism is essential for normal brain function. This includes co and post-transcriptional regulation, which are extremely prevalent in neurons. Recently, circular RNAs (circRNAs), a highly abundant new type of regulatory non-coding RNA have been found across the animal kingdom. Two of these RNAs have been shown to act as miRNA sponges but no function is known for the thousands of other circRNAs, indicating the existence of a widespread layer of previously unknown gene regulation.
The present proposal aims to comprehensively determine the role and mode of actions of circRNAs in gene expression and RNA metabolism in the fly brain. We will do so by studying their biogenesis, transport, and mechanism of action, as well as by determining the roles of circRNAs in neuronal function and behaviour. Briefly, we will: 1) identify factors involved in the biogenesis, localization, and stabilization of circRNAs; 2) determine neuro-developmental, molecular, neural and behavioural phenotypes associated with down or up regulation of specific circRNAs; 3) study the molecular mechanisms of action of circRNAs: identify circRNAs that work as miRNA sponges and determine whether circRNAs can encode proteins or act as signalling molecules and 4) perform mechanistic studies in order to determine cause-effect relationships between circRNA function and brain physiology and behaviour.
The present proposal will reveal the key pathways by which circRNAs control gene expression and influence neuronal function and behaviour. Therefore it will be one of the pioneer works in the study of this new and important area of research, which we predict will fundamentally transform the study of gene expression regulation in the brain
Summary
Tight regulation of RNA metabolism is essential for normal brain function. This includes co and post-transcriptional regulation, which are extremely prevalent in neurons. Recently, circular RNAs (circRNAs), a highly abundant new type of regulatory non-coding RNA have been found across the animal kingdom. Two of these RNAs have been shown to act as miRNA sponges but no function is known for the thousands of other circRNAs, indicating the existence of a widespread layer of previously unknown gene regulation.
The present proposal aims to comprehensively determine the role and mode of actions of circRNAs in gene expression and RNA metabolism in the fly brain. We will do so by studying their biogenesis, transport, and mechanism of action, as well as by determining the roles of circRNAs in neuronal function and behaviour. Briefly, we will: 1) identify factors involved in the biogenesis, localization, and stabilization of circRNAs; 2) determine neuro-developmental, molecular, neural and behavioural phenotypes associated with down or up regulation of specific circRNAs; 3) study the molecular mechanisms of action of circRNAs: identify circRNAs that work as miRNA sponges and determine whether circRNAs can encode proteins or act as signalling molecules and 4) perform mechanistic studies in order to determine cause-effect relationships between circRNA function and brain physiology and behaviour.
The present proposal will reveal the key pathways by which circRNAs control gene expression and influence neuronal function and behaviour. Therefore it will be one of the pioneer works in the study of this new and important area of research, which we predict will fundamentally transform the study of gene expression regulation in the brain
Max ERC Funding
1 971 750 €
Duration
Start date: 2016-02-01, End date: 2021-01-31
Project acronym BrainEnergy
Project Control of cerebral blood flow by capillary pericytes in health and disease
Researcher (PI) David ATTWELL
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Advanced Grant (AdG), LS5, ERC-2016-ADG
Summary Pericytes, located at intervals along capillaries, have recently been revealed as major controllers of brain blood flow. Normally, they dilate capillaries in response to neuronal activity, increasing local blood flow and energy supply. But in pathology they have a more sinister role. After artery block causes a stroke, the brain suffers from the so-called “no-reflow” phenomenon - a failure to fully reperfuse capillaries, even after the upstream occluded artery has been reperfused successfully. The resulting long-lasting decrease of energy supply damages neurons. I have shown that a major cause of no-reflow lies in pericytes: during ischaemia they constrict and then die in rigor. This reduces capillary diameter and blood flow, and probably degrades blood-brain barrier function. However, despite their crucial role in regulating blood flow physiologically and in pathology, little is known about the mechanisms by which pericytes function.
By using blood vessel imaging, patch-clamping, two-photon imaging, optogenetics, immunohistochemistry, mathematical modelling, and live human tissue obtained from neurosurgery, this programme of research will:
(i) define the signalling mechanisms controlling capillary constriction and dilation in health and disease;
(ii) identify the relative contributions of neurons, astrocytes and microglia to regulating pericyte tone;
(iii) develop approaches to preventing brain pericyte constriction and death during ischaemia;
(iv) define how pericyte constriction of capillaries and pericyte death contribute to Alzheimer’s disease;
(v) extend these results from rodent brain to human brain pericytes as a prelude to developing therapies.
The diseases to which pericytes contribute include stroke, spinal cord injury, diabetes and Alzheimer’s disease. These all have an enormous economic impact, as well as causing great suffering for patients and their carers. This work will provide novel therapeutic approaches for treating these diseases.
Summary
Pericytes, located at intervals along capillaries, have recently been revealed as major controllers of brain blood flow. Normally, they dilate capillaries in response to neuronal activity, increasing local blood flow and energy supply. But in pathology they have a more sinister role. After artery block causes a stroke, the brain suffers from the so-called “no-reflow” phenomenon - a failure to fully reperfuse capillaries, even after the upstream occluded artery has been reperfused successfully. The resulting long-lasting decrease of energy supply damages neurons. I have shown that a major cause of no-reflow lies in pericytes: during ischaemia they constrict and then die in rigor. This reduces capillary diameter and blood flow, and probably degrades blood-brain barrier function. However, despite their crucial role in regulating blood flow physiologically and in pathology, little is known about the mechanisms by which pericytes function.
By using blood vessel imaging, patch-clamping, two-photon imaging, optogenetics, immunohistochemistry, mathematical modelling, and live human tissue obtained from neurosurgery, this programme of research will:
(i) define the signalling mechanisms controlling capillary constriction and dilation in health and disease;
(ii) identify the relative contributions of neurons, astrocytes and microglia to regulating pericyte tone;
(iii) develop approaches to preventing brain pericyte constriction and death during ischaemia;
(iv) define how pericyte constriction of capillaries and pericyte death contribute to Alzheimer’s disease;
(v) extend these results from rodent brain to human brain pericytes as a prelude to developing therapies.
The diseases to which pericytes contribute include stroke, spinal cord injury, diabetes and Alzheimer’s disease. These all have an enormous economic impact, as well as causing great suffering for patients and their carers. This work will provide novel therapeutic approaches for treating these diseases.
Max ERC Funding
2 499 954 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym BrainNanoFlow
Project Nanoscale dynamics in the extracellular space of the brain in vivo
Researcher (PI) Juan Alberto VARELA
Host Institution (HI) THE UNIVERSITY COURT OF THE UNIVERSITY OF ST ANDREWS
Call Details Starting Grant (StG), LS5, ERC-2018-STG
Summary Aggregates of proteins such as amyloid-beta and alpha-synuclein circulate the extracellular space of the brain (ECS) and are thought to be key players in the development of neurodegenerative diseases. The clearance of these aggregates (among other toxic metabolites) is a fundamental physiological feature of the brain which is poorly understood due to the lack of techniques to study the nanoscale organisation of the ECS. Exciting advances in this field have recently shown that clearance is enhanced during sleep due to a major volume change in the ECS, facilitating the flow of the interstitial fluid. However, this process has only been characterised at a low spatial resolution while the physiological changes occur at the nanoscale. The recently proposed “glymphatic” pathway still remains controversial, as there are no techniques capable of distinguishing between diffusion and bulk flow in the ECS of living animals. Understanding these processes at a higher spatial resolution requires the development of single-molecule imaging techniques that can study the brain in living animals. Taking advantage of the strategies I have recently developed to target single-molecules in the brain in vivo with nanoparticles, we will do “nanoscopy” in living animals. Our proposal will test the glymphatic pathway at the spatial scale in which events happen, and explore how sleep and wake cycles alter the ECS and the diffusion of receptors in neuronal plasma membrane. Overall, BrainNanoFlow aims to understand how nanoscale changes in the ECS facilitate clearance of protein aggregates. We will also provide new insights to the pathological consequences of impaired clearance, focusing on the interactions between these aggregates and their putative receptors. Being able to perform single-molecule studies in vivo in the brain will be a major breakthrough in neurobiology, making possible the study of physiological and pathological processes that cannot be studied in simpler brain preparations.
Summary
Aggregates of proteins such as amyloid-beta and alpha-synuclein circulate the extracellular space of the brain (ECS) and are thought to be key players in the development of neurodegenerative diseases. The clearance of these aggregates (among other toxic metabolites) is a fundamental physiological feature of the brain which is poorly understood due to the lack of techniques to study the nanoscale organisation of the ECS. Exciting advances in this field have recently shown that clearance is enhanced during sleep due to a major volume change in the ECS, facilitating the flow of the interstitial fluid. However, this process has only been characterised at a low spatial resolution while the physiological changes occur at the nanoscale. The recently proposed “glymphatic” pathway still remains controversial, as there are no techniques capable of distinguishing between diffusion and bulk flow in the ECS of living animals. Understanding these processes at a higher spatial resolution requires the development of single-molecule imaging techniques that can study the brain in living animals. Taking advantage of the strategies I have recently developed to target single-molecules in the brain in vivo with nanoparticles, we will do “nanoscopy” in living animals. Our proposal will test the glymphatic pathway at the spatial scale in which events happen, and explore how sleep and wake cycles alter the ECS and the diffusion of receptors in neuronal plasma membrane. Overall, BrainNanoFlow aims to understand how nanoscale changes in the ECS facilitate clearance of protein aggregates. We will also provide new insights to the pathological consequences of impaired clearance, focusing on the interactions between these aggregates and their putative receptors. Being able to perform single-molecule studies in vivo in the brain will be a major breakthrough in neurobiology, making possible the study of physiological and pathological processes that cannot be studied in simpler brain preparations.
Max ERC Funding
1 552 948 €
Duration
Start date: 2018-12-01, End date: 2023-11-30
Project acronym BRAINPOWER
Project Brain energy supply and the consequences of its failure
Researcher (PI) David Ian Attwell
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Advanced Grant (AdG), LS5, ERC-2009-AdG
Summary Energy, supplied in the form of oxygen and glucose in the blood, is essential for the brain s cognitive power. Failure of the energy supply to the nervous system underlies the mental and physical disability occurring in a wide range of economically important neurological disorders, such as stroke, spinal cord injury and cerebral palsy. Using a combination of two-photon imaging, electrophysiological, molecular and transgenic approaches, I will investigate the control of brain energy supply at the vascular level, and at the level of individual neurons and glial cells, and study the deleterious consequences for the neurons, glia and vasculature of a failure of brain energy supply. The work will focus on the following fundamental issues: A. Vascular control of the brain energy supply (1) How important is control of energy supply at the capillary level, by pericytes? (2) Which synapses control blood flow (and thus generate functional imaging signals) in the cortex? B. Neuronal and glial control of brain energy supply (3) How is grey matter neuronal activity powered? (4) How is the white matter supplied with energy? C. The pathological consequences of a loss of brain energy supply (5) How does a fall of energy supply cause neurotoxic glutamate release? (6) How similar are events in the grey and white matter in energy deprivation conditions? (7) How does a transient loss of energy supply affect blood flow regulation? (8) How does brain energy use change after a period without energy supply? Together this work will significantly advance our understanding of how the energy supply to neurons and glia is regulated in normal conditions, and how the loss of the energy supply causes disorders which consume more than 5% of the costs of European health services (5% of ~1000 billion euro/year).
Summary
Energy, supplied in the form of oxygen and glucose in the blood, is essential for the brain s cognitive power. Failure of the energy supply to the nervous system underlies the mental and physical disability occurring in a wide range of economically important neurological disorders, such as stroke, spinal cord injury and cerebral palsy. Using a combination of two-photon imaging, electrophysiological, molecular and transgenic approaches, I will investigate the control of brain energy supply at the vascular level, and at the level of individual neurons and glial cells, and study the deleterious consequences for the neurons, glia and vasculature of a failure of brain energy supply. The work will focus on the following fundamental issues: A. Vascular control of the brain energy supply (1) How important is control of energy supply at the capillary level, by pericytes? (2) Which synapses control blood flow (and thus generate functional imaging signals) in the cortex? B. Neuronal and glial control of brain energy supply (3) How is grey matter neuronal activity powered? (4) How is the white matter supplied with energy? C. The pathological consequences of a loss of brain energy supply (5) How does a fall of energy supply cause neurotoxic glutamate release? (6) How similar are events in the grey and white matter in energy deprivation conditions? (7) How does a transient loss of energy supply affect blood flow regulation? (8) How does brain energy use change after a period without energy supply? Together this work will significantly advance our understanding of how the energy supply to neurons and glia is regulated in normal conditions, and how the loss of the energy supply causes disorders which consume more than 5% of the costs of European health services (5% of ~1000 billion euro/year).
Max ERC Funding
2 499 947 €
Duration
Start date: 2010-04-01, End date: 2016-03-31
Project acronym BrainReadFBPredCode
Project Brain reading of contextual feedback and predictions
Researcher (PI) Lars Muckli
Host Institution (HI) UNIVERSITY OF GLASGOW
Call Details Starting Grant (StG), LS5, ERC-2012-StG_20111109
Summary We are currently witnessing a paradigm shift in our understanding of human brain function, moving towards a clearer description of cortical processing. Sensory systems are no longer considered as 'passively recording' but rather as dynamically anticipating and adapting to the rapidly changing environment. These new ideas are encompassed in the predictive coding framework, and indeed in a unifying theory of the brain (Friston, 2010). In terms of brain computation, a predictive model is created in higher cortical areas and communicated to lower sensory areas through feedback connections. Based on my pioneering research I propose experiments that are capable of ‘brain-reading’ cortical feedback– which would contribute invaluable data to theoretical frameworks.
The proposed research project will advance our understanding of ongoing brain activity, contextual processing, and cortical feedback - contributing to what is known about general cortical functions. By providing new insights as to the information content of cortical feedback, the proposal will fill one of the most important gaps in today’s knowledge about brain function. Friston’s unifying theory of the brain (Friston, 2010) and contemporary models of the predictive-coding framework (Hawkins and Blakeslee, 2004;Mumford, 1992;Rao and Ballard, 1999) assign feedback processing an essential role in cortical processing. Compared to feedforward information processing, our knowledge about feedback processing is in its infancy. The proposal introduces parametric and explorative brain reading designs to investigate this feedback processing. The chief goal of my proposal will be precision measures of cortical feedback, and a more ambitious objective is to read mental images and inner thoughts.
Summary
We are currently witnessing a paradigm shift in our understanding of human brain function, moving towards a clearer description of cortical processing. Sensory systems are no longer considered as 'passively recording' but rather as dynamically anticipating and adapting to the rapidly changing environment. These new ideas are encompassed in the predictive coding framework, and indeed in a unifying theory of the brain (Friston, 2010). In terms of brain computation, a predictive model is created in higher cortical areas and communicated to lower sensory areas through feedback connections. Based on my pioneering research I propose experiments that are capable of ‘brain-reading’ cortical feedback– which would contribute invaluable data to theoretical frameworks.
The proposed research project will advance our understanding of ongoing brain activity, contextual processing, and cortical feedback - contributing to what is known about general cortical functions. By providing new insights as to the information content of cortical feedback, the proposal will fill one of the most important gaps in today’s knowledge about brain function. Friston’s unifying theory of the brain (Friston, 2010) and contemporary models of the predictive-coding framework (Hawkins and Blakeslee, 2004;Mumford, 1992;Rao and Ballard, 1999) assign feedback processing an essential role in cortical processing. Compared to feedforward information processing, our knowledge about feedback processing is in its infancy. The proposal introduces parametric and explorative brain reading designs to investigate this feedback processing. The chief goal of my proposal will be precision measures of cortical feedback, and a more ambitious objective is to read mental images and inner thoughts.
Max ERC Funding
1 494 714 €
Duration
Start date: 2012-12-01, End date: 2017-11-30
Project acronym BRAINVISIONREHAB
Project ‘Seeing’ with the ears, hands and bionic eyes: from theories about brain organization to visual rehabilitation
Researcher (PI) Amir Amedi
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS5, ERC-2012-StG_20111109
Summary My lab's work ranges from basic science, querying brain plasticity and sensory integration, to technological developments, allowing the blind to be more independent and even “see” using sounds and touch similar to bats and dolphins (a.k.a. Sensory Substitution Devices, SSDs), and back to applying these devices in research. We propose that, with proper training, any brain area or network can change the type of sensory input it uses to retrieve behaviorally task-relevant information within a matter of days. If this is true, it can have far reaching implications also for clinical rehabilitation. To achieve this, we are developing several innovative SSDs which encode the most crucial aspects of vision and increase their accessibility the blind, along with targeted, structured training protocols both in virtual environments and in real life. For instance, the “EyeMusic”, encodes colored complex images using pleasant musical scales and instruments, and the “EyeCane”, a palm-size cane, which encodes distance and depth in several directions accurately and efficiently. We provide preliminary but compelling evidence that following such training, SSDs can enable almost blind to recognize daily objects, colors, faces and facial expressions, read street signs, and aiding mobility and navigation. SSDs can also be used in conjunction with (any) invasive approach for visual rehabilitation. We are developing a novel hybrid Visual Rehabilitation Device which combines SSD and bionic eyes. In this set up, the SSDs is used in training the brain to “see” prior to surgery, in providing explanatory signal after surgery and in augmenting the capabilities of the bionic-eyes using information arriving from the same image. We will chart the dynamics of the plastic changes in the brain by performing unprecedented longitudinal Neuroimaging, Electrophysiological and Neurodisruptive approaches while individuals learn to ‘see’ using each of the visual rehabilitation approaches suggested here.
Summary
My lab's work ranges from basic science, querying brain plasticity and sensory integration, to technological developments, allowing the blind to be more independent and even “see” using sounds and touch similar to bats and dolphins (a.k.a. Sensory Substitution Devices, SSDs), and back to applying these devices in research. We propose that, with proper training, any brain area or network can change the type of sensory input it uses to retrieve behaviorally task-relevant information within a matter of days. If this is true, it can have far reaching implications also for clinical rehabilitation. To achieve this, we are developing several innovative SSDs which encode the most crucial aspects of vision and increase their accessibility the blind, along with targeted, structured training protocols both in virtual environments and in real life. For instance, the “EyeMusic”, encodes colored complex images using pleasant musical scales and instruments, and the “EyeCane”, a palm-size cane, which encodes distance and depth in several directions accurately and efficiently. We provide preliminary but compelling evidence that following such training, SSDs can enable almost blind to recognize daily objects, colors, faces and facial expressions, read street signs, and aiding mobility and navigation. SSDs can also be used in conjunction with (any) invasive approach for visual rehabilitation. We are developing a novel hybrid Visual Rehabilitation Device which combines SSD and bionic eyes. In this set up, the SSDs is used in training the brain to “see” prior to surgery, in providing explanatory signal after surgery and in augmenting the capabilities of the bionic-eyes using information arriving from the same image. We will chart the dynamics of the plastic changes in the brain by performing unprecedented longitudinal Neuroimaging, Electrophysiological and Neurodisruptive approaches while individuals learn to ‘see’ using each of the visual rehabilitation approaches suggested here.
Max ERC Funding
1 499 900 €
Duration
Start date: 2013-09-01, End date: 2018-08-31
Project acronym C9ND
Project C9orf72-mediated neurodegeneration: mechanisms and therapeutics
Researcher (PI) Adrian Michael Isaacs
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Consolidator Grant (CoG), LS5, ERC-2014-CoG
Summary An expanded GGGGCC repeat in a non-coding region of the C9orf72 gene is the most common known cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). The repeat RNA is transcribed and accumulates in neuronal RNA aggregates, implicating RNA toxicity as a key pathogenic mechanism. However, the pathways that lead to neurodegeneration are unknown. My lab has made pioneering contributions to the understanding of C9orf72 FTD/ALS, and reported the first structure of the repeat RNA, and the first description of both sense and antisense RNA aggregates in patient brain. We have now developed new disease models that allow, for the first time, the dissection of RNA toxicity both in vivo and in sophisticated neuronal culture models. We have also used our knowledge of the repeat structure to identify novel small molecules that show very strong binding to the repeats. We will utilise our innovative disease models in a multidisciplinary approach to fully dissect the cellular pathways underlying C9orf72 repeat RNA toxicity in vivo, on a genome-wide scale. Altered RNA metabolism has been implicated in a wide range of neurodegenerative diseases, indicating that our findings will provide profound new insight into fundamental mechanisms of neuronal maintenance and survival. This research programme will also deliver a step change in our understanding of C9orf72 FTD/ALS pathogenesis and provide essential insight for the identification of small molecules with genuine therapeutic potential. RNA-mediated mechanisms are now known to be a common theme in neurodegeneration, suggesting these findings will have broad significance.
Summary
An expanded GGGGCC repeat in a non-coding region of the C9orf72 gene is the most common known cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). The repeat RNA is transcribed and accumulates in neuronal RNA aggregates, implicating RNA toxicity as a key pathogenic mechanism. However, the pathways that lead to neurodegeneration are unknown. My lab has made pioneering contributions to the understanding of C9orf72 FTD/ALS, and reported the first structure of the repeat RNA, and the first description of both sense and antisense RNA aggregates in patient brain. We have now developed new disease models that allow, for the first time, the dissection of RNA toxicity both in vivo and in sophisticated neuronal culture models. We have also used our knowledge of the repeat structure to identify novel small molecules that show very strong binding to the repeats. We will utilise our innovative disease models in a multidisciplinary approach to fully dissect the cellular pathways underlying C9orf72 repeat RNA toxicity in vivo, on a genome-wide scale. Altered RNA metabolism has been implicated in a wide range of neurodegenerative diseases, indicating that our findings will provide profound new insight into fundamental mechanisms of neuronal maintenance and survival. This research programme will also deliver a step change in our understanding of C9orf72 FTD/ALS pathogenesis and provide essential insight for the identification of small molecules with genuine therapeutic potential. RNA-mediated mechanisms are now known to be a common theme in neurodegeneration, suggesting these findings will have broad significance.
Max ERC Funding
1 985 699 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym CAM-RIG
Project ConfocAl Microscopy and real-time Rheology of dynamIc hyroGels
Researcher (PI) Oren Alexander SCHERMAN
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Consolidator Grant (CoG), PE5, ERC-2016-COG
Summary Hydrogels cross-linked through supramolecular interactions are highly dependant on the dynamic charac- teristics of the physical cross-links. Few fundamental studies have been undertaken to quantitatively de- scribe structure-property relationships for these types of systems. Hydrogels formed from CB[8]-mediated supramolecular physical cross-linking mechanisms have gained significant interest on account of their excel- lent physical and mechanical properties such as self-healing and shear-thinning. This supramolecular motif has been further exploited to introduce and compatibilise a wide variety of different materials into hydrogel networks without phase separation, forming hybrid composite hydrogels attributed with unique and emergent properties. This proposal aims to pioneer the combination of several state-of-the-art characterisation tech- niques into an unique experimental setup (CAM-RIG), which will combine super-resolution and confocal microscopy imaging modalities with simultaneous strain-controlled rheological measurements to investigate fundamental structure-property relationships of these systems. For the first time it will be possible to decon- volute the molecular-level dynamics of the supramolecular physical cross-links from chain entanglement of the polymeric networks and understand their relative contributions on the resultant properties of the hydrogels. Using the fundamental insight gained, a set of key parameters will be determined to maximise the potential of supramolecular biocompatible hydrogels, driving paradigm shifts in sustainable science and biomaterial applications through the precise tuning of physical properties.
Summary
Hydrogels cross-linked through supramolecular interactions are highly dependant on the dynamic charac- teristics of the physical cross-links. Few fundamental studies have been undertaken to quantitatively de- scribe structure-property relationships for these types of systems. Hydrogels formed from CB[8]-mediated supramolecular physical cross-linking mechanisms have gained significant interest on account of their excel- lent physical and mechanical properties such as self-healing and shear-thinning. This supramolecular motif has been further exploited to introduce and compatibilise a wide variety of different materials into hydrogel networks without phase separation, forming hybrid composite hydrogels attributed with unique and emergent properties. This proposal aims to pioneer the combination of several state-of-the-art characterisation tech- niques into an unique experimental setup (CAM-RIG), which will combine super-resolution and confocal microscopy imaging modalities with simultaneous strain-controlled rheological measurements to investigate fundamental structure-property relationships of these systems. For the first time it will be possible to decon- volute the molecular-level dynamics of the supramolecular physical cross-links from chain entanglement of the polymeric networks and understand their relative contributions on the resultant properties of the hydrogels. Using the fundamental insight gained, a set of key parameters will be determined to maximise the potential of supramolecular biocompatible hydrogels, driving paradigm shifts in sustainable science and biomaterial applications through the precise tuning of physical properties.
Max ERC Funding
2 038 120 €
Duration
Start date: 2017-05-01, End date: 2022-04-30
Project acronym CatHet
Project New Catalytic Asymmetric Strategies for N-Heterocycle Synthesis
Researcher (PI) John Forwood Bower
Host Institution (HI) UNIVERSITY OF BRISTOL
Call Details Starting Grant (StG), PE5, ERC-2014-STG
Summary Medicinal chemistry requires more efficient and diverse methods for the asymmetric synthesis of chiral scaffolds. Over 60% of the world’s top selling small molecule drug compounds are chiral and, of these, approximately 80% are marketed as single enantiomers. There is a compelling correlation between drug candidate “chiral complexity” and the likelihood of progression to the marketplace. Surprisingly, and despite the tremendous advances made in catalysis over the past several decades, the “chiral complexity” of drug discovery libraries has actually decreased, while, at the same time, for the reasons mentioned above, the “chiral complexity” of marketed drugs has increased. Since the mid-1990s, there has been a notable acceleration of this “complexity divergence”. Consequently, there is now an urgent need to provide efficient processes that directly access privileged chiral scaffolds. It is our philosophy that catalysis holds the key here and new processes should be based upon platforms that can exert control over both absolute and relative stereochemistry. In this proposal we outline the development of a range of N-heteroannulation processes based upon the catalytic generation and trapping of unique or unusual classes of organometallic intermediate derived from transition metal insertion into C-C and C-N sigma-bonds. We will provide a variety of enabling methodologies and demonstrate applicability in flexible total syntheses of important natural product scaffolds. The processes proposed are synthetically flexible, operationally simple and amenable to asymmetric catalysis. Likely starting points, based upon preliminary results, will set the stage for the realisation of aspirational and transformative goals. Through the study of the organometallic intermediates involved here, there is potential to generalise these new catalytic manifolds, such that this research will transcend N heterocyclic chemistry to provide enabling methods for organic chemistry as a whole.
Summary
Medicinal chemistry requires more efficient and diverse methods for the asymmetric synthesis of chiral scaffolds. Over 60% of the world’s top selling small molecule drug compounds are chiral and, of these, approximately 80% are marketed as single enantiomers. There is a compelling correlation between drug candidate “chiral complexity” and the likelihood of progression to the marketplace. Surprisingly, and despite the tremendous advances made in catalysis over the past several decades, the “chiral complexity” of drug discovery libraries has actually decreased, while, at the same time, for the reasons mentioned above, the “chiral complexity” of marketed drugs has increased. Since the mid-1990s, there has been a notable acceleration of this “complexity divergence”. Consequently, there is now an urgent need to provide efficient processes that directly access privileged chiral scaffolds. It is our philosophy that catalysis holds the key here and new processes should be based upon platforms that can exert control over both absolute and relative stereochemistry. In this proposal we outline the development of a range of N-heteroannulation processes based upon the catalytic generation and trapping of unique or unusual classes of organometallic intermediate derived from transition metal insertion into C-C and C-N sigma-bonds. We will provide a variety of enabling methodologies and demonstrate applicability in flexible total syntheses of important natural product scaffolds. The processes proposed are synthetically flexible, operationally simple and amenable to asymmetric catalysis. Likely starting points, based upon preliminary results, will set the stage for the realisation of aspirational and transformative goals. Through the study of the organometallic intermediates involved here, there is potential to generalise these new catalytic manifolds, such that this research will transcend N heterocyclic chemistry to provide enabling methods for organic chemistry as a whole.
Max ERC Funding
1 548 738 €
Duration
Start date: 2015-04-01, End date: 2020-03-31
Project acronym CEIDNFSTTAIS
Project Controlling excitability in developing neurons: from synapses to the axon initial segment
Researcher (PI) Juan Burrone
Host Institution (HI) KING'S COLLEGE LONDON
Call Details Starting Grant (StG), LS5, ERC-2011-StG_20101109
Summary A critical question in neuroscience is to understand how neurons wire up to form a functional network. During the wiring of the brain it is important to establish mechanisms that act as safeguards to control and stabilize neuronal excitability in the face of large, chronic changes in neuronal or network activity. This is especially true for developing systems that undergo rapid and large scale forms of plasticity, which could easily lead to large imbalances in activity. If left unchecked, they could lead the network to its extremes: a complete loss of signal or epileptic-like activity. For this reason neurons employ different strategies to maintain their excitability within reasonable bounds. This proposal will focus on two crucial sites for neuronal information processing and integration: the synapse and the axon initial segment (AIS). Both sites undergo important structural and functional rearrangements in response to chronic activity changes, thus controlling the input-output function of a neuron and allowing the network to function efficiently. This proposal will explore novel forms of plasticity that occur during development and which are key to establishing a functional network. They range from understanding the role of activity during synapse formation to how pre- and postsynaptic structure and function become matched during development. Finally, it tackles a novel form of plasticity that lies downstream of synaptic inputs and is responsible for setting the threshold of action potential firing: the axon initial segment. Here, chronic changes in network activity results in a physical relocation of the AIS along the axon, which in turn alters the excitability of the neuron. This proposal will focus on the central issue of how a neuron alters both its input (synapses) and output (AIS) during development to maintain its activity levels within a set range and allow a functional network to form.
Summary
A critical question in neuroscience is to understand how neurons wire up to form a functional network. During the wiring of the brain it is important to establish mechanisms that act as safeguards to control and stabilize neuronal excitability in the face of large, chronic changes in neuronal or network activity. This is especially true for developing systems that undergo rapid and large scale forms of plasticity, which could easily lead to large imbalances in activity. If left unchecked, they could lead the network to its extremes: a complete loss of signal or epileptic-like activity. For this reason neurons employ different strategies to maintain their excitability within reasonable bounds. This proposal will focus on two crucial sites for neuronal information processing and integration: the synapse and the axon initial segment (AIS). Both sites undergo important structural and functional rearrangements in response to chronic activity changes, thus controlling the input-output function of a neuron and allowing the network to function efficiently. This proposal will explore novel forms of plasticity that occur during development and which are key to establishing a functional network. They range from understanding the role of activity during synapse formation to how pre- and postsynaptic structure and function become matched during development. Finally, it tackles a novel form of plasticity that lies downstream of synaptic inputs and is responsible for setting the threshold of action potential firing: the axon initial segment. Here, chronic changes in network activity results in a physical relocation of the AIS along the axon, which in turn alters the excitability of the neuron. This proposal will focus on the central issue of how a neuron alters both its input (synapses) and output (AIS) during development to maintain its activity levels within a set range and allow a functional network to form.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-03-01, End date: 2017-02-28
Project acronym CerebralHominoids
Project Evolutionary biology of human and great ape brain development in cerebral organoids
Researcher (PI) Madeline LANCASTER
Host Institution (HI) UNITED KINGDOM RESEARCH AND INNOVATION
Call Details Starting Grant (StG), LS5, ERC-2017-STG
Summary Humans are endowed with a number of advanced cognitive abilities not seen in other species. So what allows the human brain to stand out from the rest in these capabilities? In general, the brains of primates, including humans, have more neurons per unit volume than other mammals. But humans are also in the fortunate position of having the largest of the primate brains, making the number of neurons in the human cerebral cortex greatly expanded. Thus, the difference seems to be a matter of quantity, not quality. My laboratory is interested in understanding how neuron number, and thus brain size, is determined in human brain development.
The research proposed here is aimed at taking an evolutionary approach to this question and comparing brain development in an in vitro 3D model system, cerebral organoids. This method, which relies on self-organization from differentiating pluripotent stem cells, recapitulates remarkably well the endogenous developmental program of the human brain. Having previously established the brain organoid approach, and more recently improved upon it with the application of bioengineering, my laboratory is in a unique position to carry out functional studies of human brain development. I propose to use this approach to compare developing human brain tissue to that of other hominid species and tease apart unique features of human neural stem cells and progenitors that allow them to generate more neurons and therefore a greater cerebral cortical size. Furthermore, we will perform transcriptomic and functional screening to identify factors underlying this expansion, followed by careful genetic substitution to test the contributions of putative evolutionary changes. In this way, we will functionally test putative human evolutionary changes in a manner not previously possible.
Summary
Humans are endowed with a number of advanced cognitive abilities not seen in other species. So what allows the human brain to stand out from the rest in these capabilities? In general, the brains of primates, including humans, have more neurons per unit volume than other mammals. But humans are also in the fortunate position of having the largest of the primate brains, making the number of neurons in the human cerebral cortex greatly expanded. Thus, the difference seems to be a matter of quantity, not quality. My laboratory is interested in understanding how neuron number, and thus brain size, is determined in human brain development.
The research proposed here is aimed at taking an evolutionary approach to this question and comparing brain development in an in vitro 3D model system, cerebral organoids. This method, which relies on self-organization from differentiating pluripotent stem cells, recapitulates remarkably well the endogenous developmental program of the human brain. Having previously established the brain organoid approach, and more recently improved upon it with the application of bioengineering, my laboratory is in a unique position to carry out functional studies of human brain development. I propose to use this approach to compare developing human brain tissue to that of other hominid species and tease apart unique features of human neural stem cells and progenitors that allow them to generate more neurons and therefore a greater cerebral cortical size. Furthermore, we will perform transcriptomic and functional screening to identify factors underlying this expansion, followed by careful genetic substitution to test the contributions of putative evolutionary changes. In this way, we will functionally test putative human evolutionary changes in a manner not previously possible.
Max ERC Funding
1 444 911 €
Duration
Start date: 2018-07-01, End date: 2023-06-30
Project acronym CheSSTaG
Project Chemotactic Super-Selective Targeting of Gliomas
Researcher (PI) Giuseppe BATTAGLIA
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Consolidator Grant (CoG), PE5, ERC-2017-COG
Summary I propose here a research program aimed to the design a completely new platform for drug delivery. I will combine our existing repertoire of molecular engineering tools based around our established approach to design responsive nanoparticles known as Polymersomes to integrate new features using clinically safe and biodegradable components that will make them super-selective and chemotactic toward glucose gradients so to deliver large therapeutic payload into the central nervous systems and the brain in particular targeting cancer cells harbouring within the healthy. We will do so by engineering components using supramolecular interaction inspired by biological complexity equipping carriers with the ability to self-propelled as a function of glucose gradient. I will complement our proposed design with advanced biological characterisation associating functional information arising form the physiological barrier to structural parameters integrated into the final carrier design.
Summary
I propose here a research program aimed to the design a completely new platform for drug delivery. I will combine our existing repertoire of molecular engineering tools based around our established approach to design responsive nanoparticles known as Polymersomes to integrate new features using clinically safe and biodegradable components that will make them super-selective and chemotactic toward glucose gradients so to deliver large therapeutic payload into the central nervous systems and the brain in particular targeting cancer cells harbouring within the healthy. We will do so by engineering components using supramolecular interaction inspired by biological complexity equipping carriers with the ability to self-propelled as a function of glucose gradient. I will complement our proposed design with advanced biological characterisation associating functional information arising form the physiological barrier to structural parameters integrated into the final carrier design.
Max ERC Funding
2 081 747 €
Duration
Start date: 2018-05-01, End date: 2023-04-30
Project acronym CHIME
Project The Role of Cortico-Hippocampal Interactions during Memory Encoding
Researcher (PI) Daniel (Ari) Bendor
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Starting Grant (StG), LS5, ERC-2014-STG
Summary This research proposal’s goal is to investigate the role of cortico-hippocampal interactions during the encoding and consolidation of a memory. Current memory consolidation models postulate that memory storage in our brains occurs by a dynamic process- a recent episodic experience is initially encoded in the hippocampus, and during off-line states such as sleep, the encoded memory is gradually transferred to neocortex for long-term storage. One potential neural mechanism by which this could occur is replay, a phenomenon where neural activity patterns in the hippocampus evoked by a previous experience reactivate spontaneously during non-REM sleep, leading to coordinated cortical reactivation. While previous work suggests that hippocampal replay is important for encoding new memories, how memory consolidation is accomplished through cortico-hippocampal interactions is not well understood.
This research project has three major aims- 1) examine how cortical feedback influences which spatial trajectory is replayed by the hippocampus, 2) investigate how the hippocampal replay of a behavioural episode modifies cortical circuits, 3) measure the causal role of cortico-hippocampal interactions in consolidating memories. We will record ensemble activity from freely moving rats during an auditory-spatial association task and during post-behavioural sleep sessions. We will focus our ensemble recordings on two brain regions: 1) the dorsal CA1 region of the hippocampus, where the phenomenon of sleep replay has been most extensively examined, and 2) auditory cortex, a region of the brain critical for both auditory perception and long-term memory storage. This work will use behavioral and molecular-genetic techniques in combination with large-scale electrophysiological recordings, to help elucidate the role of cortico-hippocampal interactions in memory encoding and consolidation.
Summary
This research proposal’s goal is to investigate the role of cortico-hippocampal interactions during the encoding and consolidation of a memory. Current memory consolidation models postulate that memory storage in our brains occurs by a dynamic process- a recent episodic experience is initially encoded in the hippocampus, and during off-line states such as sleep, the encoded memory is gradually transferred to neocortex for long-term storage. One potential neural mechanism by which this could occur is replay, a phenomenon where neural activity patterns in the hippocampus evoked by a previous experience reactivate spontaneously during non-REM sleep, leading to coordinated cortical reactivation. While previous work suggests that hippocampal replay is important for encoding new memories, how memory consolidation is accomplished through cortico-hippocampal interactions is not well understood.
This research project has three major aims- 1) examine how cortical feedback influences which spatial trajectory is replayed by the hippocampus, 2) investigate how the hippocampal replay of a behavioural episode modifies cortical circuits, 3) measure the causal role of cortico-hippocampal interactions in consolidating memories. We will record ensemble activity from freely moving rats during an auditory-spatial association task and during post-behavioural sleep sessions. We will focus our ensemble recordings on two brain regions: 1) the dorsal CA1 region of the hippocampus, where the phenomenon of sleep replay has been most extensively examined, and 2) auditory cortex, a region of the brain critical for both auditory perception and long-term memory storage. This work will use behavioral and molecular-genetic techniques in combination with large-scale electrophysiological recordings, to help elucidate the role of cortico-hippocampal interactions in memory encoding and consolidation.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-04-01, End date: 2021-03-31
Project acronym CHOLINOMIRS
Project CholinomiRs: MicroRNA Regulators of Cholinergic Signalling in the Neuro-Immune Interface
Researcher (PI) Hermona Soreq
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS5, ERC-2012-ADG_20120314
Summary "Communication between the nervous and the immune system is pivotal for maintaining homeostasis and ensuring rapid and efficient reaction to stress and infection insults. The emergence of microRNAs (miRs) as regulators of gene expression and of acetylcholine (ACh) signalling as regulator of anxiety and inflammation provides a model for studying this interaction. My hypothesis is that 1) a specific sub-group of miRs, designated ""CholinomiRs"", may silence multiple target genes in the neuro-immune interface; 2) these miRs compete with each other on the interaction with their targets, and 3) mutations interfering with miR binding lead to inherited susceptibility to anxiety and inflammation disorders by modifying these interactions. Our preliminary findings have shown that by targeting acetylcholinesterase (AChE), CholinomiR-132 can intensify acute stress, resolve intestinal inflammation and change post-ischemic stroke responses. Further, we have identified clustered single nucleotide polymorphisms (SNPs) interfering with AChE silencing by several miRs which associate with elevated trait anxiety, blood pressure and inflammation. To further study miR regulators of ACh signalling, I plan to: (1) Identify anxiety and inflammation-induced changes in CholinomiRs and their targets in challenged brain and immune cells. (2) Establish the roles of these targets for one selected CholinomiR by tissue-specific manipulations. (3) Study primate-specific CholinomiRs by continued human DNA screens to identify SNPs and in ""humanized"" mice with knocked-in human AChE and transgenic CholinomiR-608. (4) Test if therapeutic modulation of aberrant CholinomiR expression can restore homeostasis. This research will clarify how miRs interact with each other in health and disease, introduce the dimension of complexity of multi-target competition and miR interactions and make a conceptual change in miRs research while enhancing the ability to intervene with diseases involving impaired ACh signalling."
Summary
"Communication between the nervous and the immune system is pivotal for maintaining homeostasis and ensuring rapid and efficient reaction to stress and infection insults. The emergence of microRNAs (miRs) as regulators of gene expression and of acetylcholine (ACh) signalling as regulator of anxiety and inflammation provides a model for studying this interaction. My hypothesis is that 1) a specific sub-group of miRs, designated ""CholinomiRs"", may silence multiple target genes in the neuro-immune interface; 2) these miRs compete with each other on the interaction with their targets, and 3) mutations interfering with miR binding lead to inherited susceptibility to anxiety and inflammation disorders by modifying these interactions. Our preliminary findings have shown that by targeting acetylcholinesterase (AChE), CholinomiR-132 can intensify acute stress, resolve intestinal inflammation and change post-ischemic stroke responses. Further, we have identified clustered single nucleotide polymorphisms (SNPs) interfering with AChE silencing by several miRs which associate with elevated trait anxiety, blood pressure and inflammation. To further study miR regulators of ACh signalling, I plan to: (1) Identify anxiety and inflammation-induced changes in CholinomiRs and their targets in challenged brain and immune cells. (2) Establish the roles of these targets for one selected CholinomiR by tissue-specific manipulations. (3) Study primate-specific CholinomiRs by continued human DNA screens to identify SNPs and in ""humanized"" mice with knocked-in human AChE and transgenic CholinomiR-608. (4) Test if therapeutic modulation of aberrant CholinomiR expression can restore homeostasis. This research will clarify how miRs interact with each other in health and disease, introduce the dimension of complexity of multi-target competition and miR interactions and make a conceptual change in miRs research while enhancing the ability to intervene with diseases involving impaired ACh signalling."
Max ERC Funding
2 375 600 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym ChromatidCohesion
Project Establishment of Sister Chromatid Cohesion
Researcher (PI) Frank Uhlmann
Host Institution (HI) THE FRANCIS CRICK INSTITUTE LIMITED
Call Details Advanced Grant (AdG), LS1, ERC-2014-ADG
Summary Following their synthesis during DNA replication, sister chromatids remain paired by the cohesin complex, which forms the basis for their faithful segregation during cell division. Cohesin is a large ring-shaped protein complex, incorporating an ABC-type ATPase module. Despite its importance for genome stability, the molecular mechanism of cohesin action remains as intriguing as it remains poorly understood. How is cohesin topologically loaded onto chromatin? How is it unloaded again? What happens to cohesin during DNA replication in S-phase, so that it establishes cohesion between newly synthesized sister chromatids? We propose to capitalise on our recent success in the biochemical reconstitution of topological cohesin loading onto DNA. This lays the foundation for a work programme encompassing a combination of biochemical, single molecule, structural and genetic approaches to address the above questions. Five work packages will investigate cohesin’s molecular behaviour during its life-cycle on chromosomes, including the ATP binding and hydrolysis-dependent conformational changes that make this molecular machine work. It will be complemented by mechanistic analyses of the cofactors that help cohesin to load onto chromosomes and establish sister chromatid cohesion. The insight gained will not only advance our molecular knowledge of sister chromatid cohesion. It will more generally advance our understanding of the ubiquitous family of chromosomal SMC ATPases, of which cohesin is a member, and their activity of shaping and segregating genomes.
Summary
Following their synthesis during DNA replication, sister chromatids remain paired by the cohesin complex, which forms the basis for their faithful segregation during cell division. Cohesin is a large ring-shaped protein complex, incorporating an ABC-type ATPase module. Despite its importance for genome stability, the molecular mechanism of cohesin action remains as intriguing as it remains poorly understood. How is cohesin topologically loaded onto chromatin? How is it unloaded again? What happens to cohesin during DNA replication in S-phase, so that it establishes cohesion between newly synthesized sister chromatids? We propose to capitalise on our recent success in the biochemical reconstitution of topological cohesin loading onto DNA. This lays the foundation for a work programme encompassing a combination of biochemical, single molecule, structural and genetic approaches to address the above questions. Five work packages will investigate cohesin’s molecular behaviour during its life-cycle on chromosomes, including the ATP binding and hydrolysis-dependent conformational changes that make this molecular machine work. It will be complemented by mechanistic analyses of the cofactors that help cohesin to load onto chromosomes and establish sister chromatid cohesion. The insight gained will not only advance our molecular knowledge of sister chromatid cohesion. It will more generally advance our understanding of the ubiquitous family of chromosomal SMC ATPases, of which cohesin is a member, and their activity of shaping and segregating genomes.
Max ERC Funding
2 120 100 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym CHROMOREP
Project Reconstitution of Chromosome Replication and Epigenetic Inheritance
Researcher (PI) John Diffley
Host Institution (HI) THE FRANCIS CRICK INSTITUTE LIMITED
Call Details Advanced Grant (AdG), LS1, ERC-2014-ADG
Summary A PubMed search for ‘epigenetic’ identifies nearly 35,000 entries, yet the molecular mechanisms by which chromatin modification and gene expression patterns are actually inherited during chromosome replication — mechanisms which lie at the heart of epigenetic inheritance of gene expression — are still largely uncharacterised. Understanding these mechanisms would be greatly aided if we could reconstitute the replication of chromosomes with purified proteins. The past few years have seen great progress in understanding eukaryotic DNA replication through the use of cell-free replication systems and reconstitution of individual steps in replication with purified proteins and naked DNA. We will use these in vitro replication systems together with both established and novel chromatin assembly systems to understand: a) how chromatin influences replication origin choice and timing, b) how nucleosomes on parental chromosomes are disrupted during replication and are distributed to daughter chromatids, and c) how chromatin states and gene expression patterns are re-established after passage of the replication fork. We will begin with simple, defined templates to learn basic principles, and we will use this knowledge to reconstitute genome-wide replication patterns. The experimental plan will exploit our well-characterised yeast systems, and where feasible explore these questions with human proteins. Our work will help explain how epigenetic inheritance works at a molecular level, and will complement work in vivo by many others. It will also underpin our long-term research goals aimed at making functional chromosomes from purified, defined components to understand how DNA replication interacts with gene expression, DNA repair and chromosome segregation.
Summary
A PubMed search for ‘epigenetic’ identifies nearly 35,000 entries, yet the molecular mechanisms by which chromatin modification and gene expression patterns are actually inherited during chromosome replication — mechanisms which lie at the heart of epigenetic inheritance of gene expression — are still largely uncharacterised. Understanding these mechanisms would be greatly aided if we could reconstitute the replication of chromosomes with purified proteins. The past few years have seen great progress in understanding eukaryotic DNA replication through the use of cell-free replication systems and reconstitution of individual steps in replication with purified proteins and naked DNA. We will use these in vitro replication systems together with both established and novel chromatin assembly systems to understand: a) how chromatin influences replication origin choice and timing, b) how nucleosomes on parental chromosomes are disrupted during replication and are distributed to daughter chromatids, and c) how chromatin states and gene expression patterns are re-established after passage of the replication fork. We will begin with simple, defined templates to learn basic principles, and we will use this knowledge to reconstitute genome-wide replication patterns. The experimental plan will exploit our well-characterised yeast systems, and where feasible explore these questions with human proteins. Our work will help explain how epigenetic inheritance works at a molecular level, and will complement work in vivo by many others. It will also underpin our long-term research goals aimed at making functional chromosomes from purified, defined components to understand how DNA replication interacts with gene expression, DNA repair and chromosome segregation.
Max ERC Funding
1 983 019 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym CHROMOSOME STABILITY
Project Coordination of DNA replication and DNA repair at single-forks: the role of the Smc5-Smc6 complex in replication fork stalling and resumption
Researcher (PI) Luis Fernando Aragon Alcaide
Host Institution (HI) IMPERIAL COLLEGE OF SCIENCE TECHNOLOGY AND MEDICINE
Call Details Starting Grant (StG), LS1, ERC-2007-StG
Summary DNA replication represents a dangerous moment in the life of the cell as endogenous and exogenous events challenge genome integrity by interfering with the progression, stability and restart of the replication fork. Failure to protect stalled forks or to process the replication fork appropriately contribute to the pathological mechanisms giving rise to cancer, therefore an understanding of the intricate mechanisms that ensure fork integrity can provide targets for new chemotherapeutic assays. Smc5-Smc6 is a multi-subunit complex with a poorly understood function in DNA replication and repair. One of its subunits, Nse2, is able to promote the addition of a small ubiquitin-like protein modifier (SUMO) to specific target proteins. Recent work has revealed that the Smc5-Smc6 complex is required for the progression of replication forks through damaged DNA and is recruited de novo to forks that undergo collapse. In addition, Smc5-Smc6 mediate repair of DNA breaks by homologous recombination between sister-chromatids. Thus, Smc5-Smc6 is anticipated to promote recombinational repair at stalled/collapsed replication forks. My laboratory proposes to develop molecular techniques to study replication at the level of single replication forks. We will employ these assays to identify and dissect the function of factors involved in replication fork stability and repair. We will place an emphasis on the study of the Smc5-Smc6 complex in these processes because of its potential roles in recombination-dependent fork repair and restart. We also propose to identify novel Nse2 substrates involved in DNA repair using yeast model systems. Specifically, we will address the following points: (1) Development of assays for analysis of factors involved in stabilisation, collapse and re-start of single-forks, (2) Analysis of the roles of Smc5-Smc6 in fork biology using developed techniques, (3) Isolation and functional analysis of novel Nse2 substrates.
Summary
DNA replication represents a dangerous moment in the life of the cell as endogenous and exogenous events challenge genome integrity by interfering with the progression, stability and restart of the replication fork. Failure to protect stalled forks or to process the replication fork appropriately contribute to the pathological mechanisms giving rise to cancer, therefore an understanding of the intricate mechanisms that ensure fork integrity can provide targets for new chemotherapeutic assays. Smc5-Smc6 is a multi-subunit complex with a poorly understood function in DNA replication and repair. One of its subunits, Nse2, is able to promote the addition of a small ubiquitin-like protein modifier (SUMO) to specific target proteins. Recent work has revealed that the Smc5-Smc6 complex is required for the progression of replication forks through damaged DNA and is recruited de novo to forks that undergo collapse. In addition, Smc5-Smc6 mediate repair of DNA breaks by homologous recombination between sister-chromatids. Thus, Smc5-Smc6 is anticipated to promote recombinational repair at stalled/collapsed replication forks. My laboratory proposes to develop molecular techniques to study replication at the level of single replication forks. We will employ these assays to identify and dissect the function of factors involved in replication fork stability and repair. We will place an emphasis on the study of the Smc5-Smc6 complex in these processes because of its potential roles in recombination-dependent fork repair and restart. We also propose to identify novel Nse2 substrates involved in DNA repair using yeast model systems. Specifically, we will address the following points: (1) Development of assays for analysis of factors involved in stabilisation, collapse and re-start of single-forks, (2) Analysis of the roles of Smc5-Smc6 in fork biology using developed techniques, (3) Isolation and functional analysis of novel Nse2 substrates.
Max ERC Funding
893 396 €
Duration
Start date: 2008-09-01, End date: 2013-08-31
Project acronym CilDyn
Project Molecular analysis of the Hedgehog signal transduction complex in the primary cilium
Researcher (PI) Christian Siebold
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Consolidator Grant (CoG), LS1, ERC-2014-CoG
Summary The unexpected connection between the primary cilium and cell-to-cell signalling is one of the most exciting discoveries in cell and developmental biology in the last decade. In particular, the Hedgehog (Hh) pathway relies on the primary cilium to fulfil its fundamental functions in orchestrating vertebrate development. This microtubule-based antenna, up to 5 µm long, protrudes from the plasma membrane of almost every human cell and is the essential compartment for the entire Hh signalling cascade. All its molecular components, from the most upstream transmembrane Hh receptor down to the ultimate transcription factors, are dynamically localised and enriched in the primary cilium. The aim of this proposal, which combines structural biology and live cell imaging, is to understand the function and signalling consequences of the multivalent interactions between Hh signal transducer proteins as well as their spatial and temporal regulation in the primary cilium. The key questions my laboratory will address are: What are the rules for assembly of Hh signal transduction complexes? How dynamic are these complexes in size and organisation? How are these processes linked to the transport and accumulation in the primary cilium?
I will combine state-of-the art structural biology techniques (with an emphasis on X-ray crystallography) to study the molecular architecture of binary and higher-order Hh signal transduction complexes and live cell fluorescence microscopy (for protein localisation and direct protein interactions). These two approaches will allow me to identify and define specific protein-protein interfaces at the atomic level and test their functional consequences in the cell in real time. My goal is to consolidate a world-class morphogen signal transduction laboratory, deciphering fundamental biological insights. Importantly, my results and reagents can potentially feed into the development of novel anti-cancer therapeutics and reagents promoting stem cell therapy.
Summary
The unexpected connection between the primary cilium and cell-to-cell signalling is one of the most exciting discoveries in cell and developmental biology in the last decade. In particular, the Hedgehog (Hh) pathway relies on the primary cilium to fulfil its fundamental functions in orchestrating vertebrate development. This microtubule-based antenna, up to 5 µm long, protrudes from the plasma membrane of almost every human cell and is the essential compartment for the entire Hh signalling cascade. All its molecular components, from the most upstream transmembrane Hh receptor down to the ultimate transcription factors, are dynamically localised and enriched in the primary cilium. The aim of this proposal, which combines structural biology and live cell imaging, is to understand the function and signalling consequences of the multivalent interactions between Hh signal transducer proteins as well as their spatial and temporal regulation in the primary cilium. The key questions my laboratory will address are: What are the rules for assembly of Hh signal transduction complexes? How dynamic are these complexes in size and organisation? How are these processes linked to the transport and accumulation in the primary cilium?
I will combine state-of-the art structural biology techniques (with an emphasis on X-ray crystallography) to study the molecular architecture of binary and higher-order Hh signal transduction complexes and live cell fluorescence microscopy (for protein localisation and direct protein interactions). These two approaches will allow me to identify and define specific protein-protein interfaces at the atomic level and test their functional consequences in the cell in real time. My goal is to consolidate a world-class morphogen signal transduction laboratory, deciphering fundamental biological insights. Importantly, my results and reagents can potentially feed into the development of novel anti-cancer therapeutics and reagents promoting stem cell therapy.
Max ERC Funding
1 727 456 €
Duration
Start date: 2015-08-01, End date: 2020-07-31
Project acronym CIRCOMMUNICATION
Project Deciphering molecular pathways of circadian clock communication
Researcher (PI) gad ASHER
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS1, ERC-2017-COG
Summary The overarching objective of this interdisciplinary project is to elucidate mechanisms through which billions of individual clocks in the body communicate with each other and tick in harmony. The mammalian circadian timing system consists of a master clock in the brain and subsidiary oscillators in almost every cell of the body. Since these clocks anticipate environmental changes and function together to orchestrate daily physiology and behavior their temporal synchronization is critical.
Our recent finding that oxygen serves as a resetting cue for circadian clocks points towards the unprecedented involvement of blood gases as time signals. We will apply cutting edge continuous physiological measurements in freely moving animals, alongside biochemical/molecular biology approaches and advanced cell culture setup to determine the molecular role of oxygen, carbon dioxide and pH in circadian clock communication and function.
The intricate nature of the mammalian circadian system demands the presence of communication mechanisms between clocks throughout the body at multiple levels. While previous studies primarily addressed the role of the master clock in resetting peripheral clocks, our knowledge regarding the communication among clocks between and within peripheral organs is rudimentary. We will reconstruct the mammalian circadian system from the bottom up, sequentially restoring clocks in peripheral tissues of a non-rhythmic animal to (i) obtain a system-view of the peripheral circadian communication network; and (ii) study novel tissue-derived circadian communication mechanisms.
This integrative proposal addresses fundamental aspects of circadian biology. It is expected to unravel the circadian communication network and shed light on how billions of clocks in the body function in unison. Its impact extends beyond circadian rhythms and bears great potential for research on communication between cells/tissues in various fields of biology.
Summary
The overarching objective of this interdisciplinary project is to elucidate mechanisms through which billions of individual clocks in the body communicate with each other and tick in harmony. The mammalian circadian timing system consists of a master clock in the brain and subsidiary oscillators in almost every cell of the body. Since these clocks anticipate environmental changes and function together to orchestrate daily physiology and behavior their temporal synchronization is critical.
Our recent finding that oxygen serves as a resetting cue for circadian clocks points towards the unprecedented involvement of blood gases as time signals. We will apply cutting edge continuous physiological measurements in freely moving animals, alongside biochemical/molecular biology approaches and advanced cell culture setup to determine the molecular role of oxygen, carbon dioxide and pH in circadian clock communication and function.
The intricate nature of the mammalian circadian system demands the presence of communication mechanisms between clocks throughout the body at multiple levels. While previous studies primarily addressed the role of the master clock in resetting peripheral clocks, our knowledge regarding the communication among clocks between and within peripheral organs is rudimentary. We will reconstruct the mammalian circadian system from the bottom up, sequentially restoring clocks in peripheral tissues of a non-rhythmic animal to (i) obtain a system-view of the peripheral circadian communication network; and (ii) study novel tissue-derived circadian communication mechanisms.
This integrative proposal addresses fundamental aspects of circadian biology. It is expected to unravel the circadian communication network and shed light on how billions of clocks in the body function in unison. Its impact extends beyond circadian rhythms and bears great potential for research on communication between cells/tissues in various fields of biology.
Max ERC Funding
1 999 945 €
Duration
Start date: 2018-03-01, End date: 2023-02-28
Project acronym CLAUSTRUM
Project The Claustrum: A Circuit Hub for Attention
Researcher (PI) Amihai CITRI
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Consolidator Grant (CoG), LS5, ERC-2017-COG
Summary Our senses face a constant barrage of information. Hence, understanding how our brain enables us to attend to relevant stimuli, while ignoring distractions, is of increasing biomedical importance. Recently, I discovered that the claustrum, a multi-sensory hub and recipient of extensive neuromodulatory input, enables resilience to distraction.
In my ERC project, I will explore the mechanisms underlying claustral mediation of resilience to distraction and develop novel approaches for assessing and modulating attention in mice, with implications for humans. Transgenic mouse models that I identified as enabling selective access to claustral neurons overcome its limiting anatomy, making the claustrum accessible to functional investigation. Using this novel genetic access, I obtained preliminary results strongly suggesting that the claustrum functions to filter distractions by adjusting cortical sensory gain.
My specific aims are: 1) To delineate the mechanisms whereby the claustrum achieves sensory gain control, by applying in-vivo cell-attached, multi-unit and fiber photometry recordings from claustral and cortical neurons during attention-demanding tasks. 2) To discriminate between the functions of the claustrum in multi-sensory integration and implementation of attention strategies, by employing multi-sensory behavioral paradigms while modulating claustral function. 3) To develop validated complementary physiological and behavioral protocols for adjusting claustral mediation of attention via neuromodulation.
This study is unique in its focus and aims: it will provide a stringent neurophysiological framework for defining a key mechanism underlying cognitive concepts of attention, and establish a novel platform for studying the function of the claustrum and manipulating its activity. The project is designed to achieve breakthroughs of fundamental nature and potentially lead to diagnostic and therapeutic advances relevant to attention disorders.
Summary
Our senses face a constant barrage of information. Hence, understanding how our brain enables us to attend to relevant stimuli, while ignoring distractions, is of increasing biomedical importance. Recently, I discovered that the claustrum, a multi-sensory hub and recipient of extensive neuromodulatory input, enables resilience to distraction.
In my ERC project, I will explore the mechanisms underlying claustral mediation of resilience to distraction and develop novel approaches for assessing and modulating attention in mice, with implications for humans. Transgenic mouse models that I identified as enabling selective access to claustral neurons overcome its limiting anatomy, making the claustrum accessible to functional investigation. Using this novel genetic access, I obtained preliminary results strongly suggesting that the claustrum functions to filter distractions by adjusting cortical sensory gain.
My specific aims are: 1) To delineate the mechanisms whereby the claustrum achieves sensory gain control, by applying in-vivo cell-attached, multi-unit and fiber photometry recordings from claustral and cortical neurons during attention-demanding tasks. 2) To discriminate between the functions of the claustrum in multi-sensory integration and implementation of attention strategies, by employing multi-sensory behavioral paradigms while modulating claustral function. 3) To develop validated complementary physiological and behavioral protocols for adjusting claustral mediation of attention via neuromodulation.
This study is unique in its focus and aims: it will provide a stringent neurophysiological framework for defining a key mechanism underlying cognitive concepts of attention, and establish a novel platform for studying the function of the claustrum and manipulating its activity. The project is designed to achieve breakthroughs of fundamental nature and potentially lead to diagnostic and therapeutic advances relevant to attention disorders.
Max ERC Funding
1 995 000 €
Duration
Start date: 2018-03-01, End date: 2023-02-28
Project acronym CLIP
Project Mapping functional protein-RNA interactions to identify new targets for oligonucleotide-based therapy
Researcher (PI) Jernej Ule
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Starting Grant (StG), LS1, ERC-2007-StG
Summary An important question of modern neurobiology is how neurons regulate synaptic function in response to excitation. In particular, the roles of alternative pre-mRNA splicing and mRNA translation regulation in this response are poorly understood. We will study the RNA-binding proteins (RBPs) that control these post-transcriptional changes using a UV crosslinking-based purification method (CLIP) and ultra-high throughput sequencing. Computational analysis of the resulting data will define the sequence and structural features of RNA motifs recognized by each RBP. Splicing microarrays and translation reporter assays will then allow us to examine the regulatory functions of RBPs and RNA motifs. By integrating the biochemical and functional datasets, we will relate the position of RNA motifs to the activity of bound RBPs, and predict the interactions that act as central nodes in the regulatory network. The physiological role of these core RBP-RNA interactions will then be tested using antisense RNAs. Together, these projects will provide insights to the regulatory mechanisms underlying neuronal activity-dependent changes, and provide new opportunities for future treatments of neurodegenerative disorders.
Summary
An important question of modern neurobiology is how neurons regulate synaptic function in response to excitation. In particular, the roles of alternative pre-mRNA splicing and mRNA translation regulation in this response are poorly understood. We will study the RNA-binding proteins (RBPs) that control these post-transcriptional changes using a UV crosslinking-based purification method (CLIP) and ultra-high throughput sequencing. Computational analysis of the resulting data will define the sequence and structural features of RNA motifs recognized by each RBP. Splicing microarrays and translation reporter assays will then allow us to examine the regulatory functions of RBPs and RNA motifs. By integrating the biochemical and functional datasets, we will relate the position of RNA motifs to the activity of bound RBPs, and predict the interactions that act as central nodes in the regulatory network. The physiological role of these core RBP-RNA interactions will then be tested using antisense RNAs. Together, these projects will provide insights to the regulatory mechanisms underlying neuronal activity-dependent changes, and provide new opportunities for future treatments of neurodegenerative disorders.
Max ERC Funding
900 000 €
Duration
Start date: 2008-09-01, End date: 2013-08-31
Project acronym Clock Mechanics
Project Mechanosensation and the circadian clock: a reciprocal analysis
Researcher (PI) Joerg Albert
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Consolidator Grant (CoG), LS5, ERC-2014-CoG
Summary All forms of life adjust themselves to the daily rhythms of their environments using endogenous oscillators collectively referred to as circadian clocks. Peripheral and central body clocks exist, which both require extrinsic information (e.g. light or temperature changes) to keep in sync with the geophysical cycle (entrainment). In addition, intrinsic cues (e.g. activity levels) have been linked to clock entrainment. Recently, we could show that activation of proprioceptors is sufficient to entrain the central clock of the fruit fly Drosophila melanogaster. Proprioceptors are mechanosensors that monitor the positions, and relative movements, of an animal’s own body parts. The existence of proprioceptive entrainment pathways has significant implications; it implies that an animal’s ‘clock time’ is computed by integrating, and weighting, various external and internal conditions, suggesting the existence of external and internal time.
Using Drosophila, I will investigate the relationship between mechanosensory and circadian systems in a dual, and bidirectional, approach. The project’s first aim is to dissect the neurobiological bases of proprioceptive clock entrainment (i) identifying the specific stimulus requirements for effective entrainment, (ii) determining its mechanosensory pathways and, in a combined computational and experimental strategy, (iii) quantifying the precise contributions of an animal’s activity to its sense of time. The project’s second aim, in turn, is to unravel the roles of the clock, and clock genes, for the function of mechanosensory systems. Previous studies have linked the clock to noise vulnerability in mammalian ears, and clock genes to regeneration in avian ears. Our own preliminary data reveal severe mechanosensory defects in flies mutant for core clock genes. I will use the Drosophila ear as a unifying model to analyse the specific roles of the clock, and clock genes, for the function of mechanotransducer systems.
Summary
All forms of life adjust themselves to the daily rhythms of their environments using endogenous oscillators collectively referred to as circadian clocks. Peripheral and central body clocks exist, which both require extrinsic information (e.g. light or temperature changes) to keep in sync with the geophysical cycle (entrainment). In addition, intrinsic cues (e.g. activity levels) have been linked to clock entrainment. Recently, we could show that activation of proprioceptors is sufficient to entrain the central clock of the fruit fly Drosophila melanogaster. Proprioceptors are mechanosensors that monitor the positions, and relative movements, of an animal’s own body parts. The existence of proprioceptive entrainment pathways has significant implications; it implies that an animal’s ‘clock time’ is computed by integrating, and weighting, various external and internal conditions, suggesting the existence of external and internal time.
Using Drosophila, I will investigate the relationship between mechanosensory and circadian systems in a dual, and bidirectional, approach. The project’s first aim is to dissect the neurobiological bases of proprioceptive clock entrainment (i) identifying the specific stimulus requirements for effective entrainment, (ii) determining its mechanosensory pathways and, in a combined computational and experimental strategy, (iii) quantifying the precise contributions of an animal’s activity to its sense of time. The project’s second aim, in turn, is to unravel the roles of the clock, and clock genes, for the function of mechanosensory systems. Previous studies have linked the clock to noise vulnerability in mammalian ears, and clock genes to regeneration in avian ears. Our own preliminary data reveal severe mechanosensory defects in flies mutant for core clock genes. I will use the Drosophila ear as a unifying model to analyse the specific roles of the clock, and clock genes, for the function of mechanotransducer systems.
Max ERC Funding
1 899 549 €
Duration
Start date: 2015-09-01, End date: 2021-08-31
Project acronym CLUE-BGD
Project Closing the Loop between Understanding and Effective Treatment of the Basal Ganglia and their Disorders
Researcher (PI) Hagai Bergman
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS5, ERC-2012-ADG_20120314
Summary In this project, the basal ganglia are defined as actor-critic reinforcement learning networks that aim at an optimal tradeoff between the maximization of future cumulative rewards and the minimization of the cost (the reinforcement driven multi objective optimization RDMOO model).
This computational model will be tested by multiple neuron recordings in the major basal ganglia structures of monkeys engaged in a similar behavioral task. We will further validate the RMDOO computational model of the basal ganglia by extending our previous studies of neural activity in the MPTP primate model of Parkinson's disease to a primate model of central serotonin depletion and emotional dysregulation disorders. The findings in the primate model of emotional dysregulation will then be compared to electrophysiological recordings carried out in human patients with treatment-resistant major depression and obsessive compulsive disorder during deep brain stimulation (DBS) procedures. I aim to find neural signatures (e.g., synchronous gamma oscillations in the actor part of the basal ganglia as predicted by the RMDOO model) characterizing these emotional disorders and to use them as triggers for closed loop adaptive DBS. Our working hypothesis holds that, as for the MPTP model of Parkinson's disease, closed loop DBS will lead to greater amelioration of the emotional deficits in serotonin depleted monkeys.
This project incorporates extensive collaborations with a team of neurosurgeons, neurologists, psychiatrists, and computer science/ neural network researchers. If successful, the findings will provide a firm understanding of the computational physiology of the basal ganglia networks and their disorders. Importantly, they will pave the way to better treatment of human patients with severe mental disorders.
Summary
In this project, the basal ganglia are defined as actor-critic reinforcement learning networks that aim at an optimal tradeoff between the maximization of future cumulative rewards and the minimization of the cost (the reinforcement driven multi objective optimization RDMOO model).
This computational model will be tested by multiple neuron recordings in the major basal ganglia structures of monkeys engaged in a similar behavioral task. We will further validate the RMDOO computational model of the basal ganglia by extending our previous studies of neural activity in the MPTP primate model of Parkinson's disease to a primate model of central serotonin depletion and emotional dysregulation disorders. The findings in the primate model of emotional dysregulation will then be compared to electrophysiological recordings carried out in human patients with treatment-resistant major depression and obsessive compulsive disorder during deep brain stimulation (DBS) procedures. I aim to find neural signatures (e.g., synchronous gamma oscillations in the actor part of the basal ganglia as predicted by the RMDOO model) characterizing these emotional disorders and to use them as triggers for closed loop adaptive DBS. Our working hypothesis holds that, as for the MPTP model of Parkinson's disease, closed loop DBS will lead to greater amelioration of the emotional deficits in serotonin depleted monkeys.
This project incorporates extensive collaborations with a team of neurosurgeons, neurologists, psychiatrists, and computer science/ neural network researchers. If successful, the findings will provide a firm understanding of the computational physiology of the basal ganglia networks and their disorders. Importantly, they will pave the way to better treatment of human patients with severe mental disorders.
Max ERC Funding
2 476 922 €
Duration
Start date: 2013-12-01, End date: 2018-11-30
Project acronym CMetC
Project Selective Carbon-Carbon Bond Activation: A Wellspring of Untapped Reactivity
Researcher (PI) Ilan Marek
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Advanced Grant (AdG), PE5, ERC-2013-ADG
Summary The creation of new molecular entities and subsequent exploitation of their properties is central to a broad spectrum of research disciplines from medicine to materials. Most –if not all- of the efforts of organic chemists were directed to the development of creative strategies to built carbon-carbon and carbon-heteroatom bonds in a predictable and efficient manner. But is the creation of new bonds the only approach that organic chemistry should follow? Could we design the synthesis of challenging molecular skeleton no more through the construction of carbon-carbon bonds but rather through selective cleavage of carbon-carbon bonds (C-C bond activation)? The goal of this work is to develop powerful synthetic approaches for the selective C-C bond activation and demonstrate that it has the potential to be a general principle in organic synthesis for the regio-, diastereo- and even enantiomerically enriched preparation of adducts despite that C-C single bonds belong among the least reactive functional groups in chemistry. The realization of this synthetic potential requires the ability to functionalize selectively one C-C bond in compounds containing many such bonds and an array of functional groups. This site selective C-C bond activation is one of the greatest challenges that must be met to be used widely in complex-molecular synthesis. To emphasize the practicality of C-C bond activation, we will prepare in a single-pot operation challenging molecular framework possessing various stereogenic centers from very simple starting materials through selective C-C bond activation. Ideally, alkenes will be in-situ transformed into alkanes that will subsequently undergo the C-C activation even in the presence of functional group. This work will lead to ground-breaking advances when non-strained cycloalkanes (cyclopentane, cyclohexane) will undergo this smooth C-C bond activation with friendly and non toxic organometallic species.
Summary
The creation of new molecular entities and subsequent exploitation of their properties is central to a broad spectrum of research disciplines from medicine to materials. Most –if not all- of the efforts of organic chemists were directed to the development of creative strategies to built carbon-carbon and carbon-heteroatom bonds in a predictable and efficient manner. But is the creation of new bonds the only approach that organic chemistry should follow? Could we design the synthesis of challenging molecular skeleton no more through the construction of carbon-carbon bonds but rather through selective cleavage of carbon-carbon bonds (C-C bond activation)? The goal of this work is to develop powerful synthetic approaches for the selective C-C bond activation and demonstrate that it has the potential to be a general principle in organic synthesis for the regio-, diastereo- and even enantiomerically enriched preparation of adducts despite that C-C single bonds belong among the least reactive functional groups in chemistry. The realization of this synthetic potential requires the ability to functionalize selectively one C-C bond in compounds containing many such bonds and an array of functional groups. This site selective C-C bond activation is one of the greatest challenges that must be met to be used widely in complex-molecular synthesis. To emphasize the practicality of C-C bond activation, we will prepare in a single-pot operation challenging molecular framework possessing various stereogenic centers from very simple starting materials through selective C-C bond activation. Ideally, alkenes will be in-situ transformed into alkanes that will subsequently undergo the C-C activation even in the presence of functional group. This work will lead to ground-breaking advances when non-strained cycloalkanes (cyclopentane, cyclohexane) will undergo this smooth C-C bond activation with friendly and non toxic organometallic species.
Max ERC Funding
2 367 495 €
Duration
Start date: 2013-11-01, End date: 2018-10-31
Project acronym CNTBBB
Project Targeting potential of carbon nanotubes at the blood brain barrier
Researcher (PI) Alexandra Elizabeth Porter
Host Institution (HI) IMPERIAL COLLEGE OF SCIENCE TECHNOLOGY AND MEDICINE
Call Details Starting Grant (StG), PE5, ERC-2010-StG_20091028
Summary Targeted drug delivery across the blood brain barrier (BBB) to the central nervous system is a large challenge for the treatment of neurological disorders. This 4 year ERC program is aimed towards the evaluating the BBB penetration capacity and toxicological potential of novel carbon nanotube (CNT) carriers using an integrated multidisciplinary approach. State-of-art characterisation techniques developed by the PI will be applied and further developed to detect the interaction of carbon nanotubes with in vitro BBB model and neuronal cells. Specific aims:
1. Identify the mechanisms of translocation of CNT across the endothelial cells which comprise the BBB, as well as uptake by neuronal cells in vitro.
2. To investigate the effect of length, diameter and surface charge of CNTs on the BBB and neuronal cells penetration capacity in vitro.
3. To investigate the toxicological profile of CNT on the BBB and the various neuronal cell types (immortalised and primary neuronal cultures).
4. Develop protocols to assess whether the CNTs degrade inside the cell.
The ERC Grant will consolidate the new Research Group in nanomaterials-cell interfaces, and allow them to perform stimulating investigator-initiated frontier research in nanotoxicology and nanomedicine. To this end, a multi-disciplinary laboratory will be realized within the framework of this 4-year the ERC Programme. This will permit the group around the PI, to expand activities, push limits, create new boundaries, and develop new protocols for studying nanoparticle-cell interactions in close collaboration with ICL s Department of medicine and chemistry. Within the proposed program there is an underlying ambition both to gain a fundamental understanding for which parameters of CNTs determine their penetration capacity through the BBB and also to assess their toxicological potential at the BBB two highlighted themes by the ERC.
Summary
Targeted drug delivery across the blood brain barrier (BBB) to the central nervous system is a large challenge for the treatment of neurological disorders. This 4 year ERC program is aimed towards the evaluating the BBB penetration capacity and toxicological potential of novel carbon nanotube (CNT) carriers using an integrated multidisciplinary approach. State-of-art characterisation techniques developed by the PI will be applied and further developed to detect the interaction of carbon nanotubes with in vitro BBB model and neuronal cells. Specific aims:
1. Identify the mechanisms of translocation of CNT across the endothelial cells which comprise the BBB, as well as uptake by neuronal cells in vitro.
2. To investigate the effect of length, diameter and surface charge of CNTs on the BBB and neuronal cells penetration capacity in vitro.
3. To investigate the toxicological profile of CNT on the BBB and the various neuronal cell types (immortalised and primary neuronal cultures).
4. Develop protocols to assess whether the CNTs degrade inside the cell.
The ERC Grant will consolidate the new Research Group in nanomaterials-cell interfaces, and allow them to perform stimulating investigator-initiated frontier research in nanotoxicology and nanomedicine. To this end, a multi-disciplinary laboratory will be realized within the framework of this 4-year the ERC Programme. This will permit the group around the PI, to expand activities, push limits, create new boundaries, and develop new protocols for studying nanoparticle-cell interactions in close collaboration with ICL s Department of medicine and chemistry. Within the proposed program there is an underlying ambition both to gain a fundamental understanding for which parameters of CNTs determine their penetration capacity through the BBB and also to assess their toxicological potential at the BBB two highlighted themes by the ERC.
Max ERC Funding
1 229 998 €
Duration
Start date: 2011-02-01, End date: 2017-01-31
Project acronym COFBMIX
Project Cortical feedback in figure background segregation of odors.
Researcher (PI) Dan ROKNI
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS5, ERC-2017-STG
Summary A key question in neuroscience is how information is processed by sensory systems to guide behavior. Most of our knowledge about sensory processing is based on presentation of simple isolated stimuli and recording corresponding neural activity in relevant brain areas. Yet sensory stimuli in real life are never isolated and typically not simple. How the brain processes complex stimuli, simultaneously arising from multiple objects is unknown. Our daily experience (as well as well-controlled experiments) shows that only parts of a complex sensory scene can be perceived - we cannot listen to more than one speaker in a party. Importantly, one can easily choose what is important and should be processed and what can be ignored as background. These observations lead to the prevalent hypothesis that feedback projections from ‘higher’ brain areas to more peripheral sensory areas are involved in processing of complex stimuli. However experimental analysis of signals conveyed by feedback projections in behaving animals is scarce. The nature of these signals and how they relate to behavior is unknown.
Here I propose a cutting edge approach to directly record feedback signals in the olfactory system of behaving mice. We will use chronically implanted electrodes to record the modulation of olfactory bulb (OB) principal neurons by task related context. Additionally, we will record from piriform cortical (PC) neurons that project back to the OB. These will be tagged with channelrhodopsin-2 and identified by light sensitivity. Finally, we will express the spectrally distinct Ca++ indicators GCaMP6 and RCaMP2 in PC neurons and in olfactory sensory neurons, respectively, and use 2-photon microscopy to analyze the spatio-temporal relationship between feedforward and feedback inputs in the OB. This comprehensive approach will provide an explanation of how feedforward and feedback inputs are integrated to process complex stimuli.
Summary
A key question in neuroscience is how information is processed by sensory systems to guide behavior. Most of our knowledge about sensory processing is based on presentation of simple isolated stimuli and recording corresponding neural activity in relevant brain areas. Yet sensory stimuli in real life are never isolated and typically not simple. How the brain processes complex stimuli, simultaneously arising from multiple objects is unknown. Our daily experience (as well as well-controlled experiments) shows that only parts of a complex sensory scene can be perceived - we cannot listen to more than one speaker in a party. Importantly, one can easily choose what is important and should be processed and what can be ignored as background. These observations lead to the prevalent hypothesis that feedback projections from ‘higher’ brain areas to more peripheral sensory areas are involved in processing of complex stimuli. However experimental analysis of signals conveyed by feedback projections in behaving animals is scarce. The nature of these signals and how they relate to behavior is unknown.
Here I propose a cutting edge approach to directly record feedback signals in the olfactory system of behaving mice. We will use chronically implanted electrodes to record the modulation of olfactory bulb (OB) principal neurons by task related context. Additionally, we will record from piriform cortical (PC) neurons that project back to the OB. These will be tagged with channelrhodopsin-2 and identified by light sensitivity. Finally, we will express the spectrally distinct Ca++ indicators GCaMP6 and RCaMP2 in PC neurons and in olfactory sensory neurons, respectively, and use 2-photon microscopy to analyze the spatio-temporal relationship between feedforward and feedback inputs in the OB. This comprehensive approach will provide an explanation of how feedforward and feedback inputs are integrated to process complex stimuli.
Max ERC Funding
1 500 000 €
Duration
Start date: 2018-04-01, End date: 2023-03-31
Project acronym COLORLANDS
Project COLOR Ordering Templated by Hierarchized Supramolecular Porous FlatLANDS
Researcher (PI) Davide Bonifazi
Host Institution (HI) CARDIFF UNIVERSITY
Call Details Starting Grant (StG), PE5, ERC-2011-StG_20101014
Summary The idea of this research project is to take advantage of molecular self-assembly to create a new generation of periodically-organized porous organic materials that, acting as specific molecular hosts, can structurally control the positioning of multiple functional guests on surfaces, opening new horizons toward the understanding and development of rationale protocols for the patterning of unprecedented materials. Taking advantage of a supramolecular approach to engineer extended mono- and two-dimensional organic networks, the ultimate aim of COLORLANDS is to create novel hosting frameworks accommodating in a predetermined fashion organic chromophores and/or fluorophores. For instance, these can be oligophenylenes as blue emitters, cumarines/oligophenylethylenes as green emitters, or perylenebisimides conjugates as red emitters. Depending on their spatial organization, such materials will be the springboard for further technological development in the fields of electroluminescent devices or artificial leafs mimicking natural light harvesting antenna systems. The self-assembly of selected rigid molecular modules alternatively functionalized with complementary connectors (PNA strands) will yield, under equilibrium conditions, one exclusive structural pattern. This will feature controllable (in shape, size and chemical nature) periodic receptor sites, each programmed to selectively accommodate a specific molecular chromophore and/or fluorophore. Particular attention will be given to the design and fundamental understanding of specific orthogonal interactions between the self-assembled receptor sites and the functional molecular guests. This will be achieved through the lateral organic functionalization of the PNA strands with novel orthogonal H-bonding-based recognition motifs. Depending on the ratio between the different receptors, one can tailor the desired emission or absorption colour, virtually enabling unlimited surfing through the color coordinate diagram.
Summary
The idea of this research project is to take advantage of molecular self-assembly to create a new generation of periodically-organized porous organic materials that, acting as specific molecular hosts, can structurally control the positioning of multiple functional guests on surfaces, opening new horizons toward the understanding and development of rationale protocols for the patterning of unprecedented materials. Taking advantage of a supramolecular approach to engineer extended mono- and two-dimensional organic networks, the ultimate aim of COLORLANDS is to create novel hosting frameworks accommodating in a predetermined fashion organic chromophores and/or fluorophores. For instance, these can be oligophenylenes as blue emitters, cumarines/oligophenylethylenes as green emitters, or perylenebisimides conjugates as red emitters. Depending on their spatial organization, such materials will be the springboard for further technological development in the fields of electroluminescent devices or artificial leafs mimicking natural light harvesting antenna systems. The self-assembly of selected rigid molecular modules alternatively functionalized with complementary connectors (PNA strands) will yield, under equilibrium conditions, one exclusive structural pattern. This will feature controllable (in shape, size and chemical nature) periodic receptor sites, each programmed to selectively accommodate a specific molecular chromophore and/or fluorophore. Particular attention will be given to the design and fundamental understanding of specific orthogonal interactions between the self-assembled receptor sites and the functional molecular guests. This will be achieved through the lateral organic functionalization of the PNA strands with novel orthogonal H-bonding-based recognition motifs. Depending on the ratio between the different receptors, one can tailor the desired emission or absorption colour, virtually enabling unlimited surfing through the color coordinate diagram.
Max ERC Funding
1 295 400 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym CoMMaD
Project Computational Molecular Materials Discovery
Researcher (PI) Kim JELFS
Host Institution (HI) IMPERIAL COLLEGE OF SCIENCE TECHNOLOGY AND MEDICINE
Call Details Starting Grant (StG), PE5, ERC-2017-STG
Summary The objective of the project is to develop a computational approach to accelerate the discovery of molecular materials. These materials will include porous molecules, small organic molecules and macromolecular polymers, which have application as a result of either their porosity or optoelectronic properties. The applications that will be targeted include in molecular separations, sensing, (photo)catalysis and photovoltaics. To achieve my aims, I will screen libraries of building blocks through a combination of techniques including evolutionary algorithms and machine learning. Through the application of cheminformatics algorithms, I will target the most promising libraries, assess synthetic diversity and accessibility and analyse structure-property relationships. I will develop software that will predict the (macro)molecular structures and properties; the molecular property screening calculations will include void characterisation, binding energies, diffusion barriers, local assembly, charge transport and energy level assessment. A consideration of synthetic accessibility at every stage will be central to my approach, which will ensure the realisation of our predicted targets. I have several synthetic collaborators who can provide pathways to synthetic realisation. Improved materials in this field have the potential to either reduce our energy needs or provide renewable energy, helping the EU meet the targets of the 2030 Energy Strategy.
Summary
The objective of the project is to develop a computational approach to accelerate the discovery of molecular materials. These materials will include porous molecules, small organic molecules and macromolecular polymers, which have application as a result of either their porosity or optoelectronic properties. The applications that will be targeted include in molecular separations, sensing, (photo)catalysis and photovoltaics. To achieve my aims, I will screen libraries of building blocks through a combination of techniques including evolutionary algorithms and machine learning. Through the application of cheminformatics algorithms, I will target the most promising libraries, assess synthetic diversity and accessibility and analyse structure-property relationships. I will develop software that will predict the (macro)molecular structures and properties; the molecular property screening calculations will include void characterisation, binding energies, diffusion barriers, local assembly, charge transport and energy level assessment. A consideration of synthetic accessibility at every stage will be central to my approach, which will ensure the realisation of our predicted targets. I have several synthetic collaborators who can provide pathways to synthetic realisation. Improved materials in this field have the potential to either reduce our energy needs or provide renewable energy, helping the EU meet the targets of the 2030 Energy Strategy.
Max ERC Funding
1 499 390 €
Duration
Start date: 2018-04-01, End date: 2023-03-31
Project acronym COMPLEXORDER
Project The Complexity Revolution: Exploiting Unconventional Order in Next-Generation Materials Design
Researcher (PI) Andrew GOODWIN
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Advanced Grant (AdG), PE5, ERC-2017-ADG
Summary The fundamental objective of the research described in this proposal is to lay the foundations for understanding how structural complexity can give rise to materials properties inaccessible to structurally-simple states. The long-term vision is a paradigm shift in the way we as chemists design materials—the “Complexity Revolution”—where we move to thinking beyond the unit cell and harness unconventional order to generate emergent states with entirely novel behaviour. The key methodologies of the project are (i) exploitation of the rich structural information accessible using 3D-PDF / diffuse scattering techniques, (ii) exploration of the phase behaviour of unconventional ordered states using computational methods, and (iii) experimental/computational studies of a broad range of materials in which complexity arises from a large variety of different phenemona. In this way, the project will establish how we might controllably introduce complexity into materials by varying chemical composition and synthesis, how we might then characterise these complex states, and how we might exploit this complexity when designing next-generation materials with unprecedented electronic, catalytic, photonic, information storage, dielectric, topological, and magnetic properties.
Summary
The fundamental objective of the research described in this proposal is to lay the foundations for understanding how structural complexity can give rise to materials properties inaccessible to structurally-simple states. The long-term vision is a paradigm shift in the way we as chemists design materials—the “Complexity Revolution”—where we move to thinking beyond the unit cell and harness unconventional order to generate emergent states with entirely novel behaviour. The key methodologies of the project are (i) exploitation of the rich structural information accessible using 3D-PDF / diffuse scattering techniques, (ii) exploration of the phase behaviour of unconventional ordered states using computational methods, and (iii) experimental/computational studies of a broad range of materials in which complexity arises from a large variety of different phenemona. In this way, the project will establish how we might controllably introduce complexity into materials by varying chemical composition and synthesis, how we might then characterise these complex states, and how we might exploit this complexity when designing next-generation materials with unprecedented electronic, catalytic, photonic, information storage, dielectric, topological, and magnetic properties.
Max ERC Funding
3 362 635 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym CONFINEDCHEM
Project Synthetic Confined Environments as Tools for Manipulating Chemical Reactivities and Preparing New Nanostructures
Researcher (PI) Rafal Klajn
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), PE5, ERC-2013-StG
Summary "Nature has long inspired chemists with its abilities to stabilize ephemeral chemical species, to perform chemical reactions with unprecedented rates and selectivities, and to synthesize complex molecules and fascinating inorganic nanostructures. What natural systems consistently exploit - which is yet fundamentally different from how chemists perform reactions - is their aspect of nanoscale confinement. The goal of the proposed research program is to integrate the worlds of organic and inorganic colloidal chemistry by means of manipulating chemical reactivities and synthesizing novel molecules and nanostructures inside synthetic confined environments created using novel, unconventional approaches based on inorganic, nanostructured building blocks. The three types of confined spaces we propose are as follows: 1) nanopores within reversibly self-assembling colloidal crystals (""dynamic nanoflasks""), 2) cavities of bowl-shaped metallic nanoparticles (NPs), and 3) surfaces of spherical NPs. By taking advantage of these unique tools, we will attempt to develop, respectively, 1) a conceptually new method for catalyzing chemical reactions using light, 2) nanoscale inclusion chemistry (a field based on host-guest ""complexes"" assembled form nanosized components) and 3) to use NPs as platforms for the development of new organic reactions. While these objectives are predominantly of a fundamental nature, they can easily evolve into a variety of practical applications. Specifically, we will pursue diverse goals such as the preparation of 1) a new family of inverse opals (with potentially fascinating optical and mechanical properties), 2) artificial chaperones (NPs assisting in protein folding), and 3) size- and shape-controlled polymeric vesicles. Overall, it is believed that this marriage of organic and colloidal chemistry has the potential to change the fundamental way we perform chemical reactions, paving the way to the discovery of new phenomena and unique structures."
Summary
"Nature has long inspired chemists with its abilities to stabilize ephemeral chemical species, to perform chemical reactions with unprecedented rates and selectivities, and to synthesize complex molecules and fascinating inorganic nanostructures. What natural systems consistently exploit - which is yet fundamentally different from how chemists perform reactions - is their aspect of nanoscale confinement. The goal of the proposed research program is to integrate the worlds of organic and inorganic colloidal chemistry by means of manipulating chemical reactivities and synthesizing novel molecules and nanostructures inside synthetic confined environments created using novel, unconventional approaches based on inorganic, nanostructured building blocks. The three types of confined spaces we propose are as follows: 1) nanopores within reversibly self-assembling colloidal crystals (""dynamic nanoflasks""), 2) cavities of bowl-shaped metallic nanoparticles (NPs), and 3) surfaces of spherical NPs. By taking advantage of these unique tools, we will attempt to develop, respectively, 1) a conceptually new method for catalyzing chemical reactions using light, 2) nanoscale inclusion chemistry (a field based on host-guest ""complexes"" assembled form nanosized components) and 3) to use NPs as platforms for the development of new organic reactions. While these objectives are predominantly of a fundamental nature, they can easily evolve into a variety of practical applications. Specifically, we will pursue diverse goals such as the preparation of 1) a new family of inverse opals (with potentially fascinating optical and mechanical properties), 2) artificial chaperones (NPs assisting in protein folding), and 3) size- and shape-controlled polymeric vesicles. Overall, it is believed that this marriage of organic and colloidal chemistry has the potential to change the fundamental way we perform chemical reactions, paving the way to the discovery of new phenomena and unique structures."
Max ERC Funding
1 499 992 €
Duration
Start date: 2013-10-01, End date: 2018-09-30
Project acronym CONTREX
Project Controlling Triplet Excitons in Organic Semiconductors
Researcher (PI) Hugo Bronstein
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Starting Grant (StG), PE5, ERC-2015-STG
Summary The urgent need to reduce carbon emissions in order to mitigate climate change requires the development of clean, renewable energy sources. Solar power offers a virtually unlimited supply of energy, providing it can be harnessed efficiently. Traditional silicon solar cells demonstrate high performance (~20%) but their required method of manufacture prohibits large area production rendering them too expensive to be used on a global scale. Organic solar cells (made from conjugated polymers and fullerenes) have the potential to be fabricated by low cost printing methods allowing for large scale modules to be produced cheaply. Conventional organic solar cells function by generating charge from a singlet excited state. In order to achieve optimum performance the precise morphology of polymer and fullerene must be controlled which can be extremely challenging. These devices however, have attained good efficiencies (10%) but are hampered by severe loss mechanisms which generally involve the formation of a lower energy triplet excited state.
We propose to develop novel materials for organic solar cells which will instead utilise this triplet excited state to generate charges. This will enable us to not only eliminate this loss mechanism but due to the unique properties of the triplet excited state will allow for numerous benefits. Firstly, the long lifetime of the triplet excited state will be exploited to allow for a simpler organic solar cell where precise morphological control is not required. Secondly, the proposed new materials will allow for the utilisation of near-IR light which is typically wasted in ALL current solar cell devices. Thirdly, exploiting a unique photophysical process we will produce materials capable of delivering efficiencies in excess of the theoretical limit available to conventional solar cells. Thus we propose that utilisation of triplet excitons is the required step-change to allow for organic solar cells to achieve their ultimate efficiencies
Summary
The urgent need to reduce carbon emissions in order to mitigate climate change requires the development of clean, renewable energy sources. Solar power offers a virtually unlimited supply of energy, providing it can be harnessed efficiently. Traditional silicon solar cells demonstrate high performance (~20%) but their required method of manufacture prohibits large area production rendering them too expensive to be used on a global scale. Organic solar cells (made from conjugated polymers and fullerenes) have the potential to be fabricated by low cost printing methods allowing for large scale modules to be produced cheaply. Conventional organic solar cells function by generating charge from a singlet excited state. In order to achieve optimum performance the precise morphology of polymer and fullerene must be controlled which can be extremely challenging. These devices however, have attained good efficiencies (10%) but are hampered by severe loss mechanisms which generally involve the formation of a lower energy triplet excited state.
We propose to develop novel materials for organic solar cells which will instead utilise this triplet excited state to generate charges. This will enable us to not only eliminate this loss mechanism but due to the unique properties of the triplet excited state will allow for numerous benefits. Firstly, the long lifetime of the triplet excited state will be exploited to allow for a simpler organic solar cell where precise morphological control is not required. Secondly, the proposed new materials will allow for the utilisation of near-IR light which is typically wasted in ALL current solar cell devices. Thirdly, exploiting a unique photophysical process we will produce materials capable of delivering efficiencies in excess of the theoretical limit available to conventional solar cells. Thus we propose that utilisation of triplet excitons is the required step-change to allow for organic solar cells to achieve their ultimate efficiencies
Max ERC Funding
1 499 223 €
Duration
Start date: 2016-04-01, End date: 2021-03-31
Project acronym COORDSPACE
Project Chemistry of Coordination Space: Extraction, Storage, Activation and Catalysis
Researcher (PI) Martin Schroder
Host Institution (HI) THE UNIVERSITY OF NOTTINGHAM
Call Details Advanced Grant (AdG), PE5, ERC-2008-AdG
Summary The Applicant has an outstanding record of achievement and an international reputation for independent research across many areas of metal coordination chemistry. This high-impact and challenging Proposal brings together innovative ideas in coordination chemistry within a single inter- and multi-disciplinary project to open up new horizons across molecular and biological sciences, materials science and energy research. The Proposal applies coordination chemistry to the key issues of climate change, environmental and chemical sustainability, the Hydrogen Economy, carbon capture and fuel cell technologies, and atom-efficient metal extraction and clean-up. The vision is to bring together complementary areas and new applications of metal coordination chemistry and ligand design within an overarching and fundamental research program addressing: i. nanoscale functionalized framework polymers for the storage and activation of H2, CO2, CO, O2, N2, methane and volatile organic compounds; ii. new catalysts for the reversible oxidation and photochemical production of H2; iii) clean and selective recovery of precious metals (Pt, Pd, Rh, Ir, Hf, Zr) from process streams and ores. These research themes will be consolidated within a single cross-disciplinary and ambitious program focusing on the control of chemistry, reactivity and interactions within self-assembled confined and multi-functionalized space generated by designer porous framework materials. An AdG will afford the impetus and freedom via consolidated funding to undertake fundamental, speculative research with multiple potential big-hits across a wide range of disciplines. Via an extensive network of international academic and industrial collaborations, the Applicant will deliver major research breakthroughs in these vital areas, and train scientists for the future of Europe in an exciting, stimulating and curiosity-driven environment.
Summary
The Applicant has an outstanding record of achievement and an international reputation for independent research across many areas of metal coordination chemistry. This high-impact and challenging Proposal brings together innovative ideas in coordination chemistry within a single inter- and multi-disciplinary project to open up new horizons across molecular and biological sciences, materials science and energy research. The Proposal applies coordination chemistry to the key issues of climate change, environmental and chemical sustainability, the Hydrogen Economy, carbon capture and fuel cell technologies, and atom-efficient metal extraction and clean-up. The vision is to bring together complementary areas and new applications of metal coordination chemistry and ligand design within an overarching and fundamental research program addressing: i. nanoscale functionalized framework polymers for the storage and activation of H2, CO2, CO, O2, N2, methane and volatile organic compounds; ii. new catalysts for the reversible oxidation and photochemical production of H2; iii) clean and selective recovery of precious metals (Pt, Pd, Rh, Ir, Hf, Zr) from process streams and ores. These research themes will be consolidated within a single cross-disciplinary and ambitious program focusing on the control of chemistry, reactivity and interactions within self-assembled confined and multi-functionalized space generated by designer porous framework materials. An AdG will afford the impetus and freedom via consolidated funding to undertake fundamental, speculative research with multiple potential big-hits across a wide range of disciplines. Via an extensive network of international academic and industrial collaborations, the Applicant will deliver major research breakthroughs in these vital areas, and train scientists for the future of Europe in an exciting, stimulating and curiosity-driven environment.
Max ERC Funding
2 492 372 €
Duration
Start date: 2008-12-01, End date: 2013-11-30
Project acronym CORTEX
Project Computations by Neurons and Populations in Visual Cortex
Researcher (PI) Matteo Carandini
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Advanced Grant (AdG), LS5, ERC-2008-AdG
Summary Neurons in primary visual cortex (area V1) receive feedforward inputs from thalamic afferents and lateral inputs from other cortical neurons. Little is known about how these components interact to determine the responses of a V1 neuron. One camp ascribes most responses to feedforward mechanisms. The other camp ascribes them mostly to lateral interactions. We propose that these two apparently opposed views can be simply reconciled in a single framework. We hypothesize that area V1 can operate both in a feedforward regime and in a lateral interaction regime, depending on the nature of the stimulus and on the cognitive task at hand, and that the transition from one regime to the other is governed by synaptic inhibition. We will test these hypotheses by recording from individual V1 neurons while monitoring the activity of nearby populations of cortical neurons via multiprobe electrodes. In Aim 1 we will relate the activity of V1 neurons to that of nearby populations. We will use simple measures of correlation and nonlinear models that predict individual spikes to measure how responses depend on a feedforward contribution (the receptive field ) and on a lateral contribution (the connection field ). We will test our first hypothesis, concerning the role of the stimulus in changing this dependence. In Aim 2 we will extend these results to a behaving animal. We will record from V1 of mice performing a 2-alternative forced-choice psychophysical task, and we will test our second hypothesis, concerning the role of the cognitive task in determining the operating regime of the cortex. In Aim 3 we will seek a biophysical interpretation of the functional mechanisms and effective connectivity revealed by the previous Aims. We will test our third hypothesis, concerning the role of synaptic inhibition. The tools involved will include intracellular recordings and optical stimulation in transgenic mice whose cortical neurons are sensitive to light.
Summary
Neurons in primary visual cortex (area V1) receive feedforward inputs from thalamic afferents and lateral inputs from other cortical neurons. Little is known about how these components interact to determine the responses of a V1 neuron. One camp ascribes most responses to feedforward mechanisms. The other camp ascribes them mostly to lateral interactions. We propose that these two apparently opposed views can be simply reconciled in a single framework. We hypothesize that area V1 can operate both in a feedforward regime and in a lateral interaction regime, depending on the nature of the stimulus and on the cognitive task at hand, and that the transition from one regime to the other is governed by synaptic inhibition. We will test these hypotheses by recording from individual V1 neurons while monitoring the activity of nearby populations of cortical neurons via multiprobe electrodes. In Aim 1 we will relate the activity of V1 neurons to that of nearby populations. We will use simple measures of correlation and nonlinear models that predict individual spikes to measure how responses depend on a feedforward contribution (the receptive field ) and on a lateral contribution (the connection field ). We will test our first hypothesis, concerning the role of the stimulus in changing this dependence. In Aim 2 we will extend these results to a behaving animal. We will record from V1 of mice performing a 2-alternative forced-choice psychophysical task, and we will test our second hypothesis, concerning the role of the cognitive task in determining the operating regime of the cortex. In Aim 3 we will seek a biophysical interpretation of the functional mechanisms and effective connectivity revealed by the previous Aims. We will test our third hypothesis, concerning the role of synaptic inhibition. The tools involved will include intracellular recordings and optical stimulation in transgenic mice whose cortical neurons are sensitive to light.
Max ERC Funding
2 499 921 €
Duration
Start date: 2009-04-01, End date: 2014-03-31
Project acronym CORTICAL ASSEMBLY
Project Excitatory and inhibitory cell assemblies
in the cerebral cortex
Researcher (PI) Oscar Marin Parra
Host Institution (HI) KING'S COLLEGE LONDON
Call Details Advanced Grant (AdG), LS5, ERC-2011-ADG_20110310
Summary The neural assembly underlying the formation of functional networks in the cerebral cortex is conceivably the most complex biological system that exists. Much of this complexity arises during development through the interaction of dozens of different neuronal populations, which belong to two general classes: excitatory glutamatergic pyramidal cells and inhibitory gamma-aminobutyric containing (GABAergic) interneurons. Perhaps the most fascinating aspect of the assembly of cortical circuits is that pyramidal cells and interneurons are generated in distant germinal zones. Pyramidal cells are born locally from progenitors located in the cortical anlage, while interneurons derive from progenitors in the embryonic subpallium. Much progress has been made recently in understanding the molecular mechanisms that regulate the migration of interneurons towards the cortex, but how interneurons find their appropriate partners to build cortical networks with balanced excitation and inhibition remains an enigma.
The general goal of this project is to identify the mechanisms controlling the precise allocation of different classes of interneurons into specific layers of the cortex, where they assemble into neural circuits. We also aim to determine how the allocation of interneurons into specific cortical layers influences their function. This project is now possible due to the unique combination of our detailed know-how on the early development of cortical interneurons, including a variety of genetically modified mice available to us, and the application of new technologies to specifically target synchronically generated populations of interneurons. Our multidisciplinary approach, combining mouse genetics, in vivo functional genomics and electrophysiological methodologies represents a technological breakthrough that should accelerate our understanding of the general principles guiding the assembly of neuronal circuits in the cerebral cortex.
Summary
The neural assembly underlying the formation of functional networks in the cerebral cortex is conceivably the most complex biological system that exists. Much of this complexity arises during development through the interaction of dozens of different neuronal populations, which belong to two general classes: excitatory glutamatergic pyramidal cells and inhibitory gamma-aminobutyric containing (GABAergic) interneurons. Perhaps the most fascinating aspect of the assembly of cortical circuits is that pyramidal cells and interneurons are generated in distant germinal zones. Pyramidal cells are born locally from progenitors located in the cortical anlage, while interneurons derive from progenitors in the embryonic subpallium. Much progress has been made recently in understanding the molecular mechanisms that regulate the migration of interneurons towards the cortex, but how interneurons find their appropriate partners to build cortical networks with balanced excitation and inhibition remains an enigma.
The general goal of this project is to identify the mechanisms controlling the precise allocation of different classes of interneurons into specific layers of the cortex, where they assemble into neural circuits. We also aim to determine how the allocation of interneurons into specific cortical layers influences their function. This project is now possible due to the unique combination of our detailed know-how on the early development of cortical interneurons, including a variety of genetically modified mice available to us, and the application of new technologies to specifically target synchronically generated populations of interneurons. Our multidisciplinary approach, combining mouse genetics, in vivo functional genomics and electrophysiological methodologies represents a technological breakthrough that should accelerate our understanding of the general principles guiding the assembly of neuronal circuits in the cerebral cortex.
Max ERC Funding
2 493 481 €
Duration
Start date: 2012-04-01, End date: 2017-09-30
Project acronym COSMIC
Project Complex Synthetic Mimics of the Cell Membrane
Researcher (PI) Mark Ian Wallace
Host Institution (HI) KING'S COLLEGE LONDON
Call Details Starting Grant (StG), LS1, ERC-2012-StG_20111109
Summary I propose to bridge the gap between simple in vitro measurements of biological processes, and the complexities of the cellular environment. This requires reduced in vitro systems that are sufficiently complex to reproduce the subtleties of the in vivo biological phenomenon, but sufficiently controllable to test how quantitative changes in a particular property affects function. The challenge is to step beyond the most simple and straightforward in vitro mimics of the cell membrane, and create model systems that more closely reproduce the conditions in vivo.
I propose to tackle two specific, but interrelated membrane phenomena, that are currently not captured in artificial bilayers and create new complex mimics of the cell membrane capable of tackling these systems; namely (1) protein crowding and the cytoskeleton, and (2) lateral forces and membrane curvature. Testing our synthetic mimics with models that we understand in vivo is vital. This benchmarking will ensure that the mimics we create are relevant and will help ensure the more ambitious later goals of the this proposal are successful.We will then take these tools to go on and aim to create a synthetic mimic of the bacterial membrane.
However we are not limited to creating purely natural duplicates, and we can exploit a much wider range of building material than nature. In addition to creating complex mimics, we will also create totally new synthetic systems inspired by the properties of the cell membrane, but possessing unique properties.
Summary
I propose to bridge the gap between simple in vitro measurements of biological processes, and the complexities of the cellular environment. This requires reduced in vitro systems that are sufficiently complex to reproduce the subtleties of the in vivo biological phenomenon, but sufficiently controllable to test how quantitative changes in a particular property affects function. The challenge is to step beyond the most simple and straightforward in vitro mimics of the cell membrane, and create model systems that more closely reproduce the conditions in vivo.
I propose to tackle two specific, but interrelated membrane phenomena, that are currently not captured in artificial bilayers and create new complex mimics of the cell membrane capable of tackling these systems; namely (1) protein crowding and the cytoskeleton, and (2) lateral forces and membrane curvature. Testing our synthetic mimics with models that we understand in vivo is vital. This benchmarking will ensure that the mimics we create are relevant and will help ensure the more ambitious later goals of the this proposal are successful.We will then take these tools to go on and aim to create a synthetic mimic of the bacterial membrane.
However we are not limited to creating purely natural duplicates, and we can exploit a much wider range of building material than nature. In addition to creating complex mimics, we will also create totally new synthetic systems inspired by the properties of the cell membrane, but possessing unique properties.
Max ERC Funding
1 498 523 €
Duration
Start date: 2013-02-01, End date: 2018-10-31
Project acronym CoSuN
Project Cooperative Phenomena in Supramolecular Nanostructures
Researcher (PI) Harry Laurence Anderson
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Advanced Grant (AdG), PE5, ERC-2012-ADG_20120216
Summary Many of the remarkable properties of molecular nanostructures are cooperative effects. A system is described as cooperative when it behaves differently from expectations based on the properties of its individual components. Multivalent cooperativity is crucial for biological molecular recognition, yet the factors determining the magnitude of this effect are poorly understood. Excitonic cooperativity is exploited in sensitive detectors for explosives, and is the basis of photosynthetic light harvesting. Electronic cooperativity is illustrated on the molecular scale by the phenomenon of aromaticity, and on a larger scale by metallic conductivity. Magnetic properties provide many examples of cooperativity. The magnitude of cooperative effects increases with the strength of coupling between the individual components, and with the number of coupled components. Cooperative systems exhibit sharp changes in behavior in response to small changes in conditions, such as transitions from free to bound, fluorescent to non-fluorescent, or conductive to insulating. The tendency towards an “all-or-nothing” response is often useful; in the limit of a very large ensemble, it leads to phase transitions. The CoSuN project will extend methodology developed in Oxford to create large monodisperse supramolecular nanostructures which are uniquely suited for exploring multivalent, excitonic and electronic cooperativity. The template-directed synthesis of these nanostructures is made possible by strong multivalent cooperativity, while the electronic coupling between the individual subunits results in other cooperative phenomena. This project will clarify understanding of cooperative molecular recognition. It will also help to solve some of the mysteries of photosynthesis and reveal the first molecular manifestations of coherent quantum mechanical phenomena, such as Aharonov-Bohm effects.
Summary
Many of the remarkable properties of molecular nanostructures are cooperative effects. A system is described as cooperative when it behaves differently from expectations based on the properties of its individual components. Multivalent cooperativity is crucial for biological molecular recognition, yet the factors determining the magnitude of this effect are poorly understood. Excitonic cooperativity is exploited in sensitive detectors for explosives, and is the basis of photosynthetic light harvesting. Electronic cooperativity is illustrated on the molecular scale by the phenomenon of aromaticity, and on a larger scale by metallic conductivity. Magnetic properties provide many examples of cooperativity. The magnitude of cooperative effects increases with the strength of coupling between the individual components, and with the number of coupled components. Cooperative systems exhibit sharp changes in behavior in response to small changes in conditions, such as transitions from free to bound, fluorescent to non-fluorescent, or conductive to insulating. The tendency towards an “all-or-nothing” response is often useful; in the limit of a very large ensemble, it leads to phase transitions. The CoSuN project will extend methodology developed in Oxford to create large monodisperse supramolecular nanostructures which are uniquely suited for exploring multivalent, excitonic and electronic cooperativity. The template-directed synthesis of these nanostructures is made possible by strong multivalent cooperativity, while the electronic coupling between the individual subunits results in other cooperative phenomena. This project will clarify understanding of cooperative molecular recognition. It will also help to solve some of the mysteries of photosynthesis and reveal the first molecular manifestations of coherent quantum mechanical phenomena, such as Aharonov-Bohm effects.
Max ERC Funding
2 452 688 €
Duration
Start date: 2013-05-01, End date: 2018-04-30
Project acronym CRIPTON
Project Role of ncRNAs in Chromatin and Transcription
Researcher (PI) Tony Kouzarides
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Advanced Grant (AdG), LS1, ERC-2010-AdG_20100317
Summary The human genome is highly transcribed, with over 90% of sequences contributing to the production of RNA. The function of the vast majority of these RNAs is unknown. Evidence over many years has revealed that transcription factors and chromatin regulators are associated with a variety of non-coding (nc)RNAs, but their function remains largely unknown. There are a few cases where a role has been ascribed for ncRNAs in transcription, but no clear mechanistic insight has been defined yet. We predict that many of the newly identified ncRNAs emanating from the genome will play a role in transcriptional processes. We intend to identify and characterise such ncRNAs. This will take place in two phases. In the first phase we will use biochemical approaches to identify ncRNAs involved in the regulation of chromatin and transcription. Our investigations will focus on proteins leading to the induction of pluripotency and oncogenesis. ncRNAs associated with such proteins will be identified using targeted screens. In the second phase, the importance of these RNAs in determining pluripotency and oncogenesis will be analysed. In addition, a variety of molecular approaches will be used to investigate the mechanism by which these ncRNAs regulate the function of the proteins or complexes they associate with. One particular hypothesis we will explore is that such ncRNAs play a role in guiding proteins to DNA sequences, via the formation of RNA/DNA triplexes. This concerted and focused analysis will provide mechanistic insights into the functions of ncRNAs in transcriptional regulation and validate their role in key biological processes. The identification of such new ncRNA-regulated pathways may open up new avenues for therapeutic intervention.
Summary
The human genome is highly transcribed, with over 90% of sequences contributing to the production of RNA. The function of the vast majority of these RNAs is unknown. Evidence over many years has revealed that transcription factors and chromatin regulators are associated with a variety of non-coding (nc)RNAs, but their function remains largely unknown. There are a few cases where a role has been ascribed for ncRNAs in transcription, but no clear mechanistic insight has been defined yet. We predict that many of the newly identified ncRNAs emanating from the genome will play a role in transcriptional processes. We intend to identify and characterise such ncRNAs. This will take place in two phases. In the first phase we will use biochemical approaches to identify ncRNAs involved in the regulation of chromatin and transcription. Our investigations will focus on proteins leading to the induction of pluripotency and oncogenesis. ncRNAs associated with such proteins will be identified using targeted screens. In the second phase, the importance of these RNAs in determining pluripotency and oncogenesis will be analysed. In addition, a variety of molecular approaches will be used to investigate the mechanism by which these ncRNAs regulate the function of the proteins or complexes they associate with. One particular hypothesis we will explore is that such ncRNAs play a role in guiding proteins to DNA sequences, via the formation of RNA/DNA triplexes. This concerted and focused analysis will provide mechanistic insights into the functions of ncRNAs in transcriptional regulation and validate their role in key biological processes. The identification of such new ncRNA-regulated pathways may open up new avenues for therapeutic intervention.
Max ERC Funding
2 141 470 €
Duration
Start date: 2011-05-01, End date: 2017-04-30
Project acronym CRYOMAT
Project Antifreeze GlycoProtein Mimetic Polymers
Researcher (PI) Matthew Ian Gibson
Host Institution (HI) THE UNIVERSITY OF WARWICK
Call Details Starting Grant (StG), PE5, ERC-2014-STG
Summary Fish living in polar oceans have evolved an elegant, macromolecular, solution to survive in sub-zero water: they secrete antifreeze (glyco)proteins (AFGPs) which have several ‘antifreeze’ effects, including ice recrystallization inhibition (IRI) - they slow the rate of ice crystal growth. Ice crystal growth is a major problem in settings as diverse as oil fields, wind turbines, road surfaces and frozen food. Analysis of the process of cryopreservation, whereby donor cells are frozen for later use, has revealed that ice recrystallization is a major contributor to cell death upon thawing. Enhanced cryopreservation methods are particularly needed for stem cell storage to maximize the use of this currently limited resource, but also to enable storage of clinically transfused cells such as platelets and red blood cells. AFGPs have thus far not found application in cryopreservation due to their low availability from natural sources, extremely challenging synthesis, indications of cytotoxicity, but more importantly they have a side effect of shaping ice crystals into needle-shapes which pierces cells’ membranes, killing them. The aim of this ambitious project is to take a multidisciplinary approach to develop synthetic polymers as tunable, scalable and accessible bio-mimetics of AFGPs, which specifically reproduce only the desirable IRI properties. Precision synthetic and biological methods will be applied to access both vinyl- and peptide- based materials with IRI activity. The bio-inspired approach taken here will include detailed biophysical analysis of the polymer-ice interactions and translation of this understanding to real cryopreservation scenarios using blood-borne cells and human stem cells. In summary, this ambitious project takes inspiration from Nature's defense mechanisms that have evolved to allow life to flourish in extreme environments and will employ modern polymer chemistry to apply it to a real clinical problem; cryopreservation.
Summary
Fish living in polar oceans have evolved an elegant, macromolecular, solution to survive in sub-zero water: they secrete antifreeze (glyco)proteins (AFGPs) which have several ‘antifreeze’ effects, including ice recrystallization inhibition (IRI) - they slow the rate of ice crystal growth. Ice crystal growth is a major problem in settings as diverse as oil fields, wind turbines, road surfaces and frozen food. Analysis of the process of cryopreservation, whereby donor cells are frozen for later use, has revealed that ice recrystallization is a major contributor to cell death upon thawing. Enhanced cryopreservation methods are particularly needed for stem cell storage to maximize the use of this currently limited resource, but also to enable storage of clinically transfused cells such as platelets and red blood cells. AFGPs have thus far not found application in cryopreservation due to their low availability from natural sources, extremely challenging synthesis, indications of cytotoxicity, but more importantly they have a side effect of shaping ice crystals into needle-shapes which pierces cells’ membranes, killing them. The aim of this ambitious project is to take a multidisciplinary approach to develop synthetic polymers as tunable, scalable and accessible bio-mimetics of AFGPs, which specifically reproduce only the desirable IRI properties. Precision synthetic and biological methods will be applied to access both vinyl- and peptide- based materials with IRI activity. The bio-inspired approach taken here will include detailed biophysical analysis of the polymer-ice interactions and translation of this understanding to real cryopreservation scenarios using blood-borne cells and human stem cells. In summary, this ambitious project takes inspiration from Nature's defense mechanisms that have evolved to allow life to flourish in extreme environments and will employ modern polymer chemistry to apply it to a real clinical problem; cryopreservation.
Max ERC Funding
1 496 439 €
Duration
Start date: 2015-06-01, End date: 2020-05-31
Project acronym CRYOREP
Project Chromosome Replication Visualised by Cryo-EM
Researcher (PI) Alessandro COSTA
Host Institution (HI) THE FRANCIS CRICK INSTITUTE LIMITED
Call Details Consolidator Grant (CoG), LS1, ERC-2018-COG
Summary In eukaryotic cells, DNA replication is tightly regulated to ensure that the genome is duplicated only once per cell cycle. Errors in the control mechanisms that regulate chromosome ploidy cause genomic instability, which is linked to the development of cellular abnormalities, genetic disease and the onset of cancer. Recent reconstitution experiments performed with purified proteins revealed that initiation of eukaryotic genome duplication requires three distinct steps. First, DNA replication start sites are identified and targeted for the loading of an inactive MCM helicase motor, which encircles the double helix. Second, MCM activators are recruited, causing duplex-DNA untwisting. Third, upon interaction with a firing factor, the MCM ring opens to eject one DNA strand, leading to unwinding of the replication fork and duplication by dedicated replicative polymerases. These three events are not understood at a molecular level. Structural investigations so far aimed at imaging artificially isolated replication steps and used simplified templates, such as linear duplex DNA to study helicase loading or pre-formed forks to understand unwinding. However, the natural substrate of the eukaryotic replication machinery is not DNA but rather chromatin, formed of nucleosome arrays that compact the genome. Chromatin plays important regulatory roles in all steps of DNA replication, by dictating origin start-site selection and stimulating replication fork progression. Only by studying chromatin replication, we argue, will we understand the molecular basis of genome propagation. To this end, we have developed new protocols to perform visual biochemistry experiments under the cryo-electron microscope, to image chromatin duplication at high resolution, frozen as it is being catalysed. Using these strategies we want to generate a molecular movie of the entire replication reaction. Our achievements will change the way we think about genome stability in eukaryotic cells.
Summary
In eukaryotic cells, DNA replication is tightly regulated to ensure that the genome is duplicated only once per cell cycle. Errors in the control mechanisms that regulate chromosome ploidy cause genomic instability, which is linked to the development of cellular abnormalities, genetic disease and the onset of cancer. Recent reconstitution experiments performed with purified proteins revealed that initiation of eukaryotic genome duplication requires three distinct steps. First, DNA replication start sites are identified and targeted for the loading of an inactive MCM helicase motor, which encircles the double helix. Second, MCM activators are recruited, causing duplex-DNA untwisting. Third, upon interaction with a firing factor, the MCM ring opens to eject one DNA strand, leading to unwinding of the replication fork and duplication by dedicated replicative polymerases. These three events are not understood at a molecular level. Structural investigations so far aimed at imaging artificially isolated replication steps and used simplified templates, such as linear duplex DNA to study helicase loading or pre-formed forks to understand unwinding. However, the natural substrate of the eukaryotic replication machinery is not DNA but rather chromatin, formed of nucleosome arrays that compact the genome. Chromatin plays important regulatory roles in all steps of DNA replication, by dictating origin start-site selection and stimulating replication fork progression. Only by studying chromatin replication, we argue, will we understand the molecular basis of genome propagation. To this end, we have developed new protocols to perform visual biochemistry experiments under the cryo-electron microscope, to image chromatin duplication at high resolution, frozen as it is being catalysed. Using these strategies we want to generate a molecular movie of the entire replication reaction. Our achievements will change the way we think about genome stability in eukaryotic cells.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym CytoChem
Project A Chemical Approach to Understanding Cell Division
Researcher (PI) Ulrike Sophie Eggert
Host Institution (HI) KING'S COLLEGE LONDON
Call Details Starting Grant (StG), PE5, ERC-2012-StG_20111012
Summary Many mechanisms underlying cytokinesis, the final step in cell division, remain poorly understood. The goal of my laboratory is to use chemical biology approaches to address some of the unanswered mechanistic questions by studying cytokinesis at the process, pathway and protein levels. I aim to discover small molecules that specifically target cytokinesis by different mechanisms because they are important tools to study the biology of cell division and could catalyze the discovery of therapeutics.
I am proposing here to use small molecules we discovered to study how the Rho pathway regulates cytokinesis. We will synthesize focused libraries around selected compounds to optimize their properties and to identify sites for affinity tags. I am proposing to identify our small molecules’ cellular targets using a combination of approaches, including a new strategy I designed that takes advantage of the fact that they target a discrete signalling pathway.
Rho signalling is involved in every step of cytokinesis, but there are many outstanding questions about how this occurs and which proteins are involved. We have completed a genome-wide RNAi screen that has revealed the identity of new proteins connected to Rho signalling. We will combine functional investigations into how these proteins participate in cytokinesis with our newly discovered small molecules. With this array of tools in hand, we expect to use imaging and other cell-based assays to gain of comprehensive understanding of the role of Rho signalling during cytokinesis and other Rho-dependent processes.
Summary
Many mechanisms underlying cytokinesis, the final step in cell division, remain poorly understood. The goal of my laboratory is to use chemical biology approaches to address some of the unanswered mechanistic questions by studying cytokinesis at the process, pathway and protein levels. I aim to discover small molecules that specifically target cytokinesis by different mechanisms because they are important tools to study the biology of cell division and could catalyze the discovery of therapeutics.
I am proposing here to use small molecules we discovered to study how the Rho pathway regulates cytokinesis. We will synthesize focused libraries around selected compounds to optimize their properties and to identify sites for affinity tags. I am proposing to identify our small molecules’ cellular targets using a combination of approaches, including a new strategy I designed that takes advantage of the fact that they target a discrete signalling pathway.
Rho signalling is involved in every step of cytokinesis, but there are many outstanding questions about how this occurs and which proteins are involved. We have completed a genome-wide RNAi screen that has revealed the identity of new proteins connected to Rho signalling. We will combine functional investigations into how these proteins participate in cytokinesis with our newly discovered small molecules. With this array of tools in hand, we expect to use imaging and other cell-based assays to gain of comprehensive understanding of the role of Rho signalling during cytokinesis and other Rho-dependent processes.
Max ERC Funding
1 499 080 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym DDRREAM
Project DNA-Damage responses: Regulation and mechanisms
Researcher (PI) Stephen Philip Jackson
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Advanced Grant (AdG), LS1, ERC-2010-AdG_20100317
Summary The prime objective for every life form is to deliver its genetic material, intact, to the next generation. Each human cell receives tens-of-thousands of DNA lesions per day. These lesions can block genome replication and transcription, and if not repaired or repaired incorrectly, they lead to mutations or wider genome aberrations that threaten cell viability. To counter such threats, life has evolved the DNA-damage response (DDR), to detect DNA damage, signal its presence and mediate its repair. DDR events impact on many cellular processes and, crucially, prevent diverse human diseases that include cancer, neurodegenerative diseases, immune-deficiencies and premature ageing. While much progress has been made in identifying DDR proteins, much remains to be learned about the molecular and cellular functions that they control. Furthermore, the frequent reporting of new DDR proteins in the literature suggests that many others await identification. The main goals for the proposed research are to: identify important new DDR-proteins and DDR-modulators, particularly those responding to DNA double-strand breaks (DSBs); provide mechanistic insights into how these proteins function; and determine how DDR events are affected by chromatin structure, by molecular chaperones and components of the Ubiquitin and Sumo systems. To achieve these ends, we will use molecular biology, biochemical, cell-biology and molecular genetics approaches, including synthetic-lethal and phenotypic-suppression screening methods in human cells and in the nematode worm. This work will not only be of academic importance, but will also indicate how DDR dysfunction can cause human disease and how such diseases might be better diagnosed and treated.
Summary
The prime objective for every life form is to deliver its genetic material, intact, to the next generation. Each human cell receives tens-of-thousands of DNA lesions per day. These lesions can block genome replication and transcription, and if not repaired or repaired incorrectly, they lead to mutations or wider genome aberrations that threaten cell viability. To counter such threats, life has evolved the DNA-damage response (DDR), to detect DNA damage, signal its presence and mediate its repair. DDR events impact on many cellular processes and, crucially, prevent diverse human diseases that include cancer, neurodegenerative diseases, immune-deficiencies and premature ageing. While much progress has been made in identifying DDR proteins, much remains to be learned about the molecular and cellular functions that they control. Furthermore, the frequent reporting of new DDR proteins in the literature suggests that many others await identification. The main goals for the proposed research are to: identify important new DDR-proteins and DDR-modulators, particularly those responding to DNA double-strand breaks (DSBs); provide mechanistic insights into how these proteins function; and determine how DDR events are affected by chromatin structure, by molecular chaperones and components of the Ubiquitin and Sumo systems. To achieve these ends, we will use molecular biology, biochemical, cell-biology and molecular genetics approaches, including synthetic-lethal and phenotypic-suppression screening methods in human cells and in the nematode worm. This work will not only be of academic importance, but will also indicate how DDR dysfunction can cause human disease and how such diseases might be better diagnosed and treated.
Max ERC Funding
2 482 492 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym DEDIGROWTH
Project Dedicated growth of novel 1-dimensional materials for emerging nanotechnological applications
Researcher (PI) Nicole Grobert
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Starting Grant (StG), PE5, ERC-2009-StG
Summary This proposal aims to establish growth systematics for catalytically grown nanomaterials, such as nanoparticles, nanorods, carbon and hetero-atomic nanotubes. At present there is no clear understanding of the formation mechanism of these structures. Hence, the control over their properties, a vital aspect for technological applications of nanomaterials, is limited and remains difficult. Therefore, the main target of this proposal is the controlled production of new carbon and non-carbon-based nanomaterials with the focus on achieving structural control of the nanomaterials at the atomic level. An essential step towards the controlled generation of such new nanomaterials is a comprehensive understanding of the growth reactions and the role of the metal catalyst involved in the synthesis process. To achieve this, we will use in-situ techniques to study the chemical environment in the reactor during growth and state-of-the-art electron microscopy to reveal the chemical composition of the resulting catalyst particles and structures with atomic resolution. This data will provide information on how the nanostructure may have formed. Theoretical calculations and modelling of atomic scale processes of the catalyst reactivity will be used to draw a consistent picture of the functioning of the catalyst. An improved understanding of the functioning of the catalyst will allow us to estimate how the catalyst particles and reaction conditions have to be modified in order to enhance or to suppress certain products. A new high-throughput synthesis method together with the systematic variation of the growth parameters, such as cluster particle size and composition, temperature, gas pressure and precursor, will be used to generate a nanomaterials growth library. This nanomaterials library will be made available on the Internet for use by other researchers in planning their experiments.
Summary
This proposal aims to establish growth systematics for catalytically grown nanomaterials, such as nanoparticles, nanorods, carbon and hetero-atomic nanotubes. At present there is no clear understanding of the formation mechanism of these structures. Hence, the control over their properties, a vital aspect for technological applications of nanomaterials, is limited and remains difficult. Therefore, the main target of this proposal is the controlled production of new carbon and non-carbon-based nanomaterials with the focus on achieving structural control of the nanomaterials at the atomic level. An essential step towards the controlled generation of such new nanomaterials is a comprehensive understanding of the growth reactions and the role of the metal catalyst involved in the synthesis process. To achieve this, we will use in-situ techniques to study the chemical environment in the reactor during growth and state-of-the-art electron microscopy to reveal the chemical composition of the resulting catalyst particles and structures with atomic resolution. This data will provide information on how the nanostructure may have formed. Theoretical calculations and modelling of atomic scale processes of the catalyst reactivity will be used to draw a consistent picture of the functioning of the catalyst. An improved understanding of the functioning of the catalyst will allow us to estimate how the catalyst particles and reaction conditions have to be modified in order to enhance or to suppress certain products. A new high-throughput synthesis method together with the systematic variation of the growth parameters, such as cluster particle size and composition, temperature, gas pressure and precursor, will be used to generate a nanomaterials growth library. This nanomaterials library will be made available on the Internet for use by other researchers in planning their experiments.
Max ERC Funding
1 276 038 €
Duration
Start date: 2010-02-01, End date: 2016-01-31
Project acronym DEHALORES
Project Breathing chlorinated compounds: unravelling the biochemistry underpinning (de)halorespiration, an exciting bacterial metabolism with significant bioremediation potential
Researcher (PI) David Leys
Host Institution (HI) THE UNIVERSITY OF MANCHESTER
Call Details Starting Grant (StG), LS1, ERC-2007-StG
Summary Bacterial dehalorespiration is a microbial respiratory process in which halogenated hydrocarbons, from natural or anthropogenic origin, act as terminal electron acceptors. This leads to effective dehalogenation of these compounds, and as such their degradation and detoxification. The bacterial species, their enzymes and other components responsible for this unusual metabolism have only recently been identified. Unlocking the full potential of this process for bioremediation of persistent organohalides, such as polychlorinated biphenyls (PCBs) and tetrachloroethene, requires detailed understanding of the underpinning biochemistry. However, the regulation, mechanism and structure of the reductive dehalogenase (the enzyme responsible for delivering electrons to the halogenated substrates) are poorly understood. This ambitious proposal seeks to study representatives of the distinct reductive dehalogenase classes as well as key elements of the associated regulatory systems. Our group has been at the forefront of studying the biochemistry underpinning transcriptional regulation of dehalorespiration, providing detailed insights in the protein CprK at the atomic level. However, it is now apparent that only a subset of dehalogenases are regulated by CprK homologues with little known about the other regulators. In addition, studies on the reductive dehalogenases have been hampered by the inability to purify sufficient quantities. Using an interdisciplinary, biophysical approach focused around X-ray crystallography, enzymology and molecular biology, combined with novel reductive dehalogenase production methods, we aim to provide a detailed understanding and identification of the structural elements crucial to reductive dehalogenase mechanism and regulation. At the same time, we aim to apply the knowledge gathered and study the feasibility of generating improved dehalorespiratory components for biosensing or bioremediation applications through laboratory assisted evolution.
Summary
Bacterial dehalorespiration is a microbial respiratory process in which halogenated hydrocarbons, from natural or anthropogenic origin, act as terminal electron acceptors. This leads to effective dehalogenation of these compounds, and as such their degradation and detoxification. The bacterial species, their enzymes and other components responsible for this unusual metabolism have only recently been identified. Unlocking the full potential of this process for bioremediation of persistent organohalides, such as polychlorinated biphenyls (PCBs) and tetrachloroethene, requires detailed understanding of the underpinning biochemistry. However, the regulation, mechanism and structure of the reductive dehalogenase (the enzyme responsible for delivering electrons to the halogenated substrates) are poorly understood. This ambitious proposal seeks to study representatives of the distinct reductive dehalogenase classes as well as key elements of the associated regulatory systems. Our group has been at the forefront of studying the biochemistry underpinning transcriptional regulation of dehalorespiration, providing detailed insights in the protein CprK at the atomic level. However, it is now apparent that only a subset of dehalogenases are regulated by CprK homologues with little known about the other regulators. In addition, studies on the reductive dehalogenases have been hampered by the inability to purify sufficient quantities. Using an interdisciplinary, biophysical approach focused around X-ray crystallography, enzymology and molecular biology, combined with novel reductive dehalogenase production methods, we aim to provide a detailed understanding and identification of the structural elements crucial to reductive dehalogenase mechanism and regulation. At the same time, we aim to apply the knowledge gathered and study the feasibility of generating improved dehalorespiratory components for biosensing or bioremediation applications through laboratory assisted evolution.
Max ERC Funding
1 148 522 €
Duration
Start date: 2008-09-01, End date: 2013-08-31
Project acronym DENDRITE
Project Cellular and circuit determinants of dendritic computation
Researcher (PI) Michael Andreas Hausser
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Advanced Grant (AdG), LS5, ERC-2009-AdG
Summary What is the fundamental unit of computation in the brain? Answering this question is crucial not only for understanding how the brain works, but also for building accurate models of brain function, which require abstraction based on identification of the essential elements for carrying out computations relevant to behaviour. We will directly test the possibility that single dendritic branches may act as individual computational units during behaviour, challenging the classical view that the neuron is the fundamental unit of computation. We will address this question using a combination of electrophysiological, anatomical, imaging, molecular, and modeling approaches to probe dendritic integration in pyramidal cells and Purkinje cells in mouse cortex and cerebellum. We will define the computational rules for integration of synaptic input in dendrites by examining the responses to different spatiotemporal patterns of excitatory and inhibitory inputs. We will use computational modeling to extract simple rules describing dendritic integration that captures the essence of the computation. Next, we will determine how these rules are engaged by patterns of sensory stimulation in vivo, by using various strategies to map the spatiotemporal patterns of synaptic inputs to dendrites. To understand how physiological patterns of activity in the circuit engage these dendritic computations, we will use anatomical approaches to map the wiring diagram of synaptic inputs to individual dendrites. Finally, we will manipulate dendritic function using molecular tools, in order to provide causal links between specific dendritic computations and sensory processing. These experiments will provide us with deeper insights into how single neurons act as computing devices, and how fundamental computations that drive behaviour are implemented on the level of single cells and neural circuits.
Summary
What is the fundamental unit of computation in the brain? Answering this question is crucial not only for understanding how the brain works, but also for building accurate models of brain function, which require abstraction based on identification of the essential elements for carrying out computations relevant to behaviour. We will directly test the possibility that single dendritic branches may act as individual computational units during behaviour, challenging the classical view that the neuron is the fundamental unit of computation. We will address this question using a combination of electrophysiological, anatomical, imaging, molecular, and modeling approaches to probe dendritic integration in pyramidal cells and Purkinje cells in mouse cortex and cerebellum. We will define the computational rules for integration of synaptic input in dendrites by examining the responses to different spatiotemporal patterns of excitatory and inhibitory inputs. We will use computational modeling to extract simple rules describing dendritic integration that captures the essence of the computation. Next, we will determine how these rules are engaged by patterns of sensory stimulation in vivo, by using various strategies to map the spatiotemporal patterns of synaptic inputs to dendrites. To understand how physiological patterns of activity in the circuit engage these dendritic computations, we will use anatomical approaches to map the wiring diagram of synaptic inputs to individual dendrites. Finally, we will manipulate dendritic function using molecular tools, in order to provide causal links between specific dendritic computations and sensory processing. These experiments will provide us with deeper insights into how single neurons act as computing devices, and how fundamental computations that drive behaviour are implemented on the level of single cells and neural circuits.
Max ERC Funding
2 416 078 €
Duration
Start date: 2010-06-01, End date: 2016-05-31
Project acronym DENDRITECIRCUITS
Project The origins of dendritic computation within mammalian neural circuits
Researcher (PI) Michael HAUSSER
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Advanced Grant (AdG), LS5, ERC-2015-AdG
Summary This proposal aims to address a simple question: what is the fundamental unit of computation in the brain? Answering this question is crucial not only for understanding how the brain works, but also if we are to build accurate models of brain function, which require abstraction based on identification of the essential elements for carrying out computations relevant to behaviour. In this proposal, we will build on recent work demonstrating that dendrites are highly electrically excitable to test the possibility that single dendritic branches may act as individual computational units during behaviour, challenging the classical view that the neuron is the fundamental unit of computation. We will address this question using a combination of electrophysiolgical, anatomical, imaging, molecular, and modeling approaches to probe dendritic integration in pyramidal cells and Purkinje cells in mouse cortex and cerebellum.
We will first define the computational rules for integration of synaptic input in single and multiple dendrites by examining the somatic and dendritic responses to different spatiotemporal patterns of excitatory and inhibitory inputs in brain slices. Next, we will determine how these rules are engaged by patterns of sensory stimulation in vivo, by using various strategies to map the spatiotemporal patterns of synaptic inputs onto single dendrites. To understand how physiological patterns of activity in the circuit engage these dendritic computations, we will use anatomical approaches to map the wiring diagram of synaptic inputs to individual dendrites. Finally, we will perturb the dendritic computational rules by manipulating dendritic function using molecular and optogenetic tools, in order to provide causal links between specific dendritic computations and sensory processing relevant to behaviour.
These experiments will provide us with deeper insights into how single neurons act as computing devices.
Summary
This proposal aims to address a simple question: what is the fundamental unit of computation in the brain? Answering this question is crucial not only for understanding how the brain works, but also if we are to build accurate models of brain function, which require abstraction based on identification of the essential elements for carrying out computations relevant to behaviour. In this proposal, we will build on recent work demonstrating that dendrites are highly electrically excitable to test the possibility that single dendritic branches may act as individual computational units during behaviour, challenging the classical view that the neuron is the fundamental unit of computation. We will address this question using a combination of electrophysiolgical, anatomical, imaging, molecular, and modeling approaches to probe dendritic integration in pyramidal cells and Purkinje cells in mouse cortex and cerebellum.
We will first define the computational rules for integration of synaptic input in single and multiple dendrites by examining the somatic and dendritic responses to different spatiotemporal patterns of excitatory and inhibitory inputs in brain slices. Next, we will determine how these rules are engaged by patterns of sensory stimulation in vivo, by using various strategies to map the spatiotemporal patterns of synaptic inputs onto single dendrites. To understand how physiological patterns of activity in the circuit engage these dendritic computations, we will use anatomical approaches to map the wiring diagram of synaptic inputs to individual dendrites. Finally, we will perturb the dendritic computational rules by manipulating dendritic function using molecular and optogenetic tools, in order to provide causal links between specific dendritic computations and sensory processing relevant to behaviour.
These experiments will provide us with deeper insights into how single neurons act as computing devices.
Max ERC Funding
2 495 563 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym DEVINCI
Project Developmental principles for the functional specialisation of inhibitory circuits in neocortical areas
Researcher (PI) Oscar MARIN
Host Institution (HI) KING'S COLLEGE LONDON
Call Details Advanced Grant (AdG), LS5, ERC-2017-ADG
Summary The mammalian neocortex consists of discrete, but highly interconnected, functional areas that collectively encode features of the environment, form associations between stimuli and drive behaviour by transforming sensory input into motor output. All neocortical areas are organised into six layers containing two major classes of neurons, excitatory glutamatergic pyramidal cells and inhibitory GABAergic interneurons. However, each area has distinctive cytoarchitectonical features and inputs that largely determine its computational capabilities. As pyramidal cells comprise the large majority of neurons in the cerebral cortex, much emphasis has been made on their contribution to the differential organisation of cortical areas. In contrast, interneurons have received little attention in the context of the functional specialisation of cortical areas, even though their distribution is highly heterogeneous.
The central tenet of this research proposal is that distinct patterns of inhibitory connectivity may accompany, and perhaps even determine, the functional specialisation of neocortical areas. We hypothesise that interneurons play an important role in the tuning of circuits in each cortical area, and therefore that quantitative differences in the relative distribution of specific classes of interneurons, which arise during development, reflect functional specialisations. The overall aim of this research project is to understand how developmental mechanisms ‘sculpting’ the distribution of inhibitory neurons across different neocortical areas contribute to their functional specialisation. This project has the potential to transform our understanding of the organisation of inhibitory circuits in the mammalian neocortex.
Summary
The mammalian neocortex consists of discrete, but highly interconnected, functional areas that collectively encode features of the environment, form associations between stimuli and drive behaviour by transforming sensory input into motor output. All neocortical areas are organised into six layers containing two major classes of neurons, excitatory glutamatergic pyramidal cells and inhibitory GABAergic interneurons. However, each area has distinctive cytoarchitectonical features and inputs that largely determine its computational capabilities. As pyramidal cells comprise the large majority of neurons in the cerebral cortex, much emphasis has been made on their contribution to the differential organisation of cortical areas. In contrast, interneurons have received little attention in the context of the functional specialisation of cortical areas, even though their distribution is highly heterogeneous.
The central tenet of this research proposal is that distinct patterns of inhibitory connectivity may accompany, and perhaps even determine, the functional specialisation of neocortical areas. We hypothesise that interneurons play an important role in the tuning of circuits in each cortical area, and therefore that quantitative differences in the relative distribution of specific classes of interneurons, which arise during development, reflect functional specialisations. The overall aim of this research project is to understand how developmental mechanisms ‘sculpting’ the distribution of inhibitory neurons across different neocortical areas contribute to their functional specialisation. This project has the potential to transform our understanding of the organisation of inhibitory circuits in the mammalian neocortex.
Max ERC Funding
2 500 000 €
Duration
Start date: 2018-07-01, End date: 2023-06-30
Project acronym DEVSPACE
Project The development of the hippocampal spatial representation system
Researcher (PI) Francesca Cacucci
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Starting Grant (StG), LS5, ERC-2011-StG_20101109
Summary This proposal will address how a multimodal cognitive system, the neural representation of space in the hippocampus, emerges during development. There is a long tradition in neuroscience of studying the development of primary sensory systems, but fewer studies have concentrated on the development of brain networks supporting higher-order cognitive representations.
Our recent findings (Wills, Cacucci et al. Science, 2010) provide a starting point to fill this gap, charting the emergence of spatial responses of hippocampal formation neurons, using in vivo recording in awake, behaving rats.
The hippocampal formation supports neural representations of the environment ('cognitive maps') by means of which an animal can locate itself and navigate to a goal location. It contains three classes of spatially-tuned cells: place cells, which code for location, head direction cells, which code for directional orientation and grid cells, which may code for distance travelled.
The key aim of this proposal is to delineate the developmental processes that create this neural representation of space, focusing on the representations of place and direction.
We will delineate which sensory information is capable of driving spatial firing, and whether early hippocampal coding is truly spatial in the sense of representing configurations of stimuli and not single cues. How are abstract spatial constructs (place and head direction) built from raw sensory information during development? We will test whether boundary sensitive neurons and angular velocity tuned neurons are the elemental 'building blocks' making up place and directional signals, as suggested by many theoretical models.
We will also investigate the role of experience in the construction of spatial representations. Do the network architectures underlying spatial firing emerge through experience-dependent learning mechanisms, or are they the result of self-organizing processes which take place independently of experience?
Summary
This proposal will address how a multimodal cognitive system, the neural representation of space in the hippocampus, emerges during development. There is a long tradition in neuroscience of studying the development of primary sensory systems, but fewer studies have concentrated on the development of brain networks supporting higher-order cognitive representations.
Our recent findings (Wills, Cacucci et al. Science, 2010) provide a starting point to fill this gap, charting the emergence of spatial responses of hippocampal formation neurons, using in vivo recording in awake, behaving rats.
The hippocampal formation supports neural representations of the environment ('cognitive maps') by means of which an animal can locate itself and navigate to a goal location. It contains three classes of spatially-tuned cells: place cells, which code for location, head direction cells, which code for directional orientation and grid cells, which may code for distance travelled.
The key aim of this proposal is to delineate the developmental processes that create this neural representation of space, focusing on the representations of place and direction.
We will delineate which sensory information is capable of driving spatial firing, and whether early hippocampal coding is truly spatial in the sense of representing configurations of stimuli and not single cues. How are abstract spatial constructs (place and head direction) built from raw sensory information during development? We will test whether boundary sensitive neurons and angular velocity tuned neurons are the elemental 'building blocks' making up place and directional signals, as suggested by many theoretical models.
We will also investigate the role of experience in the construction of spatial representations. Do the network architectures underlying spatial firing emerge through experience-dependent learning mechanisms, or are they the result of self-organizing processes which take place independently of experience?
Max ERC Funding
1 491 930 €
Duration
Start date: 2012-01-01, End date: 2017-12-31
Project acronym DIREVOLFUN
Project Directed Evolution of Function within Chemical Systems: Adaptive Capsules and Polymers
Researcher (PI) Jonathan Russell Nitschke
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Starting Grant (StG), PE5, ERC-2010-StG_20091028
Summary A signature trait of living systems is their ability to dynamically adjust to features of their environments, adapting to stay alive, and evolving to take better advantage of the resources in their environments. This proposed research aims to synthesise new chemical systems that are capable of adaptation and evolution, with the achievement of specified functions being used as the benchmarks by which we may be judged to have succeeded in setting the direction for our systems evolution. Two parallel lines of inquiry will be followed. First, we will build upon results that we have recently published in Science[1] to create a series of new molecular capsules that are capable of dynamically adapting to different guest molecules. These capsules will serve as sensors and as enzyme-like catalysts through the use of transition-state-analogue guests. Second, we will prepare new metal-containing conjugated polymers through self-assembly, which will be capable of dynamically exchanging building blocks in solution. These polymers will have potential applications as electrically-conductive materials, with functional properties that may be tuned and optimised by the application of evolutionary pressures.
The success of these studies will thus create novel materials with uses as self-assembled sensors, catalysts, and electrical conductors. We will also shed light upon the question of how chemical systems may be induced to evolve under selective pressure. These studies thus have long-term bearing upon the questions of how living systems evolved from pre-biological mixtures of molecules.
[1] P. Mal, B. Breiner, K. Rissanen, J.R. Nitschke, Science 2009, 324, 1697-1699.
Summary
A signature trait of living systems is their ability to dynamically adjust to features of their environments, adapting to stay alive, and evolving to take better advantage of the resources in their environments. This proposed research aims to synthesise new chemical systems that are capable of adaptation and evolution, with the achievement of specified functions being used as the benchmarks by which we may be judged to have succeeded in setting the direction for our systems evolution. Two parallel lines of inquiry will be followed. First, we will build upon results that we have recently published in Science[1] to create a series of new molecular capsules that are capable of dynamically adapting to different guest molecules. These capsules will serve as sensors and as enzyme-like catalysts through the use of transition-state-analogue guests. Second, we will prepare new metal-containing conjugated polymers through self-assembly, which will be capable of dynamically exchanging building blocks in solution. These polymers will have potential applications as electrically-conductive materials, with functional properties that may be tuned and optimised by the application of evolutionary pressures.
The success of these studies will thus create novel materials with uses as self-assembled sensors, catalysts, and electrical conductors. We will also shed light upon the question of how chemical systems may be induced to evolve under selective pressure. These studies thus have long-term bearing upon the questions of how living systems evolved from pre-biological mixtures of molecules.
[1] P. Mal, B. Breiner, K. Rissanen, J.R. Nitschke, Science 2009, 324, 1697-1699.
Max ERC Funding
1 357 006 €
Duration
Start date: 2011-01-01, End date: 2016-12-31
Project acronym DiSCo MRI SFN
Project Developing Integrated Susceptibility and Conductivity MRI for Next Generation Structural and Functional Neuroimaging
Researcher (PI) Karin SHMUELI
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Consolidator Grant (CoG), LS5, ERC-2017-COG
Summary MRI is indispensable in the diagnosis of neurodegenerative diseases. These are poorly understood while their prevalence and socio-economic burden continue to rise. Structural and functional MRI can provide biomarkers for early diagnosis and potential therapeutic intervention. My research vision is to develop novel MRI methods for structural and functional mapping of tissue magnetic susceptibility and electrical conductivity as these show great promise for neuroimaging in diseases such as Alzheimer’s (AD).
Susceptibility mapping (SM), which I pioneered, is uniquely sensitive to tissue composition including iron content affected in AD while conductivity mapping (CM) probably reflects cellular disruption in AD. Resting-state functional MRI (rsfMRI) reveals how AD affects brain networks without any tasks or stimulation equipment. However, each technique currently needs a separate time-consuming MRI scan. I will develop an integrated scan for simultaneous structural SM and CM, and rsfMRI functional connectivity characterisation. This efficient scan, ideal for AD patients, will reveal totally new resting-state networks based on electromagnetic properties: resting-state functional SM and resting-state functional CM for the first time. As changes in blood susceptibility underlie fMRI, rsfSM should measure functional connectivity more directly. This also makes it sensitive to physiological noise so I will develop noise removal methods building on fMRI techniques I established. Initial fSM studies have been at 7 Tesla but I will target the more widespread 3T field to maximise applicability. As a leader in both SM and rsfMRI physiological noise removal I have the ideal background to integrate SM and CM with fMRI and extend them for ground-breaking functional electromagnetic connectivity. This research will yield a rich set of novel, multimodal MRI contrasts to allow development of new combined structural and functional biomarkers for early diagnosis of AD and other diseases.
Summary
MRI is indispensable in the diagnosis of neurodegenerative diseases. These are poorly understood while their prevalence and socio-economic burden continue to rise. Structural and functional MRI can provide biomarkers for early diagnosis and potential therapeutic intervention. My research vision is to develop novel MRI methods for structural and functional mapping of tissue magnetic susceptibility and electrical conductivity as these show great promise for neuroimaging in diseases such as Alzheimer’s (AD).
Susceptibility mapping (SM), which I pioneered, is uniquely sensitive to tissue composition including iron content affected in AD while conductivity mapping (CM) probably reflects cellular disruption in AD. Resting-state functional MRI (rsfMRI) reveals how AD affects brain networks without any tasks or stimulation equipment. However, each technique currently needs a separate time-consuming MRI scan. I will develop an integrated scan for simultaneous structural SM and CM, and rsfMRI functional connectivity characterisation. This efficient scan, ideal for AD patients, will reveal totally new resting-state networks based on electromagnetic properties: resting-state functional SM and resting-state functional CM for the first time. As changes in blood susceptibility underlie fMRI, rsfSM should measure functional connectivity more directly. This also makes it sensitive to physiological noise so I will develop noise removal methods building on fMRI techniques I established. Initial fSM studies have been at 7 Tesla but I will target the more widespread 3T field to maximise applicability. As a leader in both SM and rsfMRI physiological noise removal I have the ideal background to integrate SM and CM with fMRI and extend them for ground-breaking functional electromagnetic connectivity. This research will yield a rich set of novel, multimodal MRI contrasts to allow development of new combined structural and functional biomarkers for early diagnosis of AD and other diseases.
Max ERC Funding
1 721 726 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym DISCOVER
Project Design of Mixed Anion Inorganic Semiconductors for Energy Conversion
Researcher (PI) David SCANLON
Host Institution (HI) UNIVERSITY COLLEGE LONDON
Call Details Starting Grant (StG), PE5, ERC-2017-STG
Summary Multi-component systems offer the chemical and structural flexibility necessary to meet the needs of next-generation energy conversion. The vast majority of work in the field has focused on mixed-metal compounds. DISCOVER will computationally explore mixed-anion compounds. These are complex systems that provide significant technical challenges for atomistic and electronic structure modelling. Currently, structure-property relationships are poorly developed and there is a distinct lack of understanding of order-disorder transitions. Crucially, no systematic approach has been established for designing new combinations which can be tailored to match the criteria for technological applications.
This project aims to utilize advanced computational techniques to: (i) understand trends in existing mixed anion systems, and (ii) to employ state of the art crystal structure prediction codes to investigate novel ternary and quaternary mixed-anion compositions. The structure-property information emanating from this analysis will allow us to develop design principles for mixed anion semiconductors, which we will use to predict prototype systems for energy conversion. Promising candidates will be experimentally tested through a collaborative network of experts in the field. This ambitious project will push the boundaries of computational materials design, through the use of both classical and electronic structure simulation techniques for bulk, surface and excited states calculations.
The principle outcome will be a novel understanding of how to controllably design mixed anion semiconductors for technological applications, which will drive this material class to the forefront of materials science, while establishing my group at the frontier of computational materials science.
Summary
Multi-component systems offer the chemical and structural flexibility necessary to meet the needs of next-generation energy conversion. The vast majority of work in the field has focused on mixed-metal compounds. DISCOVER will computationally explore mixed-anion compounds. These are complex systems that provide significant technical challenges for atomistic and electronic structure modelling. Currently, structure-property relationships are poorly developed and there is a distinct lack of understanding of order-disorder transitions. Crucially, no systematic approach has been established for designing new combinations which can be tailored to match the criteria for technological applications.
This project aims to utilize advanced computational techniques to: (i) understand trends in existing mixed anion systems, and (ii) to employ state of the art crystal structure prediction codes to investigate novel ternary and quaternary mixed-anion compositions. The structure-property information emanating from this analysis will allow us to develop design principles for mixed anion semiconductors, which we will use to predict prototype systems for energy conversion. Promising candidates will be experimentally tested through a collaborative network of experts in the field. This ambitious project will push the boundaries of computational materials design, through the use of both classical and electronic structure simulation techniques for bulk, surface and excited states calculations.
The principle outcome will be a novel understanding of how to controllably design mixed anion semiconductors for technological applications, which will drive this material class to the forefront of materials science, while establishing my group at the frontier of computational materials science.
Max ERC Funding
1 499 998 €
Duration
Start date: 2018-02-01, End date: 2023-01-31
Project acronym DNA-REPAIR-CHROMATIN
Project Biochemical reconstitution of DNA repair reactions on physiological chromatin substrates
Researcher (PI) Matthew John Neale
Host Institution (HI) THE UNIVERSITY OF SUSSEX
Call Details Starting Grant (StG), LS1, ERC-2012-StG_20111109
Summary For cells and organisms to survive and propagate, they must accurately pass on their genetic information to the next generation. Errors in this process may arise from spontaneous mistakes in normal cellular metabolism, or from exposure to external agents, such as chemical mutagens and radiation. To protect themselves from the consequences of DNA damage, cells have evolved a vast array of pathways DNA repair mechanisms, each optimised for the resolution of a particular problem. One method of DNA repair, called homologous recombination (HR), involves using intact undamaged DNA sequences as a template to repair the damaged copy. HR is used extensively in meiotic cells to repair DNA breaks that are purposely created by the cell. In this context, HR is not just a repair mechanism, but also a method to drive interaction and genetic exchange between maternally and paternally inherited chromosomes, creating haploid genomes which are chimeras of the parental genetic information. Thus, the study of DNA repair and recombination informs our understanding of mechanisms that maintain genome stability, but which also generate genetic diversity, topics that are as critical to the survival of an individual cell as they are for the evolution and survival of an entire ecosystem. In recent decades a great deal has been learned of the genetic and biochemical control of the DNA repair and recombination mechanism. In general we infer gene function from what happens (or doesn’t happen) when we mutate a pathway of interest, and use biochemistry to test function using surrogate, simplified in vitro assays. Here, to bridge the divide between these classic approaches, I propose to develop biochemical methods using intact chromatin prepared from living cells. I believe that integrating chromatin biochemistry, with cell biology and genome-wide analysis will enable a new mode of scientific investigation, detailing how molecular reactions occur on biologically-relevant chromosomal substrates.
Summary
For cells and organisms to survive and propagate, they must accurately pass on their genetic information to the next generation. Errors in this process may arise from spontaneous mistakes in normal cellular metabolism, or from exposure to external agents, such as chemical mutagens and radiation. To protect themselves from the consequences of DNA damage, cells have evolved a vast array of pathways DNA repair mechanisms, each optimised for the resolution of a particular problem. One method of DNA repair, called homologous recombination (HR), involves using intact undamaged DNA sequences as a template to repair the damaged copy. HR is used extensively in meiotic cells to repair DNA breaks that are purposely created by the cell. In this context, HR is not just a repair mechanism, but also a method to drive interaction and genetic exchange between maternally and paternally inherited chromosomes, creating haploid genomes which are chimeras of the parental genetic information. Thus, the study of DNA repair and recombination informs our understanding of mechanisms that maintain genome stability, but which also generate genetic diversity, topics that are as critical to the survival of an individual cell as they are for the evolution and survival of an entire ecosystem. In recent decades a great deal has been learned of the genetic and biochemical control of the DNA repair and recombination mechanism. In general we infer gene function from what happens (or doesn’t happen) when we mutate a pathway of interest, and use biochemistry to test function using surrogate, simplified in vitro assays. Here, to bridge the divide between these classic approaches, I propose to develop biochemical methods using intact chromatin prepared from living cells. I believe that integrating chromatin biochemistry, with cell biology and genome-wide analysis will enable a new mode of scientific investigation, detailing how molecular reactions occur on biologically-relevant chromosomal substrates.
Max ERC Funding
1 747 823 €
Duration
Start date: 2013-01-01, End date: 2018-12-31
Project acronym DNA2REPAIR
Project DNA strand break repair and links to human disease
Researcher (PI) Stephen West
Host Institution (HI) THE FRANCIS CRICK INSTITUTE LIMITED
Call Details Advanced Grant (AdG), LS1, ERC-2014-ADG
Summary Our genetic material is continually subjected to damage, either from endogenous sources such as reactive oxygen species, produced as by-products of oxidative metabolism, from the breakdown of replication forks during cell growth, or by agents in the environment such as ionising radiation or carcinogenic chemicals. To cope with DNA damage, cells employ elaborate and effective repair processes that specifically recognise a wide variety of lesions in DNA. These repair systems are essential for the maintenance of genome integrity. Unfortunately, some individuals are genetically predisposed to crippling diseases or cancers that are the direct result of mutations in genes involved in the DNA damage response. For several years our work has been at the forefront of basic biological research in the area of DNA repair, and in particular we have made significant contributions to the understanding of inheritable diseases such as breast cancer, Fanconi anemia, and the neurodegenerative disorder Ataxia with Oculomotor Apraxia (AOA). The focus of this ERC proposal is: (i) to determine the mechanism of action and high-resolution structure of the BRCA2 tumour suppressor, and to provide a detailed picture of the interplay between BRCA2, PALB2, RAD51AP1 and the RAD51 paralogs, in terms of RAD51 filament assembly, using biochemical, electron microscopic and cell biological approaches, (ii) to determine the biological role of a unique structure-selective tri-nuclease complex (SLX1-SLX4-MUS81-EME1-XPF-ERCC1), with particular emphasis on its roles in DNA crosslink repair and Fanconi anemia, and (iii) to understand the actions of Senataxin, which is defective in AOA2, in protecting against genome instability in neuronal cells. These three distinct and yet inter-related areas of the research programme will provide an improved understanding of basic mechanisms of DNA repair and thereby underpin future therapeutic developments that will help individuals afflicted with these diseases.
Summary
Our genetic material is continually subjected to damage, either from endogenous sources such as reactive oxygen species, produced as by-products of oxidative metabolism, from the breakdown of replication forks during cell growth, or by agents in the environment such as ionising radiation or carcinogenic chemicals. To cope with DNA damage, cells employ elaborate and effective repair processes that specifically recognise a wide variety of lesions in DNA. These repair systems are essential for the maintenance of genome integrity. Unfortunately, some individuals are genetically predisposed to crippling diseases or cancers that are the direct result of mutations in genes involved in the DNA damage response. For several years our work has been at the forefront of basic biological research in the area of DNA repair, and in particular we have made significant contributions to the understanding of inheritable diseases such as breast cancer, Fanconi anemia, and the neurodegenerative disorder Ataxia with Oculomotor Apraxia (AOA). The focus of this ERC proposal is: (i) to determine the mechanism of action and high-resolution structure of the BRCA2 tumour suppressor, and to provide a detailed picture of the interplay between BRCA2, PALB2, RAD51AP1 and the RAD51 paralogs, in terms of RAD51 filament assembly, using biochemical, electron microscopic and cell biological approaches, (ii) to determine the biological role of a unique structure-selective tri-nuclease complex (SLX1-SLX4-MUS81-EME1-XPF-ERCC1), with particular emphasis on its roles in DNA crosslink repair and Fanconi anemia, and (iii) to understand the actions of Senataxin, which is defective in AOA2, in protecting against genome instability in neuronal cells. These three distinct and yet inter-related areas of the research programme will provide an improved understanding of basic mechanisms of DNA repair and thereby underpin future therapeutic developments that will help individuals afflicted with these diseases.
Max ERC Funding
2 203 153 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym DNAREPAIR
Project Defects in DNA strand break repair and links to inheritable disease
Researcher (PI) Stephen West
Host Institution (HI) THE FRANCIS CRICK INSTITUTE LIMITED
Call Details Advanced Grant (AdG), LS1, ERC-2009-AdG
Summary Our genetic material is continually subjected to damage, either from endogenous sources such as reactive oxygen species produced as by-products of oxidative metabolism, from the breakdown of replication forks during cell growth, or by agents in the environment such as ionising radiation or carcinogenic chemicals. To cope with DNA damage, cells employ elaborate and effective repair processes that specifically recognise a wide variety of lesions in DNA. These repair systems are essential for the maintenance of genome integrity. Unfortunately, some individuals are genetically predisposed to crippling diseases or cancers that are the direct result of mutations in genes involved in the DNA damage response. For several years our work has been at the forefront of basic biological research in the area of DNA repair, and in particular we have made significant contributions to the understanding of inheritable diseases such as breast cancer, Fanconi anemia, and the neurodegenerative disease Ataxia with Oculomotor Apraxia-1 (AOA-1). The focus of this ERC proposal is: (i) to define the phenotypic interplay between three inheritable cancer predisposition syndromes, Fanconi anemia, Bloom s syndrome and breast cancers caused by mutation of BRCA2, (ii) to determine the biological role of the newly discovered GEN1 Holliday junction resolvase in homologous recombination and repair, and (iii) to understand the actions of Aprataxin and Senataxin in relation to the inheritable neurodegenerative diseases AOA-1 and AOA-2, respectively. Our studies will provide an improved understanding of basic mechanisms of DNA repair and thereby underpin future therapeutic developments that will help individuals afflicted with these diseases.
Summary
Our genetic material is continually subjected to damage, either from endogenous sources such as reactive oxygen species produced as by-products of oxidative metabolism, from the breakdown of replication forks during cell growth, or by agents in the environment such as ionising radiation or carcinogenic chemicals. To cope with DNA damage, cells employ elaborate and effective repair processes that specifically recognise a wide variety of lesions in DNA. These repair systems are essential for the maintenance of genome integrity. Unfortunately, some individuals are genetically predisposed to crippling diseases or cancers that are the direct result of mutations in genes involved in the DNA damage response. For several years our work has been at the forefront of basic biological research in the area of DNA repair, and in particular we have made significant contributions to the understanding of inheritable diseases such as breast cancer, Fanconi anemia, and the neurodegenerative disease Ataxia with Oculomotor Apraxia-1 (AOA-1). The focus of this ERC proposal is: (i) to define the phenotypic interplay between three inheritable cancer predisposition syndromes, Fanconi anemia, Bloom s syndrome and breast cancers caused by mutation of BRCA2, (ii) to determine the biological role of the newly discovered GEN1 Holliday junction resolvase in homologous recombination and repair, and (iii) to understand the actions of Aprataxin and Senataxin in relation to the inheritable neurodegenerative diseases AOA-1 and AOA-2, respectively. Our studies will provide an improved understanding of basic mechanisms of DNA repair and thereby underpin future therapeutic developments that will help individuals afflicted with these diseases.
Max ERC Funding
2 449 091 €
Duration
Start date: 2010-06-01, End date: 2015-05-31
Project acronym DOS
Project Drugging the Undruggable: Discovery of Protein-Protein Interaction Modulators Using Diversity-Oriented Synthesis
Researcher (PI) David Spring
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Starting Grant (StG), PE5, ERC-2011-StG_20101014
Summary This proposal aims to exploit diversity-oriented synthesis in order to lay the scientific and technological foundations for the development of enzyme inhibition by protein-protein interaction (PPI) modulation as a tool for chemical biology and molecular therapeutics. We will deploy diversity-oriented synthesis lead discovery to explore concepts for PPI modulation in important enzyme families. This work will yield new chemical entities with a spectrum of properties directed against candidate macromolecular interactions important in the regulation of enzymes that mediate key biological pathways. The proposed work has the potential to transform current approaches to drug discovery, and to radically extend the repertoire of tools available for chemical biology. It will help to address the problem of identifying small-molecule inhibitors of PPIs, widely accepted to be of major fundamental and practical significance to biomedical science.
Summary
This proposal aims to exploit diversity-oriented synthesis in order to lay the scientific and technological foundations for the development of enzyme inhibition by protein-protein interaction (PPI) modulation as a tool for chemical biology and molecular therapeutics. We will deploy diversity-oriented synthesis lead discovery to explore concepts for PPI modulation in important enzyme families. This work will yield new chemical entities with a spectrum of properties directed against candidate macromolecular interactions important in the regulation of enzymes that mediate key biological pathways. The proposed work has the potential to transform current approaches to drug discovery, and to radically extend the repertoire of tools available for chemical biology. It will help to address the problem of identifying small-molecule inhibitors of PPIs, widely accepted to be of major fundamental and practical significance to biomedical science.
Max ERC Funding
1 499 723 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym DUPLEX
Project Programmable Plastics
Researcher (PI) Christopher Alexander Hunter
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Advanced Grant (AdG), PE5, ERC-2012-ADG_20120216
Summary The unique properties of nucleic acids have made them the material of choice for complex nanofabrication. High fidelity formation of duplexes via non-covalent interactions between complementary sequences provides a straightforward approach to molecular programming of multicomponent self-assembly processes. The structure of the nucleic acid backbone and bases can be changed without destroying these properties, suggesting that there are all kinds of unexplored polymeric structures that will also show sequence selective duplex formation. This proposal investigates this rich new area at the interface of supramolecular, biological and polymer chemistry. The appeal of nucleic acids is that we can dial up any desired sequence via chemical solid phase synthesis or via biological template synthesis. With recent advances in polymerisation processes, which proceed under mild conditions compatible with non-covalent chemistry, we are now in a position to develop comparable processes for synthetic polymers. This proposal explores a ground-breaking approach to the synthesis of polymeric systems equipped with defined sequences of recognition sites. The aim is to establish protocols for routine solid phase synthesis of one class of oligomer, which can be used to template the synthesis of different classes of oligomer. This template chemistry will provide tools for polymerisation of conventional monomers using templates to determine the sequence of recognition sites and hence incorporate the selective recognition properties of nucleic acids into bulk polymers like polystyrene. The ability to program polymers with recognition information will open the way to new materials of unprecedented complexity and functionality with applications in all areas of nanotechnology where precise control over macromolecular structure and supramolecular organisation will be used to program mechanical, photochemical and electronic properties into sophisticated assemblies that rival biology.
Summary
The unique properties of nucleic acids have made them the material of choice for complex nanofabrication. High fidelity formation of duplexes via non-covalent interactions between complementary sequences provides a straightforward approach to molecular programming of multicomponent self-assembly processes. The structure of the nucleic acid backbone and bases can be changed without destroying these properties, suggesting that there are all kinds of unexplored polymeric structures that will also show sequence selective duplex formation. This proposal investigates this rich new area at the interface of supramolecular, biological and polymer chemistry. The appeal of nucleic acids is that we can dial up any desired sequence via chemical solid phase synthesis or via biological template synthesis. With recent advances in polymerisation processes, which proceed under mild conditions compatible with non-covalent chemistry, we are now in a position to develop comparable processes for synthetic polymers. This proposal explores a ground-breaking approach to the synthesis of polymeric systems equipped with defined sequences of recognition sites. The aim is to establish protocols for routine solid phase synthesis of one class of oligomer, which can be used to template the synthesis of different classes of oligomer. This template chemistry will provide tools for polymerisation of conventional monomers using templates to determine the sequence of recognition sites and hence incorporate the selective recognition properties of nucleic acids into bulk polymers like polystyrene. The ability to program polymers with recognition information will open the way to new materials of unprecedented complexity and functionality with applications in all areas of nanotechnology where precise control over macromolecular structure and supramolecular organisation will be used to program mechanical, photochemical and electronic properties into sophisticated assemblies that rival biology.
Max ERC Funding
2 457 947 €
Duration
Start date: 2013-03-01, End date: 2019-02-28
Project acronym DYNAFLUORS
Project Dynamic Activatable Fluorophores
Researcher (PI) Marc VENDRELL ESCOBAR
Host Institution (HI) THE UNIVERSITY OF EDINBURGH
Call Details Consolidator Grant (CoG), PE5, ERC-2017-COG
Summary In DYNAFLUORS I will develop the first chemical toolbox for imaging in real time the activity of immune cells in tumours.
Although the management of cancer has improved over the years, the cure rates for patients with metastasis and advanced tumours remain low due to lack of appropriate therapies. Recent studies suggest that drugs empowering host immune cells (i.e. immunotherapies) are promising approaches for intractable tumours. However, there are no tools to visualise and understand how host immune cells stop cancer progression in vivo. This important unmet challenge drives the ambitious targets of this proposal.
Over the past 10 years, I have pioneered the development of chemical fluorophores that allow unparalleled analysis of biological systems. In this project, I will implement an innovative approach to unify cutting-edge methodologies in chemistry and biology and develop Dynamic Activatable Fluorophores (DYNAFLUORS) as a chemical toolbox with enhanced imaging capabilities over current technologies.
The cross-disciplinary and ambitious nature of this project will open multiple avenues for broad impact in many areas of chemistry as well as in basic biology, imaging and medicine. DYNAFLUORS will allow us to image, from the molecular level to human tissue, the activity of immune cells in tumours and the response to therapy in real time. This ground-breaking chemical platform will represent a step forward in the forefront of chemical imaging and will create new opportunities in the personalised management of cancer.
In the long term, DYNAFLUORS will become a transformative toolbox for monitoring disease in humans. The integration of functional fluorophores into imaging technologies to perform ‘optical biopsies’ in vivo and to create patient-specific drug-response assays has the potential to revolutionise the diagnosis, stratification and personalised treatment of disease.
Summary
In DYNAFLUORS I will develop the first chemical toolbox for imaging in real time the activity of immune cells in tumours.
Although the management of cancer has improved over the years, the cure rates for patients with metastasis and advanced tumours remain low due to lack of appropriate therapies. Recent studies suggest that drugs empowering host immune cells (i.e. immunotherapies) are promising approaches for intractable tumours. However, there are no tools to visualise and understand how host immune cells stop cancer progression in vivo. This important unmet challenge drives the ambitious targets of this proposal.
Over the past 10 years, I have pioneered the development of chemical fluorophores that allow unparalleled analysis of biological systems. In this project, I will implement an innovative approach to unify cutting-edge methodologies in chemistry and biology and develop Dynamic Activatable Fluorophores (DYNAFLUORS) as a chemical toolbox with enhanced imaging capabilities over current technologies.
The cross-disciplinary and ambitious nature of this project will open multiple avenues for broad impact in many areas of chemistry as well as in basic biology, imaging and medicine. DYNAFLUORS will allow us to image, from the molecular level to human tissue, the activity of immune cells in tumours and the response to therapy in real time. This ground-breaking chemical platform will represent a step forward in the forefront of chemical imaging and will create new opportunities in the personalised management of cancer.
In the long term, DYNAFLUORS will become a transformative toolbox for monitoring disease in humans. The integration of functional fluorophores into imaging technologies to perform ‘optical biopsies’ in vivo and to create patient-specific drug-response assays has the potential to revolutionise the diagnosis, stratification and personalised treatment of disease.
Max ERC Funding
1 986 650 €
Duration
Start date: 2018-06-01, End date: 2023-05-31
Project acronym DYNAMIN
Project Dynamic Control of Mineralisation
Researcher (PI) Fiona Meldrum
Host Institution (HI) UNIVERSITY OF LEEDS
Call Details Advanced Grant (AdG), PE5, ERC-2017-ADG
Summary This project will take inspiration from biomineralisation to achieve exceptional, dynamic control over crystallisation processes.
Understanding the fundamental mechanisms which govern crystallisation promises the ability to inhibit or promote crystallisation as desired, and to tailor the properties of crystalline materials towards a huge range of applications. Biomineralisation provides a perfect precedent for this approach, where organisms achieve control currently unparalleled in synthetic systems. This is achieved because mineralisation occurs within controlled environments in which an organism can interact with the nascent mineral.
Thanks to recent advances in microfabrication techniques and analytical methods we finally have the tools required to bring such control to the laboratory. DYNAMIN will exploit microfluidic and confined systems to study and interact with crystallisation processes with outstanding spatial and temporal resolution. Flowing droplet devices will be coupled to synchrotron techniques to investigate and control nucleation, using soluble additives and nucleating particles to direct the crystallisation pathway. Static chambers will be used to interact with crystallisation processes over longer length and time scales to achieve spatio-temporal control to rival that in biomineralisation, while a unique confined system – titania nanotubes – will enable the study and control of organic-mediated mineralisation, using fresh reagents and proteinases to interact with the process. Finally, a key biogenic strategy will provide the inspiration to develop a simple and potentially general method to trigger and control the transformation of amorphous precursor phases to single crystal products.
This will generate a new framework for studying and controlling crystallisation processes, where these new skills will find applications in sectors ranging from the Chemical Industry, to Healthcare, Advanced Materials, Formulated Products and the Environment.
Summary
This project will take inspiration from biomineralisation to achieve exceptional, dynamic control over crystallisation processes.
Understanding the fundamental mechanisms which govern crystallisation promises the ability to inhibit or promote crystallisation as desired, and to tailor the properties of crystalline materials towards a huge range of applications. Biomineralisation provides a perfect precedent for this approach, where organisms achieve control currently unparalleled in synthetic systems. This is achieved because mineralisation occurs within controlled environments in which an organism can interact with the nascent mineral.
Thanks to recent advances in microfabrication techniques and analytical methods we finally have the tools required to bring such control to the laboratory. DYNAMIN will exploit microfluidic and confined systems to study and interact with crystallisation processes with outstanding spatial and temporal resolution. Flowing droplet devices will be coupled to synchrotron techniques to investigate and control nucleation, using soluble additives and nucleating particles to direct the crystallisation pathway. Static chambers will be used to interact with crystallisation processes over longer length and time scales to achieve spatio-temporal control to rival that in biomineralisation, while a unique confined system – titania nanotubes – will enable the study and control of organic-mediated mineralisation, using fresh reagents and proteinases to interact with the process. Finally, a key biogenic strategy will provide the inspiration to develop a simple and potentially general method to trigger and control the transformation of amorphous precursor phases to single crystal products.
This will generate a new framework for studying and controlling crystallisation processes, where these new skills will find applications in sectors ranging from the Chemical Industry, to Healthcare, Advanced Materials, Formulated Products and the Environment.
Max ERC Funding
2 632 375 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym DYNAMITO
Project The analysis of mitochondrial dynamics in ageing and neurodegeneration
Researcher (PI) Alexander James Whitworth
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Starting Grant (StG), LS5, ERC-2012-StG_20111109
Summary The survival of our most active tissues, such as the brain and heart, throughout decades of a human lifespan presents an extraordinary biological challenge. Mitochondria are central to the life and death of these tissues. They provide the cellular energy required by these cells and protect them by buffering potentially lethal levels of cytoplasmic calcium, while at the same time mitochondria produce much of the molecules that cause cellular damage and contain a lethal arsenal of apoptotic cell death machinery. These organelles require exquisite maintenance processes to keep them intact and prevent potentially catastrophic disruption. Failure in mitochondrial homeostasis is strongly linked to age-related conditions such as neurodegeneration.
This subject has garnered intense interest recently with emergence that two genes linked to Parkinson's disease, PINK1 and parkin, regulate the autophagic degradation of mitochondria (mitophagy). Mitophagy is coordinated with mitochondrial dynamics, processes vital to neuronal health. While recent work has uncovered the basic mechanisms of PINK1/parkin-induced mitophagy, many questions and caveats surround the current knowledge. Most notably, all studies to date have used in vitro approaches and non-physiological manipulations. Thus, we still have a poor understanding of this process in a physiological context.
I will principally use the powerful genetic techniques of Drosophila to investigate the influence of mitochondrial dynamics on maintaining normal neuronal function and survival, and its impact on neurodegeneration, in the context of an intact animal system. I will also use molecular, cell biology and biochemical approaches in mammalian cells to complement our in vivo findings and verify their relevance to human biology. These insights will deliver a greater understanding of the role of mitochondrial dynamics in the health and dysfunction of the nervous system in a physiological context and guide therapeutic developments.
Summary
The survival of our most active tissues, such as the brain and heart, throughout decades of a human lifespan presents an extraordinary biological challenge. Mitochondria are central to the life and death of these tissues. They provide the cellular energy required by these cells and protect them by buffering potentially lethal levels of cytoplasmic calcium, while at the same time mitochondria produce much of the molecules that cause cellular damage and contain a lethal arsenal of apoptotic cell death machinery. These organelles require exquisite maintenance processes to keep them intact and prevent potentially catastrophic disruption. Failure in mitochondrial homeostasis is strongly linked to age-related conditions such as neurodegeneration.
This subject has garnered intense interest recently with emergence that two genes linked to Parkinson's disease, PINK1 and parkin, regulate the autophagic degradation of mitochondria (mitophagy). Mitophagy is coordinated with mitochondrial dynamics, processes vital to neuronal health. While recent work has uncovered the basic mechanisms of PINK1/parkin-induced mitophagy, many questions and caveats surround the current knowledge. Most notably, all studies to date have used in vitro approaches and non-physiological manipulations. Thus, we still have a poor understanding of this process in a physiological context.
I will principally use the powerful genetic techniques of Drosophila to investigate the influence of mitochondrial dynamics on maintaining normal neuronal function and survival, and its impact on neurodegeneration, in the context of an intact animal system. I will also use molecular, cell biology and biochemical approaches in mammalian cells to complement our in vivo findings and verify their relevance to human biology. These insights will deliver a greater understanding of the role of mitochondrial dynamics in the health and dysfunction of the nervous system in a physiological context and guide therapeutic developments.
Max ERC Funding
1 486 761 €
Duration
Start date: 2013-01-01, End date: 2018-12-31
Project acronym DYNAPORE
Project Dynamic responsive porous crystals
Researcher (PI) Matthew ROSSEINSKY
Host Institution (HI) THE UNIVERSITY OF LIVERPOOL
Call Details Advanced Grant (AdG), PE5, ERC-2015-AdG
Summary The project addresses the long-term vision of man-made materials with chemical selectivity and functional efficiency produced by dynamic structural flexibility. These materials are not intended as protein mimics; they are however inspired by nature’s use of flexible rather than rigid systems, with their ability to dynamically restructure around guests and thus perform highly specific chemistry. Such materials would transform chemical processes through their precision, for example by reorganising to accelerate each step of a cascade reaction without reagent or product inhibition. The road to this vision is blocked as we do not have the methodology and understanding to control such materials.
The aim is to develop synergic, multidisciplinary experimental and computational capability to harness the dynamics of flexible crystalline porous solids for function, demonstrated in separation and catalysis. This will enable design and synthesis of materials that controllably adopt distinct structures according to their chemical environment to optimise performance. We will create a new workflow that integrates understanding of the structure-composition-dynamics-property relationship into the materials design and discovery process. This workflow builds on proof-of-concept in (i) chemical control of dynamical restructuring in flexible crystalline porous materials and in the use of dynamics to (ii) enhance function and (iii) guide synthesis.
Crystalline flexible porous materials are selected because crystallinity maximises the atomic-scale understanding generated, which is transferable to other materials classes, whilst porosity permits sorption and organisation of guests that controls function.
This inorganic materials chemistry project develops integrated capability in chemical synthesis (new metal-organic frameworks and linkers), computation (prediction and evaluation of structure and dynamical guest response), characterisation (e.g. by diffraction) and measurement of function.
Summary
The project addresses the long-term vision of man-made materials with chemical selectivity and functional efficiency produced by dynamic structural flexibility. These materials are not intended as protein mimics; they are however inspired by nature’s use of flexible rather than rigid systems, with their ability to dynamically restructure around guests and thus perform highly specific chemistry. Such materials would transform chemical processes through their precision, for example by reorganising to accelerate each step of a cascade reaction without reagent or product inhibition. The road to this vision is blocked as we do not have the methodology and understanding to control such materials.
The aim is to develop synergic, multidisciplinary experimental and computational capability to harness the dynamics of flexible crystalline porous solids for function, demonstrated in separation and catalysis. This will enable design and synthesis of materials that controllably adopt distinct structures according to their chemical environment to optimise performance. We will create a new workflow that integrates understanding of the structure-composition-dynamics-property relationship into the materials design and discovery process. This workflow builds on proof-of-concept in (i) chemical control of dynamical restructuring in flexible crystalline porous materials and in the use of dynamics to (ii) enhance function and (iii) guide synthesis.
Crystalline flexible porous materials are selected because crystallinity maximises the atomic-scale understanding generated, which is transferable to other materials classes, whilst porosity permits sorption and organisation of guests that controls function.
This inorganic materials chemistry project develops integrated capability in chemical synthesis (new metal-organic frameworks and linkers), computation (prediction and evaluation of structure and dynamical guest response), characterisation (e.g. by diffraction) and measurement of function.
Max ERC Funding
2 493 425 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym ECMneuro
Project Perineuronal net treatments for neurodegenerative disease
Researcher (PI) James Fawcett
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Advanced Grant (AdG), LS5, ERC-2011-ADG_20110310
Summary Inhibitory chondroitin sulphate proteoglycans (CSPGs) have several roles in CNS damage and repair, revealed by their digestion with chondroitinase. Most recently, digestion of CSPGs in the limbic system and cortex has led to a very substantial enhancement of memory.
The effects of CSPGs on plasticity and memory are largely through their concentration into PNNs, because transgenics lacking the PNN component link protein in the CNS have very attenuated PNNs, and show continuing plasticity into adulthood, and enhanced memory in just the same way as chondroitinase-treated animals. The PNN is therefore a novel therapeutic target that has not been explored.
This application focuses particularly on enhancement of memory through manipulation of PNNs.
In Alzheimer’s disease (AD) and ageing the main cognitive disability is loss of memory. The enhancement of memory following chondroitinase treatment or PNN knockout in object memory is many times greater than obtained using cholinesterase inhibitors (the only currently available treatment for memory enhancement). PNN manipulation is therefore a particularly promising avenue for developing treatments to overcome the main cognitive disability of AD and ageing.
The aims of the application are:
1. Test the extent of memory enhancement due to PNN manipulation in models of AD due to Abeta, tau mutations and in aged CNS.
2. Establish the molecular mechanism for PNN effects on memory, focusing on Semaphorin3 presentation by PNNs, and direct effects via the PTPsigma receptor.
3. Discover the sulphation modifications of the CSPG glycan chains that enable binding of Semaphorin3s, activation of the PTPsigma receptor.
4. Analyse molecules that bind to PNN glycans, to identify new potential effectors of PNN effects on memory and plasticity
5. Testing in memory and plasticity models of novel PNN-targeted approaches.
Summary
Inhibitory chondroitin sulphate proteoglycans (CSPGs) have several roles in CNS damage and repair, revealed by their digestion with chondroitinase. Most recently, digestion of CSPGs in the limbic system and cortex has led to a very substantial enhancement of memory.
The effects of CSPGs on plasticity and memory are largely through their concentration into PNNs, because transgenics lacking the PNN component link protein in the CNS have very attenuated PNNs, and show continuing plasticity into adulthood, and enhanced memory in just the same way as chondroitinase-treated animals. The PNN is therefore a novel therapeutic target that has not been explored.
This application focuses particularly on enhancement of memory through manipulation of PNNs.
In Alzheimer’s disease (AD) and ageing the main cognitive disability is loss of memory. The enhancement of memory following chondroitinase treatment or PNN knockout in object memory is many times greater than obtained using cholinesterase inhibitors (the only currently available treatment for memory enhancement). PNN manipulation is therefore a particularly promising avenue for developing treatments to overcome the main cognitive disability of AD and ageing.
The aims of the application are:
1. Test the extent of memory enhancement due to PNN manipulation in models of AD due to Abeta, tau mutations and in aged CNS.
2. Establish the molecular mechanism for PNN effects on memory, focusing on Semaphorin3 presentation by PNNs, and direct effects via the PTPsigma receptor.
3. Discover the sulphation modifications of the CSPG glycan chains that enable binding of Semaphorin3s, activation of the PTPsigma receptor.
4. Analyse molecules that bind to PNN glycans, to identify new potential effectors of PNN effects on memory and plasticity
5. Testing in memory and plasticity models of novel PNN-targeted approaches.
Max ERC Funding
2 450 543 €
Duration
Start date: 2012-03-01, End date: 2017-02-28
Project acronym ECO-ZEN
Project Enabling Catalytic Cross Couplings with only Zinc Electrophiles, Nucleophiles and Boranes
Researcher (PI) Michael James INGLESON
Host Institution (HI) THE UNIVERSITY OF MANCHESTER
Call Details Consolidator Grant (CoG), PE5, ERC-2017-COG
Summary This high-impact, challenging CoG Proposal integrates multiple novel ideas in boron and zinc chemistry into an overarching project to open up new horizons across synthesis and catalysis. The Applicant’s successful ERC StG has opened up new avenues of pioneering research in main group element mediated transformations that were not conceivable before the work was done. Components of this proposal extend out from the StG into new, exciting research areas that are completely different. Developing low toxicity earth abundant catalysts for important transformations is vital to the EU with the focus herein being on; (i) the Suzuki-Miyaura (S-M) cross coupling reaction which is ubiquitous in industry and academia, and (ii) the formation of organoboranes that are essential synthetic intermediates. Both of these are currently dominated by toxic, expensive and low abundance precious metal catalysts (e.g. Pd, Ir). This project will deliver innovation through utilising combinations of main group Lewis acids and nucleophilic anions that do not react with each other, i.e. are frustrated pairs. This “frustration” enables the two species to concertedly transform substrates to achieve:
(i) Precious metal-free S-M cross coupling reactions of sp3C electrophiles catalysed by zinc and boron compounds, including stereospecific couplings and one pot two step cross electrophile couplings.
(ii) Trans-elementoboration of alkynes, including the unprecedented fluoroboration of alkynes.
Other new approaches will be developed to access novel (hetero)arylboronic acid derivatives using only simple boranes and without requiring noble metal catalysts, specifically: (i) boron directed C-H borylation and (ii) directed ortho borylation to enable subsequent meta selective SEAr C-H functionalisation.
This CoG will afford the freedom and impetus via consolidated funding to undertake fundamental research to deliver high impact results, including developing a new area of cross coupling catalysis research.
Summary
This high-impact, challenging CoG Proposal integrates multiple novel ideas in boron and zinc chemistry into an overarching project to open up new horizons across synthesis and catalysis. The Applicant’s successful ERC StG has opened up new avenues of pioneering research in main group element mediated transformations that were not conceivable before the work was done. Components of this proposal extend out from the StG into new, exciting research areas that are completely different. Developing low toxicity earth abundant catalysts for important transformations is vital to the EU with the focus herein being on; (i) the Suzuki-Miyaura (S-M) cross coupling reaction which is ubiquitous in industry and academia, and (ii) the formation of organoboranes that are essential synthetic intermediates. Both of these are currently dominated by toxic, expensive and low abundance precious metal catalysts (e.g. Pd, Ir). This project will deliver innovation through utilising combinations of main group Lewis acids and nucleophilic anions that do not react with each other, i.e. are frustrated pairs. This “frustration” enables the two species to concertedly transform substrates to achieve:
(i) Precious metal-free S-M cross coupling reactions of sp3C electrophiles catalysed by zinc and boron compounds, including stereospecific couplings and one pot two step cross electrophile couplings.
(ii) Trans-elementoboration of alkynes, including the unprecedented fluoroboration of alkynes.
Other new approaches will be developed to access novel (hetero)arylboronic acid derivatives using only simple boranes and without requiring noble metal catalysts, specifically: (i) boron directed C-H borylation and (ii) directed ortho borylation to enable subsequent meta selective SEAr C-H functionalisation.
This CoG will afford the freedom and impetus via consolidated funding to undertake fundamental research to deliver high impact results, including developing a new area of cross coupling catalysis research.
Max ERC Funding
2 070 093 €
Duration
Start date: 2018-05-01, End date: 2023-04-30
Project acronym EMERGE
Project Enzyme Driven Molecular Nanosystems
Researcher (PI) Rein V Ulijn
Host Institution (HI) UNIVERSITY OF STRATHCLYDE
Call Details Starting Grant (StG), PE5, ERC-2010-StG_20091028
Summary Functional nanomaterials are predicted to have an enormous impact on some of the most pressing issues of 21st century society, including next-generation health care and energy related technologies. Bottom-up approaches, using self-assembly principles, are increasingly considered to be the most appropriate routes for their synthesis. Indeed, Science magazine highlighted How far can we push chemical self-assembly? as one of the 25 biggest questions that face scientific inquiry over the next quarter century. Despite significant advances in recent years, it is still a major challenge to access precisely defined nano-structures in the laboratory, especially if these do not represent the global free energy minimum (i.e. are asymmetric, multifunctional, compartmentalized and/or dynamic). The biological world provides numerous outstanding examples of highly complex functional nano-scale architectures with attractive features such as defect repair, adaptability, molecular recognition and programmability. It is the objective of this ERC Starting Grant to develop and exploit the concept of (bio-)catalytic self-assembly, a bio-inspired approach for bottom-up synthesis of complex nanomaterials. We will explore three unique features of these systems (i) spatiotemporal control, (ii) catalytic amplification, either towards or away from equilibrium and the tempting vision of (iii) dynamic systems with emergent properties. In our approach we aim to encompass the entire spectrum from fundamental understanding to eventual societal benefit. Alongside the fundamental aims, we wish to put our methodologies to use, in collaboration with experts in these fields, to develop novel functional materials towards applications in next-generation biomaterials and gel-phase supramolecular (opto-) electronic materials.
Summary
Functional nanomaterials are predicted to have an enormous impact on some of the most pressing issues of 21st century society, including next-generation health care and energy related technologies. Bottom-up approaches, using self-assembly principles, are increasingly considered to be the most appropriate routes for their synthesis. Indeed, Science magazine highlighted How far can we push chemical self-assembly? as one of the 25 biggest questions that face scientific inquiry over the next quarter century. Despite significant advances in recent years, it is still a major challenge to access precisely defined nano-structures in the laboratory, especially if these do not represent the global free energy minimum (i.e. are asymmetric, multifunctional, compartmentalized and/or dynamic). The biological world provides numerous outstanding examples of highly complex functional nano-scale architectures with attractive features such as defect repair, adaptability, molecular recognition and programmability. It is the objective of this ERC Starting Grant to develop and exploit the concept of (bio-)catalytic self-assembly, a bio-inspired approach for bottom-up synthesis of complex nanomaterials. We will explore three unique features of these systems (i) spatiotemporal control, (ii) catalytic amplification, either towards or away from equilibrium and the tempting vision of (iii) dynamic systems with emergent properties. In our approach we aim to encompass the entire spectrum from fundamental understanding to eventual societal benefit. Alongside the fundamental aims, we wish to put our methodologies to use, in collaboration with experts in these fields, to develop novel functional materials towards applications in next-generation biomaterials and gel-phase supramolecular (opto-) electronic materials.
Max ERC Funding
1 500 000 €
Duration
Start date: 2011-01-01, End date: 2015-12-31
Project acronym EMPSI
Project Receptors, Channels and Transporters:
Development and Application of Novel Technologies for Structure Determination
Researcher (PI) Christopher Gordon Tate
Host Institution (HI) MEDICAL RESEARCH COUNCIL
Call Details Advanced Grant (AdG), LS1, ERC-2013-ADG
Summary Structure determination of G protein-coupled receptors (GPCRs) has been exceedingly successful over the last 5 years due to the development of complimentary generic methodologies that will now allow the structure determination of virtually any GPCR. However, these technologies address only two aspects of the process, namely the stability of the receptors during purification and the ability to form well-diffracting crystals. The strategies also apply only to GPCRs and not transporters or ion channels. The recent successes have been of GPCRs that are expressed in either yeasts or in insect cells using the baculovirus expression system, but many membrane proteins are expressed poorly in these systems or may be expressed in a misfolded non-functional form. A second issue with the future structure determination of GPCRs is the lack of generic technologies to allow the crystallisation of arrestin-GPCR and G protein-GPCR complexes. Although one G protein GPCR complex has been crystallised this was exceedingly diffciult and resulted in poor resolution of the GPCR component of the complex. We believe that it is possible to thermostabilise both arrestin and heterotrimeric G proteins, which will allow a simplified strategy for the crystallisation and structure determination of GPCR complexes. This is based on the development of the strategy of conformational thermostabilisation of GPCRs developed in our lab that has resulted in the structure determination of 3 different GPCRs bound to either antagonists, partial agonists, full agonists and/or biased agonists.
The aims are:
1. The development of generic methodology for the production of eukaryotic membrane proteins in mammalian cells.
2. The development of a thermostable functional arrestin mutant
3. Structures of β1-adrenoceptor, adenosine A2A receptor and angiotensin receptor bound to a G protein and arrestin
4. Understanding the role of each amino acid residue in the activation process of GPCRs through saturation mutagenes
Summary
Structure determination of G protein-coupled receptors (GPCRs) has been exceedingly successful over the last 5 years due to the development of complimentary generic methodologies that will now allow the structure determination of virtually any GPCR. However, these technologies address only two aspects of the process, namely the stability of the receptors during purification and the ability to form well-diffracting crystals. The strategies also apply only to GPCRs and not transporters or ion channels. The recent successes have been of GPCRs that are expressed in either yeasts or in insect cells using the baculovirus expression system, but many membrane proteins are expressed poorly in these systems or may be expressed in a misfolded non-functional form. A second issue with the future structure determination of GPCRs is the lack of generic technologies to allow the crystallisation of arrestin-GPCR and G protein-GPCR complexes. Although one G protein GPCR complex has been crystallised this was exceedingly diffciult and resulted in poor resolution of the GPCR component of the complex. We believe that it is possible to thermostabilise both arrestin and heterotrimeric G proteins, which will allow a simplified strategy for the crystallisation and structure determination of GPCR complexes. This is based on the development of the strategy of conformational thermostabilisation of GPCRs developed in our lab that has resulted in the structure determination of 3 different GPCRs bound to either antagonists, partial agonists, full agonists and/or biased agonists.
The aims are:
1. The development of generic methodology for the production of eukaryotic membrane proteins in mammalian cells.
2. The development of a thermostable functional arrestin mutant
3. Structures of β1-adrenoceptor, adenosine A2A receptor and angiotensin receptor bound to a G protein and arrestin
4. Understanding the role of each amino acid residue in the activation process of GPCRs through saturation mutagenes
Max ERC Funding
2 378 162 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym ENABLE
Project Elucidating natural bilayer lipid environments
Researcher (PI) Carol Robinson
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Advanced Grant (AdG), LS1, ERC-2015-AdG
Summary Excising a membrane protein from its natural environment, preserving the lipid bilayer, and characterising the lipids that surround it is the ‘holy grail’ of membrane protein biophysics. However, with some 40,000 different lipid structures the challenges we face in understanding selective binding arise not just from the complexity and dynamics of the lipidome, but also from the transient nature of protein lipid interactions. To overcome these challenges we will take mass spectrometry (MS) into a new era, allowing, for the first time, the study of proteins in an environment as close as possible to the natural one. To do this we will (i) characterise protein lipid interactions by employing a high resolution Orbitrap mass spectrometer developed in-house, specifically for membrane proteins, (ii) capture the native lipid environment in vehicles suitable for use in conjunction with MS, and (iii) establish a new platform to be known as integral membrane protein desorption electrospray ionization (impDESI). Designed and built in-house impDESI is capable of releasing membrane proteins from surfaces directly into the mass spectrometer (MS). We will develop impDESI for membrane mimetics, and subsequently portions of natural membranes, enabling us to extract proteins with oligomeric state preserved and native lipid binding intact. The development of impDESI, in conjunction with high resolution Orbitrap MS, and coupled with the optimisation of membrane mimetics, has the potential to radically transform our understanding of native lipid binding, not only directly, but also temporally and spatially. Together these advances will answer key questions about how lipids modulate protein interfaces, occupy different binding sites, modulate membrane protein structure and modify function in vivo. Given the importance of membrane proteins as potential drugs targets understanding their modulation by lipids would be a major step towards more effective drug development.
Summary
Excising a membrane protein from its natural environment, preserving the lipid bilayer, and characterising the lipids that surround it is the ‘holy grail’ of membrane protein biophysics. However, with some 40,000 different lipid structures the challenges we face in understanding selective binding arise not just from the complexity and dynamics of the lipidome, but also from the transient nature of protein lipid interactions. To overcome these challenges we will take mass spectrometry (MS) into a new era, allowing, for the first time, the study of proteins in an environment as close as possible to the natural one. To do this we will (i) characterise protein lipid interactions by employing a high resolution Orbitrap mass spectrometer developed in-house, specifically for membrane proteins, (ii) capture the native lipid environment in vehicles suitable for use in conjunction with MS, and (iii) establish a new platform to be known as integral membrane protein desorption electrospray ionization (impDESI). Designed and built in-house impDESI is capable of releasing membrane proteins from surfaces directly into the mass spectrometer (MS). We will develop impDESI for membrane mimetics, and subsequently portions of natural membranes, enabling us to extract proteins with oligomeric state preserved and native lipid binding intact. The development of impDESI, in conjunction with high resolution Orbitrap MS, and coupled with the optimisation of membrane mimetics, has the potential to radically transform our understanding of native lipid binding, not only directly, but also temporally and spatially. Together these advances will answer key questions about how lipids modulate protein interfaces, occupy different binding sites, modulate membrane protein structure and modify function in vivo. Given the importance of membrane proteins as potential drugs targets understanding their modulation by lipids would be a major step towards more effective drug development.
Max ERC Funding
2 481 744 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
Project acronym EnBioN
Project Engineering the Biointerface of Nanowires to Direct Stem Cell Differentiation
Researcher (PI) Ciro CHIAPPINI
Host Institution (HI) KING'S COLLEGE LONDON
Call Details Starting Grant (StG), PE5, ERC-2017-STG
Summary ENBION will engineer a platform to direct the differentiation of stem cells by developing principles for the rational design of the biointerface of nanowires.
It is increasingly evident that efficient tissue regeneration can only ensue from combining the regenerative potential of stem cells with regulatory stimuli from gene therapy and niche engineering. Yet, despite significant advances towards integrating these technologies, the necessary degree of control over cell fate remains elusive.
Vertical arrays of high aspect ratio nanostructures (nanowires) are rapidly emerging as promising tools to direct cell fate. Thanks to their unique biointerface, nanowires enable gene delivery, intracellular sensing, and direct stimulation of signalling pathways, achieving dynamic manipulation of cells and their environment.
This broad manipulation potential highlights the importance and timeliness of engineering nanowires for regenerative medicine. However, developing a nanowire platform to direct stem cell fate requires design principles based on the largely unknown biological processes governing their interaction with cells. Enabling localized, vector-free gene therapy through efficient transfection relies on understanding the still debated mechanisms by which nanowires induce membrane permeability. Directing cell reprogramming requires understanding the largely unexplored mechanosensory processes and the resulting epigenetic effects arising from the direct interaction of nanowires with multiple organelles within the cell. Engineering the cell microenvironment requires yet undeveloped strategies to localize signalling and transfection with a resolution comparable to the lengthscale of cells.
ENBION will develop this critical knowledge and integrate it into guidelines for dynamic manipulation of cells. Beyond the nanowire platform, the principles highlighted by this unique interface can guide the development of nanomaterials with improved control over cellular processes.
Summary
ENBION will engineer a platform to direct the differentiation of stem cells by developing principles for the rational design of the biointerface of nanowires.
It is increasingly evident that efficient tissue regeneration can only ensue from combining the regenerative potential of stem cells with regulatory stimuli from gene therapy and niche engineering. Yet, despite significant advances towards integrating these technologies, the necessary degree of control over cell fate remains elusive.
Vertical arrays of high aspect ratio nanostructures (nanowires) are rapidly emerging as promising tools to direct cell fate. Thanks to their unique biointerface, nanowires enable gene delivery, intracellular sensing, and direct stimulation of signalling pathways, achieving dynamic manipulation of cells and their environment.
This broad manipulation potential highlights the importance and timeliness of engineering nanowires for regenerative medicine. However, developing a nanowire platform to direct stem cell fate requires design principles based on the largely unknown biological processes governing their interaction with cells. Enabling localized, vector-free gene therapy through efficient transfection relies on understanding the still debated mechanisms by which nanowires induce membrane permeability. Directing cell reprogramming requires understanding the largely unexplored mechanosensory processes and the resulting epigenetic effects arising from the direct interaction of nanowires with multiple organelles within the cell. Engineering the cell microenvironment requires yet undeveloped strategies to localize signalling and transfection with a resolution comparable to the lengthscale of cells.
ENBION will develop this critical knowledge and integrate it into guidelines for dynamic manipulation of cells. Beyond the nanowire platform, the principles highlighted by this unique interface can guide the development of nanomaterials with improved control over cellular processes.
Max ERC Funding
1 495 430 €
Duration
Start date: 2018-02-01, End date: 2023-01-31
Project acronym ENERCAPSULE
Project Nanoencapsulation for Energy Storage and Controlled Release
Researcher (PI) Dzmitry Shchukin
Host Institution (HI) THE UNIVERSITY OF LIVERPOOL
Call Details Consolidator Grant (CoG), PE5, ERC-2014-CoG
Summary The main vision of the project ENERCAPSULE is the development of nanoencapsulation technologies based on switchable nanoscale barriers for novel generation of controlled energy storage and delivery systems. These systems will be based on the “smart” nanocontainers (size below 200 nm) loaded with the energy-enriched active components: materials for thermal energy (both latent and based on chemical reactions) storage and substances for bioenergy (ATP or its components) storage for synthetic biology platforms. First novelty of the proposed project is the protection of the nanoscaled energy-enriched materials against environment during storage and controlled release of the encapsulated energy on demand only using both inherent properties of nanocontainer shell or biomimetic nanovalves introduced as shell components. Another main objective of the project is to study the structure and surface-to-volume properties of the energy enriched materials dispersed and encapsulated on nanoscale. The questions of stability of energy nanomaterials, influence of the nanocontainer shell on their energy capacity, homogeneity and operation lifetime will be investigated. Polymer organic nanocapsules with hollow interior and mesoporous carbon nanoparticles are chosen in the project as main types of the nanocontainer scaffolds for energy-enriched materials due to their high loading capacity and potential to design their shells to attain them controlled permeability properties. At the end of the project, developed novel energy storage and delivery systems will be combined within one network having several mechanisms for release and uptake of energy, which can be activated depending on type and intensity of the external impact (demand). The potential applications of such multienergy storage systems will be tested by industrial companies supporting the project.
Summary
The main vision of the project ENERCAPSULE is the development of nanoencapsulation technologies based on switchable nanoscale barriers for novel generation of controlled energy storage and delivery systems. These systems will be based on the “smart” nanocontainers (size below 200 nm) loaded with the energy-enriched active components: materials for thermal energy (both latent and based on chemical reactions) storage and substances for bioenergy (ATP or its components) storage for synthetic biology platforms. First novelty of the proposed project is the protection of the nanoscaled energy-enriched materials against environment during storage and controlled release of the encapsulated energy on demand only using both inherent properties of nanocontainer shell or biomimetic nanovalves introduced as shell components. Another main objective of the project is to study the structure and surface-to-volume properties of the energy enriched materials dispersed and encapsulated on nanoscale. The questions of stability of energy nanomaterials, influence of the nanocontainer shell on their energy capacity, homogeneity and operation lifetime will be investigated. Polymer organic nanocapsules with hollow interior and mesoporous carbon nanoparticles are chosen in the project as main types of the nanocontainer scaffolds for energy-enriched materials due to their high loading capacity and potential to design their shells to attain them controlled permeability properties. At the end of the project, developed novel energy storage and delivery systems will be combined within one network having several mechanisms for release and uptake of energy, which can be activated depending on type and intensity of the external impact (demand). The potential applications of such multienergy storage systems will be tested by industrial companies supporting the project.
Max ERC Funding
2 004 500 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym ENOLCAT
Project Emulating Nature: Reaction Diversity and Understanding through Asymmetric Catalysis
Researcher (PI) Andrew David Smith
Host Institution (HI) THE UNIVERSITY COURT OF THE UNIVERSITY OF ST ANDREWS
Call Details Starting Grant (StG), PE5, ERC-2011-StG_20101014
Summary The remarkable way that Nature prepares complex natural products has always been a source of inspiration to scientists, stimulating the development of new synthetic methods and strategies, as elegantly demonstrated by biomimetic approaches to total synthesis. Similarly, the performance and specificity of enzymes, perfected though evolution, offer ideals of selectivity and specificity that synthetic chemists aspire to. This proposal aims to develop an internationally leading research programme inspired by Nature’s ability to selectively generate diverse products from simple materials with exquisite levels of regio- and enantiocontrol. We aspire to synthetically emulate the elegant behaviour of Nature’s building blocks, such as co-enzyme A, in their ability to generate synthetic diversity (such as polyketides and alkaloids) from a simple and common starting material. Using this blueprint, we intend to selectively control the synthesis of a diverse range of bespoke stereodefined carbo- and heterocycles from readily available starting materials using simple man-made catalysts. We specifically aim to develop new strategies within the field of organic catalysis, focused upon the development of methods for the in situ catalytic generation of chiral ammonium enolates from carboxylic acids and their employment in catalysis. We also propose to develop a comprehensive mechanistic understanding of these processes. In preliminary work we have delineated a simple and efficient approach to this problem by employing chiral isothioureas as asymmetric catalysts, and we aim to build on the insight provided by these studies to develop this powerful concept into a generally applicable synthetic strategy.
Summary
The remarkable way that Nature prepares complex natural products has always been a source of inspiration to scientists, stimulating the development of new synthetic methods and strategies, as elegantly demonstrated by biomimetic approaches to total synthesis. Similarly, the performance and specificity of enzymes, perfected though evolution, offer ideals of selectivity and specificity that synthetic chemists aspire to. This proposal aims to develop an internationally leading research programme inspired by Nature’s ability to selectively generate diverse products from simple materials with exquisite levels of regio- and enantiocontrol. We aspire to synthetically emulate the elegant behaviour of Nature’s building blocks, such as co-enzyme A, in their ability to generate synthetic diversity (such as polyketides and alkaloids) from a simple and common starting material. Using this blueprint, we intend to selectively control the synthesis of a diverse range of bespoke stereodefined carbo- and heterocycles from readily available starting materials using simple man-made catalysts. We specifically aim to develop new strategies within the field of organic catalysis, focused upon the development of methods for the in situ catalytic generation of chiral ammonium enolates from carboxylic acids and their employment in catalysis. We also propose to develop a comprehensive mechanistic understanding of these processes. In preliminary work we have delineated a simple and efficient approach to this problem by employing chiral isothioureas as asymmetric catalysts, and we aim to build on the insight provided by these studies to develop this powerful concept into a generally applicable synthetic strategy.
Max ERC Funding
1 497 005 €
Duration
Start date: 2011-10-01, End date: 2017-06-30
Project acronym ENTANGLED-TM-ALKANE
Project Entangled pincer ligand architectures and their application in the transition-metal-mediated activation of alkanes
Researcher (PI) Adrian Benjamin Chaplin
Host Institution (HI) THE UNIVERSITY OF WARWICK
Call Details Starting Grant (StG), PE5, ERC-2014-STG
Summary The selective transformation of alkanes is an area of contemporary importance with wide-ranging implications for organic synthesis and the effective use of petroleum resources. While homogeneous transition metal catalysis is a potentially powerful means for achieving this objective, the fundamental organometallic chemistry of alkane activation reactions has proven to be exceedingly difficult to investigate due to the weakly interacting nature of alkanes. To address this knowledge gap and provide the foundation for future advancement of the field, ENTANGLED-TM-ALKANE outlines a systematic approach for the study of pivotal sigma–alkane complex intermediates; nominally transient and extremely reactive metal-alkane adducts formed through coordination of an intact C–H bond to the metal centre. Inspired from supramolecular chemistry, the approach involves the innovative use of systems containing alkane substrates held in close proximity to reactive metal centres through mechanical entanglement within supporting tridentate macrocyclic ‘pincer’ ligands (i.e. alkane based [2]rotaxanes and [2]catenanes). Through the interwoven topology of these systems, problematic dissociation reactions of sigma–alkane complexes will be circumvented, facilitating isolation and ultimately enabling their structure and reaction chemistry to be probed in much greater detail than has been previously possible. The project objectives are to: (a) develop and use new synthetic (supramolecular) methodologies for the preparation of these mechanically interlocked metal-alkane assemblies; (b) systematically investigate the organometallic chemistry of the metal centre and its interaction with the entangled alkane; and through variation of the macromolecules’ components (macrocycle donors and geometry, alkane, metal), (c) compile a definitive and unprecedented body of qualitative and quantitative structure-activity relationships for the activation alkanes using transition metals.
Summary
The selective transformation of alkanes is an area of contemporary importance with wide-ranging implications for organic synthesis and the effective use of petroleum resources. While homogeneous transition metal catalysis is a potentially powerful means for achieving this objective, the fundamental organometallic chemistry of alkane activation reactions has proven to be exceedingly difficult to investigate due to the weakly interacting nature of alkanes. To address this knowledge gap and provide the foundation for future advancement of the field, ENTANGLED-TM-ALKANE outlines a systematic approach for the study of pivotal sigma–alkane complex intermediates; nominally transient and extremely reactive metal-alkane adducts formed through coordination of an intact C–H bond to the metal centre. Inspired from supramolecular chemistry, the approach involves the innovative use of systems containing alkane substrates held in close proximity to reactive metal centres through mechanical entanglement within supporting tridentate macrocyclic ‘pincer’ ligands (i.e. alkane based [2]rotaxanes and [2]catenanes). Through the interwoven topology of these systems, problematic dissociation reactions of sigma–alkane complexes will be circumvented, facilitating isolation and ultimately enabling their structure and reaction chemistry to be probed in much greater detail than has been previously possible. The project objectives are to: (a) develop and use new synthetic (supramolecular) methodologies for the preparation of these mechanically interlocked metal-alkane assemblies; (b) systematically investigate the organometallic chemistry of the metal centre and its interaction with the entangled alkane; and through variation of the macromolecules’ components (macrocycle donors and geometry, alkane, metal), (c) compile a definitive and unprecedented body of qualitative and quantitative structure-activity relationships for the activation alkanes using transition metals.
Max ERC Funding
1 521 137 €
Duration
Start date: 2015-04-01, End date: 2020-03-31
Project acronym enzC-Hem
Project Creating Versatile Metallo-Enzyme Environments for Selective C-H Activation Chemistry: Lignocellulose Deconstruction and Beyond
Researcher (PI) Anthony GREEN
Host Institution (HI) THE UNIVERSITY OF MANCHESTER
Call Details Starting Grant (StG), PE5, ERC-2017-STG
Summary The availability of a versatile catalytic platform to precisely target and functionalize individual C-H bonds in complex organic molecules would revolutionize our synthetic strategies, leading to streamlined routes to high value chemicals and supporting the development of a ‘greener’ chemical industry. Although an impressive range of C-H functionalizations can be achieved with small transition metal complexes, site selectivity is often determined by features of the substrate, and not by the catalyst. A general approach to achieve the more aspirational ‘catalyst controlled’ transformations requires molecular recognition elements within the catalyst which: a) allow precise substrate orientation and b) can be tuned to alter selectivity. In principle, these requirements could be perfectly addressed by protein catalysts which can be readily adapted via laboratory evolution. However, enzyme engineering strategies are currently limited to Nature’s twenty amino acid alphabet, severely limiting the range of metal co-ordination environments, and thus catalytic activities, that are accessible within proteins.
In enzC-Hem, I will exploit advanced protein engineering technology available in my laboratory to install ‘chemically programmed’ ligands and/or noble metal co-factors into selected enzyme scaffolds. I will show that the resulting C-H activation catalysts can be systematically optimized via directed evolution with an expanded genetic code using modern ultra-high throughput methods (>100 variants per second), yielding biocatalysts with augmented selectivity/activity profiles. Thus my approach merges the broad range of C-H functionalizations accessible with small molecule catalysts with precise control of selectivity provided by proteins. The biocatalysts developed will address major global challenges in biotechnology and synthetic chemistry, from enhancing lignocellulose derived biofuel production to revealing novel bioactive molecules via late-stage functionalizations.
Summary
The availability of a versatile catalytic platform to precisely target and functionalize individual C-H bonds in complex organic molecules would revolutionize our synthetic strategies, leading to streamlined routes to high value chemicals and supporting the development of a ‘greener’ chemical industry. Although an impressive range of C-H functionalizations can be achieved with small transition metal complexes, site selectivity is often determined by features of the substrate, and not by the catalyst. A general approach to achieve the more aspirational ‘catalyst controlled’ transformations requires molecular recognition elements within the catalyst which: a) allow precise substrate orientation and b) can be tuned to alter selectivity. In principle, these requirements could be perfectly addressed by protein catalysts which can be readily adapted via laboratory evolution. However, enzyme engineering strategies are currently limited to Nature’s twenty amino acid alphabet, severely limiting the range of metal co-ordination environments, and thus catalytic activities, that are accessible within proteins.
In enzC-Hem, I will exploit advanced protein engineering technology available in my laboratory to install ‘chemically programmed’ ligands and/or noble metal co-factors into selected enzyme scaffolds. I will show that the resulting C-H activation catalysts can be systematically optimized via directed evolution with an expanded genetic code using modern ultra-high throughput methods (>100 variants per second), yielding biocatalysts with augmented selectivity/activity profiles. Thus my approach merges the broad range of C-H functionalizations accessible with small molecule catalysts with precise control of selectivity provided by proteins. The biocatalysts developed will address major global challenges in biotechnology and synthetic chemistry, from enhancing lignocellulose derived biofuel production to revealing novel bioactive molecules via late-stage functionalizations.
Max ERC Funding
1 492 424 €
Duration
Start date: 2018-01-01, End date: 2022-12-31