Project acronym AGRISCENTS
Project Scents and sensibility in agriculture: exploiting specificity in herbivore- and pathogen-induced plant volatiles for real-time crop monitoring
Researcher (PI) Theodoor Turlings
Host Institution (HI) UNIVERSITE DE NEUCHATEL
Country Switzerland
Call Details Advanced Grant (AdG), LS9, ERC-2017-ADG
Summary Plants typically release large quantities of volatiles in response to attack by herbivores or pathogens. I may claim to have contributed to various breakthroughs in this research field, including the discovery that the volatile blends induced by different attackers are astonishingly specific, resulting in characteristic, readily distinguishable odour blends. Using maize as our model plant, I wish to take several leaps forward in our understanding of this signal specificity and use this knowledge to develop sensors for the real-time detection of crop pests and diseases. For this, three interconnected work-packages will aim to:
• Develop chemical analytical techniques and statistical models to decipher the odorous vocabulary of plants, and to create a complete inventory of “odour-prints” for a wide range of herbivore-plant and pathogen-plant combinations, including simultaneous infestations.
• Develop and optimize nano-mechanical sensors for the detection of specific plant volatile mixtures. For this, we will initially adapt a prototype sensor that has been successfully developed for the detection of cancer-related volatiles in human breath.
• Genetically manipulate maize plants to release a unique blend of root-produced volatiles upon herbivory. For this, we will engineer gene cassettes that combine recently identified P450 (CYP) genes from poplar with inducible, root-specific promoters from maize. This will result in maize plants that, in response to pest attack, release easy-to-detect aldoximes and nitriles from their roots.
In short, by investigating and manipulating the specificity of inducible odour blends we will generate the necessary knowhow to develop a novel odour-detection device. The envisioned sensor technology will permit real-time monitoring of the pests and enable farmers to apply crop protection treatments at the right time and in the right place.
Summary
Plants typically release large quantities of volatiles in response to attack by herbivores or pathogens. I may claim to have contributed to various breakthroughs in this research field, including the discovery that the volatile blends induced by different attackers are astonishingly specific, resulting in characteristic, readily distinguishable odour blends. Using maize as our model plant, I wish to take several leaps forward in our understanding of this signal specificity and use this knowledge to develop sensors for the real-time detection of crop pests and diseases. For this, three interconnected work-packages will aim to:
• Develop chemical analytical techniques and statistical models to decipher the odorous vocabulary of plants, and to create a complete inventory of “odour-prints” for a wide range of herbivore-plant and pathogen-plant combinations, including simultaneous infestations.
• Develop and optimize nano-mechanical sensors for the detection of specific plant volatile mixtures. For this, we will initially adapt a prototype sensor that has been successfully developed for the detection of cancer-related volatiles in human breath.
• Genetically manipulate maize plants to release a unique blend of root-produced volatiles upon herbivory. For this, we will engineer gene cassettes that combine recently identified P450 (CYP) genes from poplar with inducible, root-specific promoters from maize. This will result in maize plants that, in response to pest attack, release easy-to-detect aldoximes and nitriles from their roots.
In short, by investigating and manipulating the specificity of inducible odour blends we will generate the necessary knowhow to develop a novel odour-detection device. The envisioned sensor technology will permit real-time monitoring of the pests and enable farmers to apply crop protection treatments at the right time and in the right place.
Max ERC Funding
2 498 086 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym AlgoRNN
Project Recurrent Neural Networks and Related Machines That Learn Algorithms
Researcher (PI) Juergen Schmidhuber
Host Institution (HI) UNIVERSITA DELLA SVIZZERA ITALIANA
Country Switzerland
Call Details Advanced Grant (AdG), PE6, ERC-2016-ADG
Summary Recurrent neural networks (RNNs) are general parallel-sequential computers. Some learn their programs or weights. Our supervised Long Short-Term Memory (LSTM) RNNs were the first to win pattern recognition contests, and recently enabled best known results in speech and handwriting recognition, machine translation, etc. They are now available to billions of users through the world's most valuable public companies including Google and Apple. Nevertheless, in lots of real-world tasks RNNs do not yet live up to their full potential. Although universal in theory, in practice they fail to learn important types of algorithms. This ERC project will go far beyond today's best RNNs through novel RNN-like systems that address some of the biggest open RNN problems and hottest RNN research topics: (1) How can RNNs learn to control (through internal spotlights of attention) separate large short-memory structures such as sub-networks with fast weights, to improve performance on many natural short-term memory-intensive tasks which are currently hard to learn by RNNs, such as answering detailed questions on recently observed videos? (2) How can such RNN-like systems metalearn entire learning algorithms that outperform the original learning algorithms? (3) How to achieve efficient transfer learning from one RNN-learned set of problem-solving programs to new RNN programs solving new tasks? In other words, how can one RNN-like system actively learn to exploit algorithmic information contained in the programs running on another? We will test our systems existing benchmarks, and create new, more challenging multi-task benchmarks. This will be supported by a rather cheap, GPU-based mini-brain for implementing large RNNs.
Summary
Recurrent neural networks (RNNs) are general parallel-sequential computers. Some learn their programs or weights. Our supervised Long Short-Term Memory (LSTM) RNNs were the first to win pattern recognition contests, and recently enabled best known results in speech and handwriting recognition, machine translation, etc. They are now available to billions of users through the world's most valuable public companies including Google and Apple. Nevertheless, in lots of real-world tasks RNNs do not yet live up to their full potential. Although universal in theory, in practice they fail to learn important types of algorithms. This ERC project will go far beyond today's best RNNs through novel RNN-like systems that address some of the biggest open RNN problems and hottest RNN research topics: (1) How can RNNs learn to control (through internal spotlights of attention) separate large short-memory structures such as sub-networks with fast weights, to improve performance on many natural short-term memory-intensive tasks which are currently hard to learn by RNNs, such as answering detailed questions on recently observed videos? (2) How can such RNN-like systems metalearn entire learning algorithms that outperform the original learning algorithms? (3) How to achieve efficient transfer learning from one RNN-learned set of problem-solving programs to new RNN programs solving new tasks? In other words, how can one RNN-like system actively learn to exploit algorithmic information contained in the programs running on another? We will test our systems existing benchmarks, and create new, more challenging multi-task benchmarks. This will be supported by a rather cheap, GPU-based mini-brain for implementing large RNNs.
Max ERC Funding
2 500 000 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym ALK7
Project Metabolic control by the TGF-² superfamily receptor ALK7: A novel regulator of insulin secretion, fat accumulation and energy balance
Researcher (PI) Carlos Ibanez
Host Institution (HI) KAROLINSKA INSTITUTET
Country Sweden
Call Details Advanced Grant (AdG), LS4, ERC-2008-AdG
Summary The aim of this proposal is to understand a novel regulatory signaling network controlling insulin secretion, fat accumulation and energy balance centered around selected components of the TGF-² signaling system, including Activins A and B, GDF-3 and their receptors ALK7 and ALK4. Recent results from my laboratory indicate that these molecules are part of paracrine signaling networks that control important functions in pancreatic islets and adipose tissue through feedback inhibition and feed-forward regulation. These discoveries have open up a new research area with important implications for the understanding of metabolic networks and the treatment of human metabolic syndromes, such as diabetes and obesity.
To drive progress in this new research area beyond the state-of-the-art it is proposed to: i) Elucidate the molecular mechanisms by which Activins regulate Ca2+ influx and insulin secretion in pancreatic ²-cells; ii) Elucidate the molecular mechanisms underlying the effects of GDF-3 on adipocyte metabolism, turnover and fat accumulation; iii) Investigate the interplay between insulin levels and fat deposition in the development of insulin resistance using mutant mice lacking Activin B and GDF-3; iv) Investigate tissue-specific contributions of ALK7 and ALK4 signaling to metabolic control by generating and characterizing conditional mutant mice; v) Investigate the effects of specific and reversible inactivation of ALK7 and ALK4 on metabolic regulation using a novel chemical-genetic approach based on analog-sensitive alleles.
This is research of a high-gain/high-risk nature. It is posed to open unique opportunities for further exploration of complex metabolic networks. The development of drugs capable of enhancing insulin secretion, limiting fat accumulation and ameliorating diet-induced obesity by targeting components of the ALK7 signaling network will find a strong rationale in the results of the proposed work.
Summary
The aim of this proposal is to understand a novel regulatory signaling network controlling insulin secretion, fat accumulation and energy balance centered around selected components of the TGF-² signaling system, including Activins A and B, GDF-3 and their receptors ALK7 and ALK4. Recent results from my laboratory indicate that these molecules are part of paracrine signaling networks that control important functions in pancreatic islets and adipose tissue through feedback inhibition and feed-forward regulation. These discoveries have open up a new research area with important implications for the understanding of metabolic networks and the treatment of human metabolic syndromes, such as diabetes and obesity.
To drive progress in this new research area beyond the state-of-the-art it is proposed to: i) Elucidate the molecular mechanisms by which Activins regulate Ca2+ influx and insulin secretion in pancreatic ²-cells; ii) Elucidate the molecular mechanisms underlying the effects of GDF-3 on adipocyte metabolism, turnover and fat accumulation; iii) Investigate the interplay between insulin levels and fat deposition in the development of insulin resistance using mutant mice lacking Activin B and GDF-3; iv) Investigate tissue-specific contributions of ALK7 and ALK4 signaling to metabolic control by generating and characterizing conditional mutant mice; v) Investigate the effects of specific and reversible inactivation of ALK7 and ALK4 on metabolic regulation using a novel chemical-genetic approach based on analog-sensitive alleles.
This is research of a high-gain/high-risk nature. It is posed to open unique opportunities for further exploration of complex metabolic networks. The development of drugs capable of enhancing insulin secretion, limiting fat accumulation and ameliorating diet-induced obesity by targeting components of the ALK7 signaling network will find a strong rationale in the results of the proposed work.
Max ERC Funding
2 462 154 €
Duration
Start date: 2009-04-01, End date: 2014-03-31
Project acronym ALMA
Project Attosecond Control of Light and Matter
Researcher (PI) Anne L'huillier
Host Institution (HI) MAX IV Laboratory, Lund University
Country Sweden
Call Details Advanced Grant (AdG), PE2, ERC-2008-AdG
Summary Attosecond light pulses are generated when an intense laser interacts with a gas target. These pulses are not only short, enabling the study of electronic processes at their natural time scale, but also coherent. The vision of this proposal is to extend temporal coherent control concepts to a completely new regime of time and energy, combining (i) ultrashort pulses (ii) broadband excitation (iii) high photon energy, allowing scientists to reach not only valence but also inner shells in atoms and molecules, and, when needed, (iv) high spatial resolution. We want to explore how elementary electronic processes in atoms, molecules and more complex systems can be controlled by using well designed sequences of attosecond pulses. The research project proposed is organized into four parts: 1. Attosecond control of light leading to controlled sequences of attosecond pulses We will develop techniques to generate sequences of attosecond pulses with a variable number of pulses and controlled carrier-envelope-phase variation between consecutive pulses. 2. Attosecond control of electronic processes in atoms and molecules We will investigate the dynamics and coherence of phenomena induced by attosecond excitation of electron wave packets in various systems and we will explore how they can be controlled by a controlled sequence of ultrashort pulses. 3. Intense attosecond sources to reach the nonlinear regime We will optimize attosecond light sources in a systematic way, including amplification of the radiation by injecting a free electron laser. This will open up the possibility to develop nonlinear measurement and control schemes. 4. Attosecond control in more complex systems, including high spatial resolution We will develop ultrafast microscopy techniques, in order to obtain meaningful temporal information in surface and solid state physics. Two directions will be explored, digital in line microscopic holography and photoemission electron microscopy.
Summary
Attosecond light pulses are generated when an intense laser interacts with a gas target. These pulses are not only short, enabling the study of electronic processes at their natural time scale, but also coherent. The vision of this proposal is to extend temporal coherent control concepts to a completely new regime of time and energy, combining (i) ultrashort pulses (ii) broadband excitation (iii) high photon energy, allowing scientists to reach not only valence but also inner shells in atoms and molecules, and, when needed, (iv) high spatial resolution. We want to explore how elementary electronic processes in atoms, molecules and more complex systems can be controlled by using well designed sequences of attosecond pulses. The research project proposed is organized into four parts: 1. Attosecond control of light leading to controlled sequences of attosecond pulses We will develop techniques to generate sequences of attosecond pulses with a variable number of pulses and controlled carrier-envelope-phase variation between consecutive pulses. 2. Attosecond control of electronic processes in atoms and molecules We will investigate the dynamics and coherence of phenomena induced by attosecond excitation of electron wave packets in various systems and we will explore how they can be controlled by a controlled sequence of ultrashort pulses. 3. Intense attosecond sources to reach the nonlinear regime We will optimize attosecond light sources in a systematic way, including amplification of the radiation by injecting a free electron laser. This will open up the possibility to develop nonlinear measurement and control schemes. 4. Attosecond control in more complex systems, including high spatial resolution We will develop ultrafast microscopy techniques, in order to obtain meaningful temporal information in surface and solid state physics. Two directions will be explored, digital in line microscopic holography and photoemission electron microscopy.
Max ERC Funding
2 250 000 €
Duration
Start date: 2008-12-01, End date: 2013-11-30
Project acronym AMIMOS
Project Agile MIMO Systems for Communications, Biomedicine, and Defense
Researcher (PI) Bjorn Ottersten
Host Institution (HI) KUNGLIGA TEKNISKA HOEGSKOLAN
Country Sweden
Call Details Advanced Grant (AdG), PE7, ERC-2008-AdG
Summary This proposal targets the emerging frontier research field of multiple-input multiple-output (MIMO) systems along with several innovative and somewhat unconventional applications of such systems. The use of arrays of transmitters and receivers will have a profound impact on future medical imaging/therapy systems, radar systems, and radio communication networks. Multiple transmitters provide a tremendous versatility and allow waveforms to be adapted temporally and spatially to environmental conditions. This is useful for individually tailored illumination of human tissue in biomedical imaging or ultrasound therapy. In radar systems, multiple transmit beams can be formed simultaneously via separate waveform designs allowing accurate target classification. In a wireless communication system, multiple communication signals can be directed to one or more users at the same time on the same frequency carrier. In addition, multiple receivers can be used in the above applications to provide increased detection performance, interference rejection, and improved estimation accuracy. The joint modelling, analysis, and design of these multidimensional transmit and receive schemes form the core of this research proposal. Ultimately, our research aims at developing the fundamental tools that will allow the design of wireless communication systems with an order-of-magnitude higher capacity at a lower cost than today; of ultrasound therapy systems maximizing delivered power while reducing treatment duration and unwanted illumination; and of distributed aperture multi-beam radars allowing more effective target location, identification, and classification. Europe has several successful industries that are active in biomedical imaging/therapy, radar systems, and wireless communications. The future success of these sectors critically depends on the ability to innovate and integrate new technology.
Summary
This proposal targets the emerging frontier research field of multiple-input multiple-output (MIMO) systems along with several innovative and somewhat unconventional applications of such systems. The use of arrays of transmitters and receivers will have a profound impact on future medical imaging/therapy systems, radar systems, and radio communication networks. Multiple transmitters provide a tremendous versatility and allow waveforms to be adapted temporally and spatially to environmental conditions. This is useful for individually tailored illumination of human tissue in biomedical imaging or ultrasound therapy. In radar systems, multiple transmit beams can be formed simultaneously via separate waveform designs allowing accurate target classification. In a wireless communication system, multiple communication signals can be directed to one or more users at the same time on the same frequency carrier. In addition, multiple receivers can be used in the above applications to provide increased detection performance, interference rejection, and improved estimation accuracy. The joint modelling, analysis, and design of these multidimensional transmit and receive schemes form the core of this research proposal. Ultimately, our research aims at developing the fundamental tools that will allow the design of wireless communication systems with an order-of-magnitude higher capacity at a lower cost than today; of ultrasound therapy systems maximizing delivered power while reducing treatment duration and unwanted illumination; and of distributed aperture multi-beam radars allowing more effective target location, identification, and classification. Europe has several successful industries that are active in biomedical imaging/therapy, radar systems, and wireless communications. The future success of these sectors critically depends on the ability to innovate and integrate new technology.
Max ERC Funding
1 872 720 €
Duration
Start date: 2009-01-01, End date: 2013-12-31
Project acronym Amygdala Circuits
Project Amygdala Circuits for Appetitive Conditioning
Researcher (PI) Andreas Luthi
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Country Switzerland
Call Details Advanced Grant (AdG), LS5, ERC-2014-ADG
Summary The project outlined here addresses the fundamental question how the brain encodes and controls behavior. While we have a reasonable understanding of the role of entire brain areas in such processes, and of mechanisms at the molecular and synaptic levels, there is a big gap in our knowledge of how behavior is controlled at the level of defined neuronal circuits.
In natural environments, chances for survival depend on learning about possible aversive and appetitive outcomes and on the appropriate behavioral responses. Most studies addressing the underlying mechanisms at the level of neuronal circuits have focused on aversive learning, such as in Pavlovian fear conditioning. Understanding how activity in defined neuronal circuits mediates appetitive learning, as well as how these circuitries are shared and interact with aversive learning circuits, is a central question in the neuroscience of learning and memory and the focus of this grant application.
Using a multidisciplinary approach in mice, combining behavioral, in vivo and in vitro electrophysiological, imaging, optogenetic and state-of-the-art viral circuit tracing techniques, we aim at dissecting the neuronal circuitry of appetitive Pavlovian conditioning with a focus on the amygdala, a key brain region important for both aversive and appetitive learning. Ultimately, elucidating these mechanisms at the level of defined neurons and circuits is fundamental not only for an understanding of memory processes in the brain in general, but also to inform a mechanistic approach to psychiatric conditions associated with amygdala dysfunction and dysregulated emotional responses including anxiety and mood disorders.
Summary
The project outlined here addresses the fundamental question how the brain encodes and controls behavior. While we have a reasonable understanding of the role of entire brain areas in such processes, and of mechanisms at the molecular and synaptic levels, there is a big gap in our knowledge of how behavior is controlled at the level of defined neuronal circuits.
In natural environments, chances for survival depend on learning about possible aversive and appetitive outcomes and on the appropriate behavioral responses. Most studies addressing the underlying mechanisms at the level of neuronal circuits have focused on aversive learning, such as in Pavlovian fear conditioning. Understanding how activity in defined neuronal circuits mediates appetitive learning, as well as how these circuitries are shared and interact with aversive learning circuits, is a central question in the neuroscience of learning and memory and the focus of this grant application.
Using a multidisciplinary approach in mice, combining behavioral, in vivo and in vitro electrophysiological, imaging, optogenetic and state-of-the-art viral circuit tracing techniques, we aim at dissecting the neuronal circuitry of appetitive Pavlovian conditioning with a focus on the amygdala, a key brain region important for both aversive and appetitive learning. Ultimately, elucidating these mechanisms at the level of defined neurons and circuits is fundamental not only for an understanding of memory processes in the brain in general, but also to inform a mechanistic approach to psychiatric conditions associated with amygdala dysfunction and dysregulated emotional responses including anxiety and mood disorders.
Max ERC Funding
2 497 200 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym ANGIOFAT
Project New mechanisms of angiogenesis modulators in switching between white and brown adipose tissues
Researcher (PI) Yihai Cao
Host Institution (HI) KAROLINSKA INSTITUTET
Country Sweden
Call Details Advanced Grant (AdG), LS4, ERC-2009-AdG
Summary Understanding the molecular mechanisms underlying adipose blood vessel growth or regression opens new fundamentally insight into novel therapeutic options for the treatment of obesity and its related metabolic diseases such as type 2 diabetes and cancer. Unlike any other tissues in the body, the adipose tissue constantly experiences expansion and shrinkage throughout the adult life. Adipocytes in the white adipose tissue have the ability to switch into metabolically highly active brown-like adipocytes. Brown adipose tissue (BAT) contains significantly higher numbers of microvessels than white adipose tissue (WAT) in order to adopt the high rates of metabolism. Thus, an angiogenic phenotype has to be switched on during the transition from WAT into BAT. We have found that acclimation of mice in cold could induce transition from inguinal and epidedymal WAT into BAT by upregulation of angiogenic factor expression and down-regulations of angiogenesis inhibitors (Xue et al, Cell Metabolism, 2009). The transition from WAT into BAT is dependent on vascular endothelial growth factor (VEGF) that primarily targets on vascular endothelial cells via a tissue hypoxia-independent mechanism. VEGF blockade significantly alters adipose tissue metabolism. In another genetic model, we show similar findings that angiogenesis is crucial to mediate the transition from WAT into BAT (Xue et al, PNAS, 2008). Here we propose that the vascular tone determines the metabolic switch between WAT and BAT. Characterization of these novel angiogenic pathways may reveal new mechanisms underlying development of obesity- and metabolism-related disease complications and may define novel therapeutic targets. Thus, the benefit of this research proposal is enormous and is aimed to treat the most common and highly risk human health conditions in the modern time.
Summary
Understanding the molecular mechanisms underlying adipose blood vessel growth or regression opens new fundamentally insight into novel therapeutic options for the treatment of obesity and its related metabolic diseases such as type 2 diabetes and cancer. Unlike any other tissues in the body, the adipose tissue constantly experiences expansion and shrinkage throughout the adult life. Adipocytes in the white adipose tissue have the ability to switch into metabolically highly active brown-like adipocytes. Brown adipose tissue (BAT) contains significantly higher numbers of microvessels than white adipose tissue (WAT) in order to adopt the high rates of metabolism. Thus, an angiogenic phenotype has to be switched on during the transition from WAT into BAT. We have found that acclimation of mice in cold could induce transition from inguinal and epidedymal WAT into BAT by upregulation of angiogenic factor expression and down-regulations of angiogenesis inhibitors (Xue et al, Cell Metabolism, 2009). The transition from WAT into BAT is dependent on vascular endothelial growth factor (VEGF) that primarily targets on vascular endothelial cells via a tissue hypoxia-independent mechanism. VEGF blockade significantly alters adipose tissue metabolism. In another genetic model, we show similar findings that angiogenesis is crucial to mediate the transition from WAT into BAT (Xue et al, PNAS, 2008). Here we propose that the vascular tone determines the metabolic switch between WAT and BAT. Characterization of these novel angiogenic pathways may reveal new mechanisms underlying development of obesity- and metabolism-related disease complications and may define novel therapeutic targets. Thus, the benefit of this research proposal is enormous and is aimed to treat the most common and highly risk human health conditions in the modern time.
Max ERC Funding
2 411 547 €
Duration
Start date: 2010-03-01, End date: 2015-02-28
Project acronym ANOBEST
Project Structure function and pharmacology of calcium-activated chloride channels: Anoctamins and Bestrophins
Researcher (PI) Raimund Dutzler
Host Institution (HI) University of Zurich
Country Switzerland
Call Details Advanced Grant (AdG), LS1, ERC-2013-ADG
Summary Calcium-activated chloride channels (CaCCs) play key roles in a range of physiological processes such as the control of membrane excitability, photoreception and epithelial secretion. Although the importance of these channels has been recognized for more than 30 years their molecular identity remained obscure. The recent discovery of two protein families encoding for CaCCs, Anoctamins and Bestrophins, was a scientific breakthrough that has provided first insight into two novel ion channel architectures. Within this proposal we aim to determine the first high resolution structures of members of both families and study their functional behavior by an interdisciplinary approach combining biochemistry, X-ray crystallography and electrophysiology. The structural investigation of eukaryotic membrane proteins is extremely challenging and will require us to investigate large numbers of candidates to single out family members with superior biochemical properties. During the last year we have made large progress in this direction. By screening numerous eukaryotic Anoctamins and prokaryotic Bestrophins we have identified well-behaved proteins for both families, which were successfully scaled-up and purified. Additional family members will be identified within the course of the project. For these stable proteins we plan to grow crystals diffracting to high resolution and to proceed with structure determination. With first structural information in hand we will perform detailed functional studies using electrophysiology and complementary biophysical techniques to gain mechanistic insight into ion permeation and gating. As the pharmacology of both families is still in its infancy we will in later stages also engage in the identification and characterization of inhibitors and activators of Anoctamins and Bestrophins to open up a field that may ultimately lead to the discovery of novel therapeutic strategies targeting calcium-activated chloride channels.
Summary
Calcium-activated chloride channels (CaCCs) play key roles in a range of physiological processes such as the control of membrane excitability, photoreception and epithelial secretion. Although the importance of these channels has been recognized for more than 30 years their molecular identity remained obscure. The recent discovery of two protein families encoding for CaCCs, Anoctamins and Bestrophins, was a scientific breakthrough that has provided first insight into two novel ion channel architectures. Within this proposal we aim to determine the first high resolution structures of members of both families and study their functional behavior by an interdisciplinary approach combining biochemistry, X-ray crystallography and electrophysiology. The structural investigation of eukaryotic membrane proteins is extremely challenging and will require us to investigate large numbers of candidates to single out family members with superior biochemical properties. During the last year we have made large progress in this direction. By screening numerous eukaryotic Anoctamins and prokaryotic Bestrophins we have identified well-behaved proteins for both families, which were successfully scaled-up and purified. Additional family members will be identified within the course of the project. For these stable proteins we plan to grow crystals diffracting to high resolution and to proceed with structure determination. With first structural information in hand we will perform detailed functional studies using electrophysiology and complementary biophysical techniques to gain mechanistic insight into ion permeation and gating. As the pharmacology of both families is still in its infancy we will in later stages also engage in the identification and characterization of inhibitors and activators of Anoctamins and Bestrophins to open up a field that may ultimately lead to the discovery of novel therapeutic strategies targeting calcium-activated chloride channels.
Max ERC Funding
2 176 000 €
Duration
Start date: 2014-02-01, End date: 2020-01-31
Project acronym AOC
Project Adversary-Oriented Computing
Researcher (PI) Rachid Guerraoui
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), PE6, ERC-2013-ADG
Summary "Recent technological evolutions, including the cloud, the multicore, the social and the mobiles ones, are turning computing ubiquitously distributed. Yet, building high-assurance distributed programs is notoriously challenging. One of the main reasons is that these systems usually seek to achieve several goals at the same time. In short, they need to be efficient, responding effectively in various average-case conditions, as well as reliable, behaving correctly in severe, worst-case conditions. As a consequence, they typically intermingle different strategies: each to cope with some specific condition, e.g., with or without node failures, message losses, time-outs, contention, cache misses,
over-sizing, malicious attacks, etc. The resulting programs end up hard to design, prove, verify, implement, test and debug. Not surprisingly, there are anecdotal evidences of the fragility of the most celebrated distributed systems.
The goal of this project is to contribute to building high-assurance distributed programs by introducing a new dimension for separating and isolating their concerns, as well as a new scheme for composing and reusing them in a modular manner. In short, the project will explore the inherent power and limitations of a novel paradigm, Adversary-Oriented Computing (AOC). Sub-programs, each implementing a specific strategy to cope with a given adversary, modelling a specific working condition, are designed, proved, verified, implemented, tested and debugged independently. They are then composed, possibly dynamically, as black-boxes within the same global program. The AOC project is ambitious and it seeks to fundamentally revisit the way distributed algorithms are designed and distributed systems are implemented. The gain expected in comparison with today's approaches is substantial, and I believe it will be proportional to the degree of difficulty of the distributed problem at hand."
Summary
"Recent technological evolutions, including the cloud, the multicore, the social and the mobiles ones, are turning computing ubiquitously distributed. Yet, building high-assurance distributed programs is notoriously challenging. One of the main reasons is that these systems usually seek to achieve several goals at the same time. In short, they need to be efficient, responding effectively in various average-case conditions, as well as reliable, behaving correctly in severe, worst-case conditions. As a consequence, they typically intermingle different strategies: each to cope with some specific condition, e.g., with or without node failures, message losses, time-outs, contention, cache misses,
over-sizing, malicious attacks, etc. The resulting programs end up hard to design, prove, verify, implement, test and debug. Not surprisingly, there are anecdotal evidences of the fragility of the most celebrated distributed systems.
The goal of this project is to contribute to building high-assurance distributed programs by introducing a new dimension for separating and isolating their concerns, as well as a new scheme for composing and reusing them in a modular manner. In short, the project will explore the inherent power and limitations of a novel paradigm, Adversary-Oriented Computing (AOC). Sub-programs, each implementing a specific strategy to cope with a given adversary, modelling a specific working condition, are designed, proved, verified, implemented, tested and debugged independently. They are then composed, possibly dynamically, as black-boxes within the same global program. The AOC project is ambitious and it seeks to fundamentally revisit the way distributed algorithms are designed and distributed systems are implemented. The gain expected in comparison with today's approaches is substantial, and I believe it will be proportional to the degree of difficulty of the distributed problem at hand."
Max ERC Funding
2 147 012 €
Duration
Start date: 2014-06-01, End date: 2019-05-31
Project acronym APPROXNP
Project Approximation of NP-hard optimization problems
Researcher (PI) Johan Haastad
Host Institution (HI) KUNGLIGA TEKNISKA HOEGSKOLAN
Country Sweden
Call Details Advanced Grant (AdG), PE6, ERC-2008-AdG
Summary The proposed project aims to create a center of excellence that aims at understanding the approximability of NP-hard optimization problems. In particular, for central problems like vertex cover, coloring of graphs, and various constraint satisfaction problems we want to study upper and lower bounds on how well they can be approximated in polynomial time. Many existing strong results are based on what is known as the Unique Games Conjecture (UGC) and a significant part of the project will be devoted to studying this conjecture. We expect that a major step needed to be taken in this process is to further develop the understanding of Boolean functions on the Boolean hypercube. We anticipate that the tools needed for this will come in the form of harmonic analysis which in its turn will rely on the corresponding results in the analysis of functions over the domain of real numbers.
Summary
The proposed project aims to create a center of excellence that aims at understanding the approximability of NP-hard optimization problems. In particular, for central problems like vertex cover, coloring of graphs, and various constraint satisfaction problems we want to study upper and lower bounds on how well they can be approximated in polynomial time. Many existing strong results are based on what is known as the Unique Games Conjecture (UGC) and a significant part of the project will be devoted to studying this conjecture. We expect that a major step needed to be taken in this process is to further develop the understanding of Boolean functions on the Boolean hypercube. We anticipate that the tools needed for this will come in the form of harmonic analysis which in its turn will rely on the corresponding results in the analysis of functions over the domain of real numbers.
Max ERC Funding
2 376 000 €
Duration
Start date: 2009-01-01, End date: 2014-12-31
Project acronym astromnesis
Project The language of astrocytes: multilevel analysis to understand astrocyte communication and its role in memory-related brain operations and in cognitive behavior
Researcher (PI) Andrea Volterra
Host Institution (HI) UNIVERSITE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS5, ERC-2013-ADG
Summary In the 90s, two landmark observations brought to a paradigm shift about the role of astrocytes in brain function: 1) astrocytes respond to signals coming from other cells with transient Ca2+ elevations; 2) Ca2+ transients in astrocytes trigger release of neuroactive and vasoactive agents. Since then, many modulatory astrocytic actions and mechanisms were described, forming a complex - partly contradictory - picture, in which the exact roles and modes of astrocyte action remain ill defined. Our project wants to bring light into the “language of astrocytes”, i.e. into how they communicate with neurons and, ultimately, address their role in brain computations and cognitive behavior. To this end we will perform 4 complementary levels of analysis using highly innovative methodologies in order to obtain unprecedented results. We will study: 1) the subcellular organization of astrocytes underlying local microdomain communications by use of correlative light-electron microscopy; 2) the way individual astrocytes integrate inputs and control synaptic ensembles using 3D two-photon imaging, genetically-encoded Ca2+ indicators, optogenetics and electrophysiology; 3) the contribution of astrocyte ensembles to behavior-relevant circuit operations using miniaturized microscopes capturing neuronal/astrocytic population dynamics in freely-moving mice during memory tests; 4) the contribution of astrocytic signalling mechanisms to cognitive behavior using a set of new mouse lines with conditional, astrocyte-specific genetic modification of signalling pathways. We expect that this combination of groundbreaking ideas, innovative technologies and multilevel analysis makes our project highly attractive to the neuroscience community at large, bridging aspects of molecular, cellular, systems and behavioral neuroscience, with the goal of leading from a provocative hypothesis to the conclusive demonstration of whether and how “the language of astrocytes” participates in memory and cognition.
Summary
In the 90s, two landmark observations brought to a paradigm shift about the role of astrocytes in brain function: 1) astrocytes respond to signals coming from other cells with transient Ca2+ elevations; 2) Ca2+ transients in astrocytes trigger release of neuroactive and vasoactive agents. Since then, many modulatory astrocytic actions and mechanisms were described, forming a complex - partly contradictory - picture, in which the exact roles and modes of astrocyte action remain ill defined. Our project wants to bring light into the “language of astrocytes”, i.e. into how they communicate with neurons and, ultimately, address their role in brain computations and cognitive behavior. To this end we will perform 4 complementary levels of analysis using highly innovative methodologies in order to obtain unprecedented results. We will study: 1) the subcellular organization of astrocytes underlying local microdomain communications by use of correlative light-electron microscopy; 2) the way individual astrocytes integrate inputs and control synaptic ensembles using 3D two-photon imaging, genetically-encoded Ca2+ indicators, optogenetics and electrophysiology; 3) the contribution of astrocyte ensembles to behavior-relevant circuit operations using miniaturized microscopes capturing neuronal/astrocytic population dynamics in freely-moving mice during memory tests; 4) the contribution of astrocytic signalling mechanisms to cognitive behavior using a set of new mouse lines with conditional, astrocyte-specific genetic modification of signalling pathways. We expect that this combination of groundbreaking ideas, innovative technologies and multilevel analysis makes our project highly attractive to the neuroscience community at large, bridging aspects of molecular, cellular, systems and behavioral neuroscience, with the goal of leading from a provocative hypothesis to the conclusive demonstration of whether and how “the language of astrocytes” participates in memory and cognition.
Max ERC Funding
2 513 896 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym AtomicGaugeSimulator
Project Classical and Atomic Quantum Simulation of Gauge Theories in Particle and Condensed Matter Physics
Researcher (PI) Uwe-Jens Richard Christian Wiese
Host Institution (HI) UNIVERSITAET BERN
Country Switzerland
Call Details Advanced Grant (AdG), PE2, ERC-2013-ADG
Summary Gauge theories play a central role in particle and condensed matter physics. Heavy-ion collisions explore the strong dynamics of quarks and gluons, which also governs the deep interior of neutron stars, while strongly correlated electrons determine the physics of high-temperature superconductors and spin liquids. Numerical simulations of such systems are often hindered by sign problems. In quantum link models - an alternative formulation of gauge theories developed by the applicant - gauge fields emerge from discrete quantum variables. In the past year, in close collaboration with atomic physicists, we have established quantum link models as a framework for the atomic quantum simulation of dynamical gauge fields. Abelian gauge theories can be realized with Bose-Fermi mixtures of ultracold atoms in an optical lattice, while non-Abelian gauge fields arise from fermionic constituents embodied by alkaline-earth atoms. Quantum simulators, which do not suffer from the sign problem, shall be constructed to address non-trivial dynamics, including quantum phase transitions in spin liquids, the real-time dynamics of confining strings as well as of chiral symmetry restoration at finite temperature and baryon density, baryon superfluidity, or color-flavor locking. New classical simulation algorithms shall be developed in order to solve severe sign problems, to investigate confining gauge theories, and to validate the proposed quantum simulators. Starting from U(1) and SU(2) gauge theories, an atomic physics tool box shall be developed for quantum simulation of gauge theories of increasing complexity, ultimately aiming at 4-d Quantum Chromodynamics (QCD). This project is based on innovative ideas from particle, condensed matter, and computational physics, and requires an interdisciplinary team of researchers. It has the potential to drastically increase the power of simulations and to address very challenging problems that cannot be solved with classical simulation methods.
Summary
Gauge theories play a central role in particle and condensed matter physics. Heavy-ion collisions explore the strong dynamics of quarks and gluons, which also governs the deep interior of neutron stars, while strongly correlated electrons determine the physics of high-temperature superconductors and spin liquids. Numerical simulations of such systems are often hindered by sign problems. In quantum link models - an alternative formulation of gauge theories developed by the applicant - gauge fields emerge from discrete quantum variables. In the past year, in close collaboration with atomic physicists, we have established quantum link models as a framework for the atomic quantum simulation of dynamical gauge fields. Abelian gauge theories can be realized with Bose-Fermi mixtures of ultracold atoms in an optical lattice, while non-Abelian gauge fields arise from fermionic constituents embodied by alkaline-earth atoms. Quantum simulators, which do not suffer from the sign problem, shall be constructed to address non-trivial dynamics, including quantum phase transitions in spin liquids, the real-time dynamics of confining strings as well as of chiral symmetry restoration at finite temperature and baryon density, baryon superfluidity, or color-flavor locking. New classical simulation algorithms shall be developed in order to solve severe sign problems, to investigate confining gauge theories, and to validate the proposed quantum simulators. Starting from U(1) and SU(2) gauge theories, an atomic physics tool box shall be developed for quantum simulation of gauge theories of increasing complexity, ultimately aiming at 4-d Quantum Chromodynamics (QCD). This project is based on innovative ideas from particle, condensed matter, and computational physics, and requires an interdisciplinary team of researchers. It has the potential to drastically increase the power of simulations and to address very challenging problems that cannot be solved with classical simulation methods.
Max ERC Funding
1 975 242 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym Attoclock
Project Clocking fundamental attosecond electron dynamics
Researcher (PI) Ursula Keller
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Country Switzerland
Call Details Advanced Grant (AdG), PE2, ERC-2012-ADG_20120216
Summary The attoclock is a powerful, new, and unconventional tool to study fundamental attosecond dynamics on an atomic scale. We established its potential by using the first attoclock to measure the tunneling delay time in laser-induced ionization of helium and argon atoms, with surprising results. Building on these first proof-of-principle measurements, I propose to amplify and expand this tool concept to explore the following key questions: How fast can light liberate electrons from a single atom, a single molecule, or a solid-state system? Related are more questions: How fast can an electron tunnel through a potential barrier? How fast is a multi-photon absorption process? How fast is single-photon photoemission? Many of these questions will undoubtedly spark more questions – revealing deeper and more detailed insights on the dynamics of some of the most fundamental and relevant optoelectronic processes.
There are still many unknown and unexplored areas here. Theory has failed to offer definitive answers. Simulations based on the exact time-dependent Schrödinger equation have not been possible in most cases. Therefore one uses approximations and simpler models to capture the essential physics. Such semi-classical models potentially will help to understand attosecond energy and charge transport in larger molecular systems. Indeed the attoclock provides a unique tool to explore different semi-classical models.
For example, the question of whether electron tunneling through an energetically forbidden region takes a finite time or is instantaneous has been subject to ongoing debate for the last sixty years. The tunnelling process, charge transfer, and energy transport all play key roles in electronics, energy conversion, chemical and biological reactions, and fundamental processes important for improved information, health, and energy technologies. We believe the attoclock can help refine and resolve key models for many of these important underlying attosecond processes.
Summary
The attoclock is a powerful, new, and unconventional tool to study fundamental attosecond dynamics on an atomic scale. We established its potential by using the first attoclock to measure the tunneling delay time in laser-induced ionization of helium and argon atoms, with surprising results. Building on these first proof-of-principle measurements, I propose to amplify and expand this tool concept to explore the following key questions: How fast can light liberate electrons from a single atom, a single molecule, or a solid-state system? Related are more questions: How fast can an electron tunnel through a potential barrier? How fast is a multi-photon absorption process? How fast is single-photon photoemission? Many of these questions will undoubtedly spark more questions – revealing deeper and more detailed insights on the dynamics of some of the most fundamental and relevant optoelectronic processes.
There are still many unknown and unexplored areas here. Theory has failed to offer definitive answers. Simulations based on the exact time-dependent Schrödinger equation have not been possible in most cases. Therefore one uses approximations and simpler models to capture the essential physics. Such semi-classical models potentially will help to understand attosecond energy and charge transport in larger molecular systems. Indeed the attoclock provides a unique tool to explore different semi-classical models.
For example, the question of whether electron tunneling through an energetically forbidden region takes a finite time or is instantaneous has been subject to ongoing debate for the last sixty years. The tunnelling process, charge transfer, and energy transport all play key roles in electronics, energy conversion, chemical and biological reactions, and fundamental processes important for improved information, health, and energy technologies. We believe the attoclock can help refine and resolve key models for many of these important underlying attosecond processes.
Max ERC Funding
2 319 796 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym AXION
Project Axions: From Heaven to Earth
Researcher (PI) Frank Wilczek
Host Institution (HI) STOCKHOLMS UNIVERSITET
Country Sweden
Call Details Advanced Grant (AdG), PE2, ERC-2016-ADG
Summary Axions are hypothetical particles whose existence would solve two major problems: the strong P, T problem (a major blemish on the standard model); and the dark matter problem. It is a most important goal to either observe or rule out the existence of a cosmic axion background. It appears that decisive observations may be possible, but only after orchestrating insight from specialities ranging from quantum field theory and astrophysical modeling to ultra-low noise quantum measurement theory. Detailed predictions for the magnitude and structure of the cosmic axion background depend on cosmological and astrophysical modeling, which can be constrained by theoretical insight and numerical simulation. In parallel, we must optimize strategies for extracting accessible signals from that very weakly interacting source.
While the existence of axions as fundamental particles remains hypothetical, the equations governing how axions interact with electromagnetic fields also govern (with different parameters) how certain materials interact with electromagnetic fields. Thus those materials embody “emergent” axions. The equations have remarkable properties, which one can test in these materials, and possibly put to practical use.
Closely related to axions, mathematically, are anyons. Anyons are particle-like excitations that elude the familiar classification into bosons and fermions. Theoretical and numerical studies indicate that they are common emergent features of highly entangled states of matter in two dimensions. Recent work suggests the existence of states of matter, both natural and engineered, in which anyon dynamics is both important and experimentally accessible. Since the equations for anyons and axions are remarkably similar, and both have common, deep roots in symmetry and topology, it will be fruitful to consider them together.
Summary
Axions are hypothetical particles whose existence would solve two major problems: the strong P, T problem (a major blemish on the standard model); and the dark matter problem. It is a most important goal to either observe or rule out the existence of a cosmic axion background. It appears that decisive observations may be possible, but only after orchestrating insight from specialities ranging from quantum field theory and astrophysical modeling to ultra-low noise quantum measurement theory. Detailed predictions for the magnitude and structure of the cosmic axion background depend on cosmological and astrophysical modeling, which can be constrained by theoretical insight and numerical simulation. In parallel, we must optimize strategies for extracting accessible signals from that very weakly interacting source.
While the existence of axions as fundamental particles remains hypothetical, the equations governing how axions interact with electromagnetic fields also govern (with different parameters) how certain materials interact with electromagnetic fields. Thus those materials embody “emergent” axions. The equations have remarkable properties, which one can test in these materials, and possibly put to practical use.
Closely related to axions, mathematically, are anyons. Anyons are particle-like excitations that elude the familiar classification into bosons and fermions. Theoretical and numerical studies indicate that they are common emergent features of highly entangled states of matter in two dimensions. Recent work suggests the existence of states of matter, both natural and engineered, in which anyon dynamics is both important and experimentally accessible. Since the equations for anyons and axions are remarkably similar, and both have common, deep roots in symmetry and topology, it will be fruitful to consider them together.
Max ERC Funding
2 324 391 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym BACNK
Project Recognition of bacteria by NK cells
Researcher (PI) Ofer Mandelboim
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Country Israel
Call Details Advanced Grant (AdG), LS6, ERC-2012-ADG_20120314
Summary NK cells that are well known by their ability to recognize and eliminate virus infected and tumor cells were also implicated in the defence against bacteria. However, the recognition of bacteria by NK cells is only poorly understood. we do not know how bacteria are recognized and the functional consequences of such recognition are also weakly understood. In the current proposal we aimed at determining the “NK cell receptor-bacterial interactome”. We will examine the hypothesis that NK inhibitory and activating receptors are directly involved in bacterial recognition. This ground breaking hypothesis is based on our preliminary results in which we show that several NK cell receptors directly recognize various bacterial strains as well as on a few other publications. We will generate various mice knockouts for NCR1 (a major NK killer receptor) and determine their microbiota to understand the physiological function of NCR1 and whether certain bacterial strains affects its activity. We will use different human and mouse NK killer and inhibitory receptors fused to IgG1 to pull-down bacteria from saliva and fecal samples and then use 16S rRNA analysis and next generation sequencing to determine the nature of the bacteria species isolated. We will identify the bacterial ligands that are recognized by the relevant NK cell receptors, using bacterial random transposon insertion mutagenesis approach. We will end this research with functional assays. In the wake of the emerging threat of bacterial drug resistance and the involvement of bacteria in the pathogenesis of many different chronic diseases and in shaping the immune response, the completion of this study will open a new field of research; the direct recognition of bacteria by NK cell receptors.
Summary
NK cells that are well known by their ability to recognize and eliminate virus infected and tumor cells were also implicated in the defence against bacteria. However, the recognition of bacteria by NK cells is only poorly understood. we do not know how bacteria are recognized and the functional consequences of such recognition are also weakly understood. In the current proposal we aimed at determining the “NK cell receptor-bacterial interactome”. We will examine the hypothesis that NK inhibitory and activating receptors are directly involved in bacterial recognition. This ground breaking hypothesis is based on our preliminary results in which we show that several NK cell receptors directly recognize various bacterial strains as well as on a few other publications. We will generate various mice knockouts for NCR1 (a major NK killer receptor) and determine their microbiota to understand the physiological function of NCR1 and whether certain bacterial strains affects its activity. We will use different human and mouse NK killer and inhibitory receptors fused to IgG1 to pull-down bacteria from saliva and fecal samples and then use 16S rRNA analysis and next generation sequencing to determine the nature of the bacteria species isolated. We will identify the bacterial ligands that are recognized by the relevant NK cell receptors, using bacterial random transposon insertion mutagenesis approach. We will end this research with functional assays. In the wake of the emerging threat of bacterial drug resistance and the involvement of bacteria in the pathogenesis of many different chronic diseases and in shaping the immune response, the completion of this study will open a new field of research; the direct recognition of bacteria by NK cell receptors.
Max ERC Funding
2 499 800 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym BACTERIAL RESPONSE
Project New Concepts in Bacterial Response to their Surroundings
Researcher (PI) Sigal Ben-Yehuda
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Country Israel
Call Details Advanced Grant (AdG), LS6, ERC-2013-ADG
Summary Bacteria in nature exhibit remarkable capacity to sense their surroundings and rapidly adapt to diverse conditions by gaining new beneficial traits. This extraordinary feature facilitates their survival when facing extreme environments. Utilizing Bacillus subtilis as our primary model organism, we propose to study two facets of this vital bacterial attribute: communication via extracellular nanotubes, and persistence as resilient spores while maintaining the potential to revive. Exploring these fascinating aspects of bacterial physiology is likely to change our view as to how bacteria sense, respond, endure and communicate with their extracellular environment.
We have recently discovered a previously uncharacterized mode of bacterial communication, mediated by tubular extensions (nanotubes) that bridge neighboring cells, providing a route for exchange of intracellular molecules. Nanotube-mediated molecular sharing may represent a key form of bacterial communication in nature, allowing for the emergence of new phenotypes and increasing survival in fluctuating environments. Here we propose to develop strategies for observing nanotube formation and molecular exchange in living bacterial cells, and to characterize the molecular composition of nanotubes. We will explore the premise that nanotubes serve as a strategy to expand the cell surface, and will determine whether nanotubes provide a conduit for phage infection and spreading. Furthermore, the formation and functionality of interspecies nanotubes will be explored. An additional mode employed by bacteria to achieve extreme robustness is the ability to reside as long lasting spores. Previously held views considered the spore to be dormant and metabolically inert. However, we have recently shown that at least one week following spore formation, during an adaptive period, the spore senses and responds to environmental cues and undergoes corresponding molecular changes, influencing subsequent emergence from quiescence.
Summary
Bacteria in nature exhibit remarkable capacity to sense their surroundings and rapidly adapt to diverse conditions by gaining new beneficial traits. This extraordinary feature facilitates their survival when facing extreme environments. Utilizing Bacillus subtilis as our primary model organism, we propose to study two facets of this vital bacterial attribute: communication via extracellular nanotubes, and persistence as resilient spores while maintaining the potential to revive. Exploring these fascinating aspects of bacterial physiology is likely to change our view as to how bacteria sense, respond, endure and communicate with their extracellular environment.
We have recently discovered a previously uncharacterized mode of bacterial communication, mediated by tubular extensions (nanotubes) that bridge neighboring cells, providing a route for exchange of intracellular molecules. Nanotube-mediated molecular sharing may represent a key form of bacterial communication in nature, allowing for the emergence of new phenotypes and increasing survival in fluctuating environments. Here we propose to develop strategies for observing nanotube formation and molecular exchange in living bacterial cells, and to characterize the molecular composition of nanotubes. We will explore the premise that nanotubes serve as a strategy to expand the cell surface, and will determine whether nanotubes provide a conduit for phage infection and spreading. Furthermore, the formation and functionality of interspecies nanotubes will be explored. An additional mode employed by bacteria to achieve extreme robustness is the ability to reside as long lasting spores. Previously held views considered the spore to be dormant and metabolically inert. However, we have recently shown that at least one week following spore formation, during an adaptive period, the spore senses and responds to environmental cues and undergoes corresponding molecular changes, influencing subsequent emergence from quiescence.
Max ERC Funding
1 497 800 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym BARRAGE
Project Cell compartmentalization, individuation and diversity
Researcher (PI) Yves Barral
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Country Switzerland
Call Details Advanced Grant (AdG), LS3, ERC-2009-AdG
Summary Asymmetric cell division is a key mechanism for the generation of cell diversity in eukaryotes. During this process, a polarized mother cell divides into non-equivalent daughters. These may differentially inherit fate determinants, irreparable damages or age determinants. Our aim is to decipher the mechanisms governing the individualization of daughters from each other. In the past ten years, our studies identified several lateral diffusion barriers located in the plasma membrane and the endoplasmic reticulum of budding yeast. These barriers all restrict molecular exchanges between the mother cell and its bud, and thereby compartmentalize the cell already long before its division. They play key roles in the asymmetric segregation of various factors. On one side, they help maintain polarized factors into the bud. Thereby, they reinforce cell polarity and sequester daughter-specific fate determinants into the bud. On the other side they prevent aging factors of the mother from entering the bud. Hence, they play key roles in the rejuvenation of the bud, in the aging of the mother, and in the differentiation of mother and daughter from each other. Recently, we accumulated evidence that some of these barriers are subject to regulation, such as to help modulate the longevity of the mother cell in response to environmental signals. Our data also suggest that barriers help the mother cell keep traces of its life history, thereby contributing to its individuation and adaption to the environment. In this project, we will address the following questions: 1 How are these barriers assembled, functioning, and regulated? 2 What type of differentiation processes are they involved in? 3 Are they conserved in other eukaryotes, and what are their functions outside of budding yeast? These studies will shed light into the principles underlying and linking aging, rejuvenation and differentiation.
Summary
Asymmetric cell division is a key mechanism for the generation of cell diversity in eukaryotes. During this process, a polarized mother cell divides into non-equivalent daughters. These may differentially inherit fate determinants, irreparable damages or age determinants. Our aim is to decipher the mechanisms governing the individualization of daughters from each other. In the past ten years, our studies identified several lateral diffusion barriers located in the plasma membrane and the endoplasmic reticulum of budding yeast. These barriers all restrict molecular exchanges between the mother cell and its bud, and thereby compartmentalize the cell already long before its division. They play key roles in the asymmetric segregation of various factors. On one side, they help maintain polarized factors into the bud. Thereby, they reinforce cell polarity and sequester daughter-specific fate determinants into the bud. On the other side they prevent aging factors of the mother from entering the bud. Hence, they play key roles in the rejuvenation of the bud, in the aging of the mother, and in the differentiation of mother and daughter from each other. Recently, we accumulated evidence that some of these barriers are subject to regulation, such as to help modulate the longevity of the mother cell in response to environmental signals. Our data also suggest that barriers help the mother cell keep traces of its life history, thereby contributing to its individuation and adaption to the environment. In this project, we will address the following questions: 1 How are these barriers assembled, functioning, and regulated? 2 What type of differentiation processes are they involved in? 3 Are they conserved in other eukaryotes, and what are their functions outside of budding yeast? These studies will shed light into the principles underlying and linking aging, rejuvenation and differentiation.
Max ERC Funding
2 200 000 €
Duration
Start date: 2010-05-01, End date: 2015-04-30
Project acronym BATESON
Project Dissecting genotype-phenotype relationships using high-throughput genomics and carefully selected study populations
Researcher (PI) Leif Andersson
Host Institution (HI) UPPSALA UNIVERSITET
Country Sweden
Call Details Advanced Grant (AdG), LS2, ERC-2011-ADG_20110310
Summary A major aim in genome research is to reveal how genetic variation affects phenotypic variation. Here I propose to use high-throughput genomics (whole genome sequencing, transcriptome and epigenome analysis) to screen carefully selected study populations where the chances are particularly favourable to obtain novel insight into genotype-phenotype relationships. The ambition is to take discoveries all the way from phenotypic characterization to the identification of the genes and the actual genetic variant causing a phenotypic effect and to understanding the underlying functional mechanisms. The program will involve a fish (the Atlantic herring), a bird (the domestic chicken) and a mammal (the European rabbit). The Atlantic herring will be studied because it provides unique opportunities to study the genetics of adaptation in a natural population and because of the possibilities to revolutionize the fishery management of this economically important marine fish. We will generate a draft assembly of the herring genome and then perform whole genome resequencing of different populations to reveal the population structure and the loci underlying genetic adaptation. The European rabbit is an excellent model for studying the genetics of speciation due to the presence of two distinct subspecies on the Iberian Peninsula. The domestication of the rabbit is also particularly interesting because it is a recent event (about 1500 years ago) and it is well established that domestication happened from the wild rabbit population in southern France. Finally, the domestic chicken provides excellent opportunities for in depth functional studies since it is both a domestic animal harbouring a rich genetic diversity and an experimental organism.
(BATESON is the acronym for this proposal because Bateson (1902) pioneered the study of genotype-phenotype relationships in animals and used the chicken for this work.)
Summary
A major aim in genome research is to reveal how genetic variation affects phenotypic variation. Here I propose to use high-throughput genomics (whole genome sequencing, transcriptome and epigenome analysis) to screen carefully selected study populations where the chances are particularly favourable to obtain novel insight into genotype-phenotype relationships. The ambition is to take discoveries all the way from phenotypic characterization to the identification of the genes and the actual genetic variant causing a phenotypic effect and to understanding the underlying functional mechanisms. The program will involve a fish (the Atlantic herring), a bird (the domestic chicken) and a mammal (the European rabbit). The Atlantic herring will be studied because it provides unique opportunities to study the genetics of adaptation in a natural population and because of the possibilities to revolutionize the fishery management of this economically important marine fish. We will generate a draft assembly of the herring genome and then perform whole genome resequencing of different populations to reveal the population structure and the loci underlying genetic adaptation. The European rabbit is an excellent model for studying the genetics of speciation due to the presence of two distinct subspecies on the Iberian Peninsula. The domestication of the rabbit is also particularly interesting because it is a recent event (about 1500 years ago) and it is well established that domestication happened from the wild rabbit population in southern France. Finally, the domestic chicken provides excellent opportunities for in depth functional studies since it is both a domestic animal harbouring a rich genetic diversity and an experimental organism.
(BATESON is the acronym for this proposal because Bateson (1902) pioneered the study of genotype-phenotype relationships in animals and used the chicken for this work.)
Max ERC Funding
2 300 000 €
Duration
Start date: 2012-05-01, End date: 2017-04-30
Project acronym BBBARRIER
Project Mechanisms of regulation of the blood-brain barrier; towards opening and closing the barrier on demand
Researcher (PI) Bjoern Christer Betsholtz
Host Institution (HI) UPPSALA UNIVERSITET
Country Sweden
Call Details Advanced Grant (AdG), LS4, ERC-2011-ADG_20110310
Summary In the bone-enclosed CNS, increased vascular permeability may cause life-threatening tissue swelling, and/or ischemia and inflammation which compromise tissue repair after trauma or stroke. The brain vasculature possesses several unique features collectively named the blood-brain barrier (BBB) in which passive permeability is almost completely abolished and replaced by a complex of specific transport mechanisms. The BBB is necessary to uphold the specific milieu necessary for neuronal function. Whereas breakdown of the BBB is part of many CNS diseases, including stroke, neuroinflammation, trauma and neurodegenerative disorders, its molecular mechanisms and consequences are unclear and debated. Conversely, the intact BBB is a huge obstacle for drug delivery to the brain. Research on the BBB therefore has two seemingly opposing aims: 1) to seal a damaged BBB and protect the brain from toxic blood products, and 2) to open the BBB “on demand” for drug delivery. A major problem in the BBB field has been the lack of in vivo animal models for molecular and functional studies. So far, available in vitro models are not recapitulating the in vivo BBB. Our recent work on mouse models lacking pericytes, a BBB-associated cell type, demonstrates a specific role for pericytes in the development and regulation of the mammalian BBB. These animal models are the first ones showing a general and significant BBB impairment in adulthood, and as such they provide a unique opportunity to address molecular mechanisms of BBB disruption in disease and in drug transport across the BBB. Importantly, the new models and tools that we have developed allow us to search for relevant druggable mechanisms and molecular targets in the BBB. The long-term goals of this proposal are to develop molecular strategies and tools to open and close the BBB “on demand” for drug delivery to the CNS, and to explore the importance and mechanisms of BBB dysfunction in neurodegenerative diseases and stroke.
Summary
In the bone-enclosed CNS, increased vascular permeability may cause life-threatening tissue swelling, and/or ischemia and inflammation which compromise tissue repair after trauma or stroke. The brain vasculature possesses several unique features collectively named the blood-brain barrier (BBB) in which passive permeability is almost completely abolished and replaced by a complex of specific transport mechanisms. The BBB is necessary to uphold the specific milieu necessary for neuronal function. Whereas breakdown of the BBB is part of many CNS diseases, including stroke, neuroinflammation, trauma and neurodegenerative disorders, its molecular mechanisms and consequences are unclear and debated. Conversely, the intact BBB is a huge obstacle for drug delivery to the brain. Research on the BBB therefore has two seemingly opposing aims: 1) to seal a damaged BBB and protect the brain from toxic blood products, and 2) to open the BBB “on demand” for drug delivery. A major problem in the BBB field has been the lack of in vivo animal models for molecular and functional studies. So far, available in vitro models are not recapitulating the in vivo BBB. Our recent work on mouse models lacking pericytes, a BBB-associated cell type, demonstrates a specific role for pericytes in the development and regulation of the mammalian BBB. These animal models are the first ones showing a general and significant BBB impairment in adulthood, and as such they provide a unique opportunity to address molecular mechanisms of BBB disruption in disease and in drug transport across the BBB. Importantly, the new models and tools that we have developed allow us to search for relevant druggable mechanisms and molecular targets in the BBB. The long-term goals of this proposal are to develop molecular strategies and tools to open and close the BBB “on demand” for drug delivery to the CNS, and to explore the importance and mechanisms of BBB dysfunction in neurodegenerative diseases and stroke.
Max ERC Funding
2 499 427 €
Duration
Start date: 2012-08-01, End date: 2017-07-31
Project acronym BETAIMAGE
Project An in vivo imaging approach to understand pancreatic beta-cell signal-transduction
Researcher (PI) Per-Olof Berggren
Host Institution (HI) KAROLINSKA INSTITUTET
Country Sweden
Call Details Advanced Grant (AdG), LS4, ERC-2013-ADG
Summary The challenge in cell physiology/pathology today is to translate in vitro findings to the living organism. We have developed a unique approach where signal-transduction can be investigated in vivo non-invasively, longitudinally at single cell resolution, using the anterior chamber of the eye as a natural body window for imaging. We will use this approach to understand how the universally important and highly complex signal Ca2+ is regulated in the pancreatic beta-cell, while localized in the vascularized and innervated islet of Langerhans, and how that affects the insulin secretory machinery in vivo. Engrafted islets in the eye take on identical innervation- and vascularization patterns as those in the pancreas and are proficient in regulating glucose homeostasis in the animal. Since the pancreatic islet constitutes a micro-organ, this imaging approach offers a seminal model system to understand Ca2+ signaling in individual cells at the organ level in real life. We will test the hypothesis that the Ca2+-signal has a key role in pancreatic beta-cell function and survival in vivo and that perturbation in the Ca2+-signal serves as a common denominator for beta-cell pathology associated with impaired glucose homeostasis and diabetes. Of special interest is how innervation impacts on Ca2+-dynamics and the integration of autocrine, paracrine and endocrine signals in fine-tuning the Ca2+-signal with regard to beta-cell function and survival. We aim to define key defects in the machinery regulating Ca2+-dynamics in association with the autoimmune reaction, inflammation and obesity eventually resulting in diabetes. Our imaging platform will be applied to clarify in vivo regulation of Ca2+-dynamics in both healthy and diabetic human beta-cells. To define novel drugable targets for treatment of diabetes, it is crucial to identify similarities and differences in the molecular machinery regulating the in vivo Ca2+-signal in the human and in the rodent beta-cell.
Summary
The challenge in cell physiology/pathology today is to translate in vitro findings to the living organism. We have developed a unique approach where signal-transduction can be investigated in vivo non-invasively, longitudinally at single cell resolution, using the anterior chamber of the eye as a natural body window for imaging. We will use this approach to understand how the universally important and highly complex signal Ca2+ is regulated in the pancreatic beta-cell, while localized in the vascularized and innervated islet of Langerhans, and how that affects the insulin secretory machinery in vivo. Engrafted islets in the eye take on identical innervation- and vascularization patterns as those in the pancreas and are proficient in regulating glucose homeostasis in the animal. Since the pancreatic islet constitutes a micro-organ, this imaging approach offers a seminal model system to understand Ca2+ signaling in individual cells at the organ level in real life. We will test the hypothesis that the Ca2+-signal has a key role in pancreatic beta-cell function and survival in vivo and that perturbation in the Ca2+-signal serves as a common denominator for beta-cell pathology associated with impaired glucose homeostasis and diabetes. Of special interest is how innervation impacts on Ca2+-dynamics and the integration of autocrine, paracrine and endocrine signals in fine-tuning the Ca2+-signal with regard to beta-cell function and survival. We aim to define key defects in the machinery regulating Ca2+-dynamics in association with the autoimmune reaction, inflammation and obesity eventually resulting in diabetes. Our imaging platform will be applied to clarify in vivo regulation of Ca2+-dynamics in both healthy and diabetic human beta-cells. To define novel drugable targets for treatment of diabetes, it is crucial to identify similarities and differences in the molecular machinery regulating the in vivo Ca2+-signal in the human and in the rodent beta-cell.
Max ERC Funding
2 499 590 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym BIGGER
Project Biophysics in gene regulation - A genome wide approach
Researcher (PI) Johan Elf
Host Institution (HI) UPPSALA UNIVERSITET
Country Sweden
Call Details Advanced Grant (AdG), LS2, ERC-2019-ADG
Summary In this project, we will develop and use technology that combines synthetic genomics and live-cell imaging. These methods make it possible to study the intracellular biophysics at single-molecule detail in thousands of genetically different bacterial strains in parallel. Our approach is based on in situ genotyping of a barcoded strain library after phenotyping has been performed by live-cell imaging. Within the scope of the proposed project, the new technology will be used to solve mechanistic and structural questions of the bacterial cell cycle.
To this end, we will explore two parallel but complementary applications. In the first application, we will determine the dynamic 3D structure of the E. coli chromosome at 1kb resolution throughout the cell cycle. The structure determination can be seen as a live-cell version of chromatin conformation capture, where we will follow the 3D distances of 10 000 pairs of chromosomal loci over the cell cycle at high resolution. In the second application, we will make a complete CRISPRi knockdown strain library where we can follow the replication forks of the E. coli chromosome and septum formation over the cell cycle in individual cells. Using this strategy, we will resolve how individual gene products contribute to the cell-to-cell accuracy in replication initiation and cell division. In particular, this approach allows us to address the challenging question of size sensing at replication initiation. How the cell can decide that it is large enough to initiate replication is still an open question despite decades of investigations.
The general principles for high-end imaging of pool-synthesized cell libraries have nearly unlimited applications throughout cell biology. The specific applications explored in this project will take the understanding of the bacterial cell cycle to a new level and answer general questions about the chromosomal organization and cell size sensing.
Summary
In this project, we will develop and use technology that combines synthetic genomics and live-cell imaging. These methods make it possible to study the intracellular biophysics at single-molecule detail in thousands of genetically different bacterial strains in parallel. Our approach is based on in situ genotyping of a barcoded strain library after phenotyping has been performed by live-cell imaging. Within the scope of the proposed project, the new technology will be used to solve mechanistic and structural questions of the bacterial cell cycle.
To this end, we will explore two parallel but complementary applications. In the first application, we will determine the dynamic 3D structure of the E. coli chromosome at 1kb resolution throughout the cell cycle. The structure determination can be seen as a live-cell version of chromatin conformation capture, where we will follow the 3D distances of 10 000 pairs of chromosomal loci over the cell cycle at high resolution. In the second application, we will make a complete CRISPRi knockdown strain library where we can follow the replication forks of the E. coli chromosome and septum formation over the cell cycle in individual cells. Using this strategy, we will resolve how individual gene products contribute to the cell-to-cell accuracy in replication initiation and cell division. In particular, this approach allows us to address the challenging question of size sensing at replication initiation. How the cell can decide that it is large enough to initiate replication is still an open question despite decades of investigations.
The general principles for high-end imaging of pool-synthesized cell libraries have nearly unlimited applications throughout cell biology. The specific applications explored in this project will take the understanding of the bacterial cell cycle to a new level and answer general questions about the chromosomal organization and cell size sensing.
Max ERC Funding
2 411 410 €
Duration
Start date: 2021-01-01, End date: 2025-12-31
Project acronym BiomeRiskFactors
Project Discovering microbiome-based disease risk factors
Researcher (PI) Eran Segal
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Country Israel
Call Details Advanced Grant (AdG), LS2, ERC-2017-ADG
Summary Identifying risk factors for diseases that can be prevented or delayed by early intervention is of major importance, and numerous genetic, lifestyle, anthropometric and clinical risk factors were found for many different diseases. Another source of potentially pertinent disease risk factors is the human microbiome - the collective genome of trillions of bacteria, viruses, fungi, and parasites that reside in the human gut. However, very few microbiome disease markers were found to date.
Here, we aim to develop risk prediction tools based on the human microbiome that predict the likelihood of an individual to develop a particular condition or disease within 5-10 years. We will use a cohort of >2200 individuals that my group previously assembled, for whom we have clinical profiles, gut microbiome data, and banked blood and stool samples. We will invite people 5-10 years after their initial recruitment time, profile disease status and blood markers, and develop algorithms for predicting 5-10 year onset of Type 2 diabetes, cardiovascular disease, and obesity, using microbiome data from recruitment time.
To increase the likelihood of finding microbiome markers predictive of disease onset, we will develop novel experimental and computational methods for in-depth characterization of microbial gene function, the metabolites produced by the microbiome, the underexplored fungal microbiome members, and the interactions between the gut microbiota and the host adaptive immune system. We will then apply these methods to >2200 banked samples from cohort recruitment time and use the resulting data in devising our microbiome-based risk prediction tools. In themselves, these novel assays and their application to >2200 samples should greatly advance the microbiome field.
If successful, our proposal will identify new disease risk factors and risk prediction tools based on the microbiome, paving the way towards using the microbiome in early disease detection and prevention.
Summary
Identifying risk factors for diseases that can be prevented or delayed by early intervention is of major importance, and numerous genetic, lifestyle, anthropometric and clinical risk factors were found for many different diseases. Another source of potentially pertinent disease risk factors is the human microbiome - the collective genome of trillions of bacteria, viruses, fungi, and parasites that reside in the human gut. However, very few microbiome disease markers were found to date.
Here, we aim to develop risk prediction tools based on the human microbiome that predict the likelihood of an individual to develop a particular condition or disease within 5-10 years. We will use a cohort of >2200 individuals that my group previously assembled, for whom we have clinical profiles, gut microbiome data, and banked blood and stool samples. We will invite people 5-10 years after their initial recruitment time, profile disease status and blood markers, and develop algorithms for predicting 5-10 year onset of Type 2 diabetes, cardiovascular disease, and obesity, using microbiome data from recruitment time.
To increase the likelihood of finding microbiome markers predictive of disease onset, we will develop novel experimental and computational methods for in-depth characterization of microbial gene function, the metabolites produced by the microbiome, the underexplored fungal microbiome members, and the interactions between the gut microbiota and the host adaptive immune system. We will then apply these methods to >2200 banked samples from cohort recruitment time and use the resulting data in devising our microbiome-based risk prediction tools. In themselves, these novel assays and their application to >2200 samples should greatly advance the microbiome field.
If successful, our proposal will identify new disease risk factors and risk prediction tools based on the microbiome, paving the way towards using the microbiome in early disease detection and prevention.
Max ERC Funding
2 500 000 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym BIOMOLECULAR_COMP
Project Biomolecular computers
Researcher (PI) Ehud Shapiro
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Country Israel
Call Details Advanced Grant (AdG), LS9, ERC-2008-AdG
Summary Autonomous programmable computing devices made of biological molecules hold the promise of interacting with the biological environment in future biological and medical applications. Our laboratory's long-term objective is to develop a 'Doctor in a cell': molecular-sized device that can roam the body, equipped with medical knowledge. It would diagnose a disease by analyzing the data available in its biochemical environment based on the encoded medical knowledge and treat it by releasing the appropriate drug molecule in situ. This kind of device might, in the future, be delivered to all cells in a specific tissue, organ or the whole organism, and cure or kill only those cells diagnosed with a disease. Our laboratory embarked on the attempt to design and build these molecular computing devices and lay the foundation for their future biomedical applications. Several important milestones have already been accomplished towards the realization of the Doctor in a cell vision. The subject of this proposal is a construction of autonomous biomolecular computers that could be delivered into a living cell, interact with endogenous biomolecules that are known to indicate diseases, logically analyze them, make a diagnostic decision and couple it to the production of an active biomolecule capable of influencing cell fate.
Summary
Autonomous programmable computing devices made of biological molecules hold the promise of interacting with the biological environment in future biological and medical applications. Our laboratory's long-term objective is to develop a 'Doctor in a cell': molecular-sized device that can roam the body, equipped with medical knowledge. It would diagnose a disease by analyzing the data available in its biochemical environment based on the encoded medical knowledge and treat it by releasing the appropriate drug molecule in situ. This kind of device might, in the future, be delivered to all cells in a specific tissue, organ or the whole organism, and cure or kill only those cells diagnosed with a disease. Our laboratory embarked on the attempt to design and build these molecular computing devices and lay the foundation for their future biomedical applications. Several important milestones have already been accomplished towards the realization of the Doctor in a cell vision. The subject of this proposal is a construction of autonomous biomolecular computers that could be delivered into a living cell, interact with endogenous biomolecules that are known to indicate diseases, logically analyze them, make a diagnostic decision and couple it to the production of an active biomolecule capable of influencing cell fate.
Max ERC Funding
2 125 980 €
Duration
Start date: 2009-01-01, End date: 2013-10-31
Project acronym BIRD
Project Bimanual Manipulation of Rigid and Deformable Objects
Researcher (PI) Danica KRAGIC JENSFELT
Host Institution (HI) KUNGLIGA TEKNISKA HOEGSKOLAN
Country Sweden
Call Details Advanced Grant (AdG), PE7, ERC-2019-ADG
Summary All day long, our fingers touch, grasp and move objects in various media such as air, water, oil. We do this almost effortlessly - it feels like we do not spend time planning and reflecting over what our hands and fingers do or how the continuous integration of various sensory modalities such as vision, touch, proprioception, hearing help us to outperform any other biological system in the variety of the interaction tasks that we can execute. Largely overlooked, and perhaps most fascinating is the ease with which we perform these interactions resulting in a belief that these are also easy to accomplish in artificial systems such as robots. However, there are still no robots that can easily hand-wash dishes, button a shirt or peel a potato. Our claim is that this is fundamentally a problem of appropriate representation or parameterization. When interacting with objects, the robot needs to consider geometric, topological, and physical properties of objects. This can be done either explicitly, by modeling and representing these properties, or implicitly, by learning them from data. The main scientific objective of this project is to create new informative and compact representations of deformable objects that incorporate both analytical and learning-based approaches and encode geometric, topological, and physical information about the robot, the object, and the environment. We will do this in the context of challenging multimodal, bimanual object interaction tasks. The focus will be on physical interaction with deformable objects using multimodal feedback. To meet these objectives, we will use theoretical and computational methods together with rigorous experimental evaluation to model skilled sensorimotor behavior in bimanual robot systems.
Summary
All day long, our fingers touch, grasp and move objects in various media such as air, water, oil. We do this almost effortlessly - it feels like we do not spend time planning and reflecting over what our hands and fingers do or how the continuous integration of various sensory modalities such as vision, touch, proprioception, hearing help us to outperform any other biological system in the variety of the interaction tasks that we can execute. Largely overlooked, and perhaps most fascinating is the ease with which we perform these interactions resulting in a belief that these are also easy to accomplish in artificial systems such as robots. However, there are still no robots that can easily hand-wash dishes, button a shirt or peel a potato. Our claim is that this is fundamentally a problem of appropriate representation or parameterization. When interacting with objects, the robot needs to consider geometric, topological, and physical properties of objects. This can be done either explicitly, by modeling and representing these properties, or implicitly, by learning them from data. The main scientific objective of this project is to create new informative and compact representations of deformable objects that incorporate both analytical and learning-based approaches and encode geometric, topological, and physical information about the robot, the object, and the environment. We will do this in the context of challenging multimodal, bimanual object interaction tasks. The focus will be on physical interaction with deformable objects using multimodal feedback. To meet these objectives, we will use theoretical and computational methods together with rigorous experimental evaluation to model skilled sensorimotor behavior in bimanual robot systems.
Max ERC Funding
2 424 186 €
Duration
Start date: 2020-09-01, End date: 2025-08-31
Project acronym BISON
Project Bio-Inspired Self-Assembled Supramolecular Organic Nanostructures
Researcher (PI) Ehud Gazit
Host Institution (HI) TEL AVIV UNIVERSITY
Country Israel
Call Details Advanced Grant (AdG), LS9, ERC-2015-AdG
Summary Peptide building blocks serve as very attractive bio-inspired elements in nanotechnology owing to their controlled self-assembly, inherent biocompatibility, chemical versatility, biological recognition abilities and facile synthesis. We have demonstrated the ability of remarkably simple aromatic peptides to form well-ordered nanostructures of exceptional physical properties. By taking inspiration from the minimal recognition modules used by nature to mediate coordinated processes of self-assembly, we have developed building blocks that form well-ordered nanostructures. The compact design of the building blocks, and therefore, the unique structural organization, resulted in metallic-like Young's modulus, blue luminescence due to quantum confinement, and notable piezoelectric properties. The goal of this proposal is to develop two new fronts for bio-inspired building block repertoire along with co-assembly to provide new avenues for organic nanotechnology. This will combine our vast experience in the assembly of aromatic peptides together with additional structural modules from nature. The new entities will be developed by exploiting the design principles of small aromatic building blocks to arrive at the smallest possible module that form super helical assembly based on the coiled coil motifs and establishing peptide nucleic acids based systems to combine the worlds of peptide and DNA nanotechnologies. The proposed research will combine extensive design and synthesis effort to provide a very diverse collection of novel buildings blocks and determination of their self-assembly process, followed by broad chemical, physical, and biological characterization of the nanostructures. Furthermore, effort will be made to establish supramolecular co-polymer systems to extend the morphological control of the assembly process. The result of the project will be a large and defined collection of novel chemical entities that will help reshape the field of bioorganic nanotechnology.
Summary
Peptide building blocks serve as very attractive bio-inspired elements in nanotechnology owing to their controlled self-assembly, inherent biocompatibility, chemical versatility, biological recognition abilities and facile synthesis. We have demonstrated the ability of remarkably simple aromatic peptides to form well-ordered nanostructures of exceptional physical properties. By taking inspiration from the minimal recognition modules used by nature to mediate coordinated processes of self-assembly, we have developed building blocks that form well-ordered nanostructures. The compact design of the building blocks, and therefore, the unique structural organization, resulted in metallic-like Young's modulus, blue luminescence due to quantum confinement, and notable piezoelectric properties. The goal of this proposal is to develop two new fronts for bio-inspired building block repertoire along with co-assembly to provide new avenues for organic nanotechnology. This will combine our vast experience in the assembly of aromatic peptides together with additional structural modules from nature. The new entities will be developed by exploiting the design principles of small aromatic building blocks to arrive at the smallest possible module that form super helical assembly based on the coiled coil motifs and establishing peptide nucleic acids based systems to combine the worlds of peptide and DNA nanotechnologies. The proposed research will combine extensive design and synthesis effort to provide a very diverse collection of novel buildings blocks and determination of their self-assembly process, followed by broad chemical, physical, and biological characterization of the nanostructures. Furthermore, effort will be made to establish supramolecular co-polymer systems to extend the morphological control of the assembly process. The result of the project will be a large and defined collection of novel chemical entities that will help reshape the field of bioorganic nanotechnology.
Max ERC Funding
3 003 125 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
Project acronym BONE SCAN
Project Traces in the bones: reconstructing the lost soft anatomy of the earliest vertebrates through ultra-high resolution synchrotron scanning
Researcher (PI) Per Erik Ahlberg
Host Institution (HI) UPPSALA UNIVERSITET
Country Sweden
Call Details Advanced Grant (AdG), LS8, ERC-2008-AdG
Summary Early vertebrate evolution involved a series of drastic structural reorganisations as new features were added and elaborated. The fossil record illuminates this evolutionary history more directly than inferences from the diversity of living forms, but the fossils usually consist only of bones whereas many of the most important and interesting changes occurred in the soft anatomy. Traditional approaches to reconstructing the musculature and other soft tissues of fossil vertebrates rely on subjective tools, like the visual identification of rough bone textures thought to indicate muscle attachments, and generally leave a lot to be desired. Here I propose a wholly novel and radically more objective approach to the identification of soft-tissue contacts, using holotomographic synchrotron CT at sub-micron resolutions to identify these contacts by the three-dimensional micro-architecture of the bone. A pilot study has already shown that such scans (performed at the ESRF synchrotron facility in Grenoble) are capable of imaging key features such as arrested growth surfaces and probable Sharpey s fibres in 380 million year old fossils. We will undertake a systematic review of the three-dimensional bone micro-architectures associated with different soft-tissue contacts in living vertebrates, and the use this as a key to reconstruct the soft-tissue contacts on fossil bones with unprecedented accuracy. This will permit us to produce far more reliable reconstructions of the soft anatomy than has hitherto been possible. Our findings will inform other areas of palaentology, particularly functional morphology, and will also be of great importance to evolutionary developmental biology.
Summary
Early vertebrate evolution involved a series of drastic structural reorganisations as new features were added and elaborated. The fossil record illuminates this evolutionary history more directly than inferences from the diversity of living forms, but the fossils usually consist only of bones whereas many of the most important and interesting changes occurred in the soft anatomy. Traditional approaches to reconstructing the musculature and other soft tissues of fossil vertebrates rely on subjective tools, like the visual identification of rough bone textures thought to indicate muscle attachments, and generally leave a lot to be desired. Here I propose a wholly novel and radically more objective approach to the identification of soft-tissue contacts, using holotomographic synchrotron CT at sub-micron resolutions to identify these contacts by the three-dimensional micro-architecture of the bone. A pilot study has already shown that such scans (performed at the ESRF synchrotron facility in Grenoble) are capable of imaging key features such as arrested growth surfaces and probable Sharpey s fibres in 380 million year old fossils. We will undertake a systematic review of the three-dimensional bone micro-architectures associated with different soft-tissue contacts in living vertebrates, and the use this as a key to reconstruct the soft-tissue contacts on fossil bones with unprecedented accuracy. This will permit us to produce far more reliable reconstructions of the soft anatomy than has hitherto been possible. Our findings will inform other areas of palaentology, particularly functional morphology, and will also be of great importance to evolutionary developmental biology.
Max ERC Funding
1 046 782 €
Duration
Start date: 2009-04-01, End date: 2014-03-31
Project acronym BOTMED
Project Microrobotics and Nanomedicine
Researcher (PI) Bradley James Nelson
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Country Switzerland
Call Details Advanced Grant (AdG), PE7, ERC-2010-AdG_20100224
Summary The introduction of minimally invasive surgery in the 1980’s created a paradigm shift in surgical procedures. Health care is now in a position to make a more dramatic leap by integrating newly developed wireless microrobotic technologies with nanomedicine to perform precisely targeted, localized endoluminal techniques. Devices capable of entering the human body through natural orifices or small incisions to deliver drugs, perform diagnostic procedures, and excise and repair tissue will be used. These new procedures will result in less trauma to the patient and faster recovery times, and will enable new therapies that have not yet been conceived. In order to realize this, many new technologies must be developed and synergistically integrated, and medical therapies for which the technology will prove successful must be aggressively pursued.
This proposed project will result in the realization of animal trials in which wireless microrobotic devices will be used to investigate a variety of extremely delicate ophthalmic therapies. The therapies to be pursued include the delivery of tissue plasminogen activator (t-PA) to blocked retinal veins, the peeling of epiretinal membranes from the retina, and the development of diagnostic procedures based on mapping oxygen concentration at the vitreous-retina interface. With successful animal trials, a path to human trials and commercialization will follow. Clearly, many systems in the body have the potential to benefit from the endoluminal technologies that this project considers, including the digestive system, the circulatory system, the urinary system, the central nervous system, the respiratory system, the female reproductive system and even the fetus. Microrobotic retinal therapies will greatly illuminate the potential that the integration of microrobotics and nanomedicine holds for society, and greatly accelerate this trend in Europe.
Summary
The introduction of minimally invasive surgery in the 1980’s created a paradigm shift in surgical procedures. Health care is now in a position to make a more dramatic leap by integrating newly developed wireless microrobotic technologies with nanomedicine to perform precisely targeted, localized endoluminal techniques. Devices capable of entering the human body through natural orifices or small incisions to deliver drugs, perform diagnostic procedures, and excise and repair tissue will be used. These new procedures will result in less trauma to the patient and faster recovery times, and will enable new therapies that have not yet been conceived. In order to realize this, many new technologies must be developed and synergistically integrated, and medical therapies for which the technology will prove successful must be aggressively pursued.
This proposed project will result in the realization of animal trials in which wireless microrobotic devices will be used to investigate a variety of extremely delicate ophthalmic therapies. The therapies to be pursued include the delivery of tissue plasminogen activator (t-PA) to blocked retinal veins, the peeling of epiretinal membranes from the retina, and the development of diagnostic procedures based on mapping oxygen concentration at the vitreous-retina interface. With successful animal trials, a path to human trials and commercialization will follow. Clearly, many systems in the body have the potential to benefit from the endoluminal technologies that this project considers, including the digestive system, the circulatory system, the urinary system, the central nervous system, the respiratory system, the female reproductive system and even the fetus. Microrobotic retinal therapies will greatly illuminate the potential that the integration of microrobotics and nanomedicine holds for society, and greatly accelerate this trend in Europe.
Max ERC Funding
2 498 044 €
Duration
Start date: 2011-04-01, End date: 2016-03-31
Project acronym BRAINCOMPATH
Project Mesoscale Brain Dynamics: Computing with Neuronal Pathways
Researcher (PI) Fritjof Helmchen
Host Institution (HI) UNIVERSITAT ZURICH
Country Switzerland
Call Details Advanced Grant (AdG), LS5, ERC-2014-ADG
Summary Brain computations rely on proper signal flow through the complex network of connected brain regions. Despite a wealth of anatomical and functional data – from microscopic to macroscopic scale – we still poorly understand the principles of how signal flow is routed through neuronal networks to generate appropriate behavior. Brain dynamics on the 'mesoscopic' scale, the intermediate level where local microcircuits communicate via axonal pathways, has remained a particular blind spot of research as it has been difficult to access under in vivo conditions. Here, I propose to tackle the mesoscopic level of brain dynamics both experimentally and theoretically, adopting a fresh perspective centered on neuronal pathway dynamics. Experimentally, we will utilize and further advance state-of-the-art genetic and optical techniques to create a toolbox for measuring and manipulating signal flow in pathway networks across a broad range of temporal scales. In particular, we will improve fiber-optic based methods for probing the activity of either individual or multiple neuronal pathways with high specificity. Using these tools we will set out to reveal mesoscopic brain dynamics across relevant cortical and subcortical regions in awake, behaving mice. Specifically, we will investigate sensorimotor learning for a reward-based texture discrimination task and rapid sensorimotor control during skilled locomotion. Moreover, by combining fiber-optic methods with two-photon microscopy and fMRI, respectively, we will start linking the meso-level to the micro- and macro-levels. Throughout the project, experiments will be complemented by computational approaches to analyse data, model pathway dynamics, and conceptualize a formal theory of mesoscopic dynamics. This project may transform the field by bridging the hierarchical brain levels and opening significant new avenues to assess physiological as well as pathological signal flow in the brain.
Summary
Brain computations rely on proper signal flow through the complex network of connected brain regions. Despite a wealth of anatomical and functional data – from microscopic to macroscopic scale – we still poorly understand the principles of how signal flow is routed through neuronal networks to generate appropriate behavior. Brain dynamics on the 'mesoscopic' scale, the intermediate level where local microcircuits communicate via axonal pathways, has remained a particular blind spot of research as it has been difficult to access under in vivo conditions. Here, I propose to tackle the mesoscopic level of brain dynamics both experimentally and theoretically, adopting a fresh perspective centered on neuronal pathway dynamics. Experimentally, we will utilize and further advance state-of-the-art genetic and optical techniques to create a toolbox for measuring and manipulating signal flow in pathway networks across a broad range of temporal scales. In particular, we will improve fiber-optic based methods for probing the activity of either individual or multiple neuronal pathways with high specificity. Using these tools we will set out to reveal mesoscopic brain dynamics across relevant cortical and subcortical regions in awake, behaving mice. Specifically, we will investigate sensorimotor learning for a reward-based texture discrimination task and rapid sensorimotor control during skilled locomotion. Moreover, by combining fiber-optic methods with two-photon microscopy and fMRI, respectively, we will start linking the meso-level to the micro- and macro-levels. Throughout the project, experiments will be complemented by computational approaches to analyse data, model pathway dynamics, and conceptualize a formal theory of mesoscopic dynamics. This project may transform the field by bridging the hierarchical brain levels and opening significant new avenues to assess physiological as well as pathological signal flow in the brain.
Max ERC Funding
2 498 915 €
Duration
Start date: 2016-02-01, End date: 2021-01-31
Project acronym BROADimmune
Project Structural, genetic and functional analyses of broadly neutralizing antibodies against human pathogens
Researcher (PI) Antonio Lanzavecchia
Host Institution (HI) FONDAZIONE PER L ISTITUTO DI RICERCA IN BIOMEDICINA
Country Switzerland
Call Details Advanced Grant (AdG), LS6, ERC-2014-ADG
Summary The overall goal of this project is to understand the molecular mechanisms that lead to the generation of potent and broadly neutralizing antibodies against medically relevant pathogens, and to identify the factors that limit their production in response to infection or vaccination with current vaccines. We will use high-throughput cellular screens to isolate from immune donors clonally related antibodies to different sites of influenza hemagglutinin, which will be fully characterized and sequenced in order to reconstruct their developmental pathways. Using this approach, we will ask fundamental questions with regards to the role of somatic mutations in affinity maturation and intraclonal diversification, which in some cases may lead to the generation of autoantibodies. We will combine crystallography and long time-scale molecular dynamics simulation to understand how mutations can increase affinity and broaden antibody specificity. By mapping the B and T cell response to all sites and conformations of influenza hemagglutinin, we will uncover the factors, such as insufficient T cell help or the instability of the pre-fusion hemagglutinin, that may limit the generation of broadly neutralizing antibodies. We will also perform a broad analysis of the antibody response to erythrocytes infected by P. falciparum to identify conserved epitopes on the parasite and to unravel the role of an enigmatic V gene that appears to be involved in response to blood-stage parasites. The hypotheses tested are strongly supported by preliminary observations from our own laboratory. While these studies will contribute to our understanding of B cell biology, the results obtained will also have translational implications for the development of potent and broad-spectrum antibodies, for the definition of correlates of protection, and for improving vaccine design.
Summary
The overall goal of this project is to understand the molecular mechanisms that lead to the generation of potent and broadly neutralizing antibodies against medically relevant pathogens, and to identify the factors that limit their production in response to infection or vaccination with current vaccines. We will use high-throughput cellular screens to isolate from immune donors clonally related antibodies to different sites of influenza hemagglutinin, which will be fully characterized and sequenced in order to reconstruct their developmental pathways. Using this approach, we will ask fundamental questions with regards to the role of somatic mutations in affinity maturation and intraclonal diversification, which in some cases may lead to the generation of autoantibodies. We will combine crystallography and long time-scale molecular dynamics simulation to understand how mutations can increase affinity and broaden antibody specificity. By mapping the B and T cell response to all sites and conformations of influenza hemagglutinin, we will uncover the factors, such as insufficient T cell help or the instability of the pre-fusion hemagglutinin, that may limit the generation of broadly neutralizing antibodies. We will also perform a broad analysis of the antibody response to erythrocytes infected by P. falciparum to identify conserved epitopes on the parasite and to unravel the role of an enigmatic V gene that appears to be involved in response to blood-stage parasites. The hypotheses tested are strongly supported by preliminary observations from our own laboratory. While these studies will contribute to our understanding of B cell biology, the results obtained will also have translational implications for the development of potent and broad-spectrum antibodies, for the definition of correlates of protection, and for improving vaccine design.
Max ERC Funding
1 867 500 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym BSMOXFORD
Project Physics Beyond the Standard Model at the LHC and with Atom Interferometers
Researcher (PI) Savas Dimopoulos
Host Institution (HI) EUROPEAN ORGANIZATION FOR NUCLEAR RESEARCH
Country Switzerland
Call Details Advanced Grant (AdG), PE2, ERC-2008-AdG
Summary Elementary particle physics is entering a spectacular new era in which experiments at the Large Hadron Collider (LHC) at CERN will soon start probing some of the deepest questions in physics, such as: Why is gravity so weak? Do elementary particles have substructure? What is the origin of mass? Are there new dimensions? Can we produce black holes in the lab? Could there be other universes with different physical laws? While the LHC pushes the energy frontier, the unprecedented precision of Atom Interferometry, has pointed me to a new tool for fundamental physics. These experiments based on the quantum interference of atoms can test General Relativity on the surface of the Earth, detect gravity waves, and test short-distance gravity, charge quantization, and quantum mechanics with unprecedented precision in the next decade. This ERC Advanced grant proposal is aimed at setting up a world-leading European center for development of a deeper theory of fundamental physics. The next 10 years is the optimal time for such studies to benefit from the wealth of new data that will emerge from the LHC, astrophysical observations and atom interferometry. This is a once-in-a-generation opportunity for making ground-breaking progress, and will open up many new research horizons.
Summary
Elementary particle physics is entering a spectacular new era in which experiments at the Large Hadron Collider (LHC) at CERN will soon start probing some of the deepest questions in physics, such as: Why is gravity so weak? Do elementary particles have substructure? What is the origin of mass? Are there new dimensions? Can we produce black holes in the lab? Could there be other universes with different physical laws? While the LHC pushes the energy frontier, the unprecedented precision of Atom Interferometry, has pointed me to a new tool for fundamental physics. These experiments based on the quantum interference of atoms can test General Relativity on the surface of the Earth, detect gravity waves, and test short-distance gravity, charge quantization, and quantum mechanics with unprecedented precision in the next decade. This ERC Advanced grant proposal is aimed at setting up a world-leading European center for development of a deeper theory of fundamental physics. The next 10 years is the optimal time for such studies to benefit from the wealth of new data that will emerge from the LHC, astrophysical observations and atom interferometry. This is a once-in-a-generation opportunity for making ground-breaking progress, and will open up many new research horizons.
Max ERC Funding
2 200 000 €
Duration
Start date: 2009-05-01, End date: 2014-04-30
Project acronym CAP
Project Computers Arguing with People
Researcher (PI) Sarit Kraus
Host Institution (HI) BAR ILAN UNIVERSITY
Country Israel
Call Details Advanced Grant (AdG), PE6, ERC-2010-AdG_20100224
Summary An important form of negotiation is argumentation. This is the ability to argue and to persuade the other party to accept a desired agreement, to acquire or give information, to coordinate goals and actions, and to find and verify evidence. This is a key capability in negotiating with humans.
While automated negotiations between software agents can often exchange offers and counteroffers, humans require persuasion. This challenges the design of agents arguing with people, with the objective that the outcome of the negotiation will meet the preferences of the arguer agent.
CAP’s objective is to enable automated agents to argue and persuade humans.
To achieve this, we intend to develop the following key components:
1) The extension of current game theory models of persuasion and bargaining to more realistic settings, 2) Algorithms and heuristics for generation and evaluation of arguments during negotiation with people, 3) Algorithms and heuristics for managing inconsistent views of the negotiation environment, and decision procedures for revelation, signalling, and requesting information, 4) The revision and update of the agent’s mental state and incorporation of social context, 5) Identifying strategies for expressing emotions in negotiations, 6) Technology for general opponent modelling from sparse and noisy data.
To demonstrate the developed methods, we will implement two training systems for people to improve their interviewing capabilities, and for training negotiators in inter-culture negotiations.
CAP will revolutionise the state of the art of automated systems negotiating with people. It will also create breakthroughs in the research of multi-agent systems in general, and will change paradigms by providing new directions for the way computers interact with people.
Summary
An important form of negotiation is argumentation. This is the ability to argue and to persuade the other party to accept a desired agreement, to acquire or give information, to coordinate goals and actions, and to find and verify evidence. This is a key capability in negotiating with humans.
While automated negotiations between software agents can often exchange offers and counteroffers, humans require persuasion. This challenges the design of agents arguing with people, with the objective that the outcome of the negotiation will meet the preferences of the arguer agent.
CAP’s objective is to enable automated agents to argue and persuade humans.
To achieve this, we intend to develop the following key components:
1) The extension of current game theory models of persuasion and bargaining to more realistic settings, 2) Algorithms and heuristics for generation and evaluation of arguments during negotiation with people, 3) Algorithms and heuristics for managing inconsistent views of the negotiation environment, and decision procedures for revelation, signalling, and requesting information, 4) The revision and update of the agent’s mental state and incorporation of social context, 5) Identifying strategies for expressing emotions in negotiations, 6) Technology for general opponent modelling from sparse and noisy data.
To demonstrate the developed methods, we will implement two training systems for people to improve their interviewing capabilities, and for training negotiators in inter-culture negotiations.
CAP will revolutionise the state of the art of automated systems negotiating with people. It will also create breakthroughs in the research of multi-agent systems in general, and will change paradigms by providing new directions for the way computers interact with people.
Max ERC Funding
2 334 057 €
Duration
Start date: 2011-07-01, End date: 2016-06-30
Project acronym CardHeal
Project Novel strategies for mammalian cardiac repair
Researcher (PI) Eldad TZAHOR
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Country Israel
Call Details Advanced Grant (AdG), LS4, ERC-2017-ADG
Summary Recent ground-breaking studies by my team and others demonstrated that latent heart regeneration machinery can be awakened even in adult mammals. My lab’s main contribution is the identification of two, apparently different, molecular mechanisms for augmenting cardiac regeneration in adult mice. The first requires transient activation of ErbB2 signalling in cardiomyocytes and the second involves extra cellular matrix-driven signalling by the proteoglycan agrin. Impressively, both mechanisms promote a major regenerative response that, in turn, enhances cardiac repair. In CardHeal we will use the two powerful regenerative models to obtain a holistic view of cardiac regeneration and repair mechanisms in mammals (mice and pigs).
In Aim 1, we will explore the molecular mechanisms underlying our discovery that transient activation of ErbB2 in adult cardiomyocytes results in massive cardiomyocyte dedifferentiation and proliferation followed by new vessels formation, scar resolution and functional cardiac repair. Specific objectives focus on ErbB2-Yap/Hippo signalling during cardiac regeneration; ErbB2 activation in a chronic heart failure model; ErbB2-induced regenerative EMT-like process; and cardiomyocyte re-differentiation.
In Aim 2, we will investigate the therapeutic effects of agrin, whose administration into injured hearts of mice and pigs elicits a significant regenerative response. Specific objectives are matrix-related cardiac regenerative cues, modulation of the immune response, angiogenesis, matrix remodeling, and developing a preclinical, large animal model to study agrin efficacy for cardiac repair.
Interrogating the differences and similarities between our two regenerative models should give us a detailed roadmap for cardiac regenerative medicine by providing deeper knowledge of the regenerative process in the heart and pointing to novel targets for cardiac repair in human patients.
Summary
Recent ground-breaking studies by my team and others demonstrated that latent heart regeneration machinery can be awakened even in adult mammals. My lab’s main contribution is the identification of two, apparently different, molecular mechanisms for augmenting cardiac regeneration in adult mice. The first requires transient activation of ErbB2 signalling in cardiomyocytes and the second involves extra cellular matrix-driven signalling by the proteoglycan agrin. Impressively, both mechanisms promote a major regenerative response that, in turn, enhances cardiac repair. In CardHeal we will use the two powerful regenerative models to obtain a holistic view of cardiac regeneration and repair mechanisms in mammals (mice and pigs).
In Aim 1, we will explore the molecular mechanisms underlying our discovery that transient activation of ErbB2 in adult cardiomyocytes results in massive cardiomyocyte dedifferentiation and proliferation followed by new vessels formation, scar resolution and functional cardiac repair. Specific objectives focus on ErbB2-Yap/Hippo signalling during cardiac regeneration; ErbB2 activation in a chronic heart failure model; ErbB2-induced regenerative EMT-like process; and cardiomyocyte re-differentiation.
In Aim 2, we will investigate the therapeutic effects of agrin, whose administration into injured hearts of mice and pigs elicits a significant regenerative response. Specific objectives are matrix-related cardiac regenerative cues, modulation of the immune response, angiogenesis, matrix remodeling, and developing a preclinical, large animal model to study agrin efficacy for cardiac repair.
Interrogating the differences and similarities between our two regenerative models should give us a detailed roadmap for cardiac regenerative medicine by providing deeper knowledge of the regenerative process in the heart and pointing to novel targets for cardiac repair in human patients.
Max ERC Funding
2 268 750 €
Duration
Start date: 2018-06-01, End date: 2023-05-31
Project acronym cdGMP
Project Time, space and speed: cdGMP signaling in cell behavior and reproduction
Researcher (PI) Urs Jenal
Host Institution (HI) UNIVERSITAT BASEL
Country Switzerland
Call Details Advanced Grant (AdG), LS6, ERC-2012-ADG_20120314
Summary Bacterial biofilms are the primary cause of chronic infections and of resulting infection relapses. To be able to interfere with bacterial persistence it is vital to understand the molecular details of biofilm formation and to define how motile planktonic cells transit into surface-grown communities. The nucleotide second messenger cyclic di-guanosinemonophosphate (cdGMP) has emerged as a central regulatory factor governing bacterial surface adaptation and biofilm formation. Although cdGMP signaling may well represent the Achilles heel of bacterial communities, cdGMP networks in bacterial pathogens are exquisitely complex and an integrated cellular system to uncover the details of cdGMP dynamics is missing.
To quantitatively describe cdGMP signaling we propose to exploit Caulobacter crescentus, an organism with a simple bimodal life-style that integrates the sessile-motile switch into its asymmetric division cycle. We aim to: 1) identify the role and regulation of all diguanylate cyclases and phosphodiesterases that contribute to the asymmetric cellular program with the goal to model the temporal and spatial distribution of cdGMP during development; 2) identify and characterize cdGMP effectors, their downstream targets and cellular pathways; 3) elucidate how cdGMP coordinates cell differentiation with cell growth and propagation; 4) unravel the role of cdGMP as an allosteric regulator in mechanosensation and in rapid adaptation of bacteria to growth on surfaces; 5) develop novel tools to quantitatively describe cdGMP network dynamics as the basis for mathematical modeling that provides the predictive power to experimentally test and refine important network parameters. We propose a multidisciplinary research program at the forefront of bacterial signal transduction that will provide the molecular and conceptual framework for a rapidly growing research field of second messenger signaling in pathogenic bacteria.
Summary
Bacterial biofilms are the primary cause of chronic infections and of resulting infection relapses. To be able to interfere with bacterial persistence it is vital to understand the molecular details of biofilm formation and to define how motile planktonic cells transit into surface-grown communities. The nucleotide second messenger cyclic di-guanosinemonophosphate (cdGMP) has emerged as a central regulatory factor governing bacterial surface adaptation and biofilm formation. Although cdGMP signaling may well represent the Achilles heel of bacterial communities, cdGMP networks in bacterial pathogens are exquisitely complex and an integrated cellular system to uncover the details of cdGMP dynamics is missing.
To quantitatively describe cdGMP signaling we propose to exploit Caulobacter crescentus, an organism with a simple bimodal life-style that integrates the sessile-motile switch into its asymmetric division cycle. We aim to: 1) identify the role and regulation of all diguanylate cyclases and phosphodiesterases that contribute to the asymmetric cellular program with the goal to model the temporal and spatial distribution of cdGMP during development; 2) identify and characterize cdGMP effectors, their downstream targets and cellular pathways; 3) elucidate how cdGMP coordinates cell differentiation with cell growth and propagation; 4) unravel the role of cdGMP as an allosteric regulator in mechanosensation and in rapid adaptation of bacteria to growth on surfaces; 5) develop novel tools to quantitatively describe cdGMP network dynamics as the basis for mathematical modeling that provides the predictive power to experimentally test and refine important network parameters. We propose a multidisciplinary research program at the forefront of bacterial signal transduction that will provide the molecular and conceptual framework for a rapidly growing research field of second messenger signaling in pathogenic bacteria.
Max ERC Funding
2 496 000 €
Duration
Start date: 2013-05-01, End date: 2018-04-30
Project acronym CellularBiographies
Project Global views of cell type specification and differentiation
Researcher (PI) Alexander Schier
Host Institution (HI) UNIVERSITAT BASEL
Country Switzerland
Call Details Advanced Grant (AdG), LS3, ERC-2018-ADG
Summary Each cell in our body has a specific biography that is defined by its pedigree relationship with other cells (lineage) and by its history of gene expression (trajectory). A fundamental question in cellular and developmental biology has been how the lineage and trajectory of a cell lead to its specification and differentiation. Remarkable progress in genome editing and single-cell sequencing has generated the opportunity to understand this process at global scales and single-cell resolution. We have recently developed methods to reconstruct the cellular ancestry and transcriptional trajectories of cells during embryogenesis. The resulting lineage and trajectory trees can be analyzed to gain comprehensive views of how cellular diversity arises and how differentiation leads to physiologically specialized cell types. To generate such global views of cellular development, we will: 1. Define the cellular diversity and gene expression trajectories during zebrafish embryogenesis and organogenesis. Trajectory trees will be generated from scRNA-seq data and analyzed to reconstruct the gene expression pathways underlying fate specification. 2. Reveal the relationships between lineage and transcriptional trajectories during fate specification. Lineage trees will be generated by marking cells via genome editing and combined with trajectory trees to reveal the cellular paths towards fate specification. 3. Discover the gene expression cascades that remodel cells into physiologically functional types. Cell biological modules will be identified by comparing gene enrichment in differentiation trajectories and reveal the specialized and shared mechanisms of differentiation. These studies will help provide the first comprehensive and global view of the trajectories and lineages underlying vertebrate development. Our focus is on the zebrafish model system, but the data and concepts developed in this project will be applicable to other developmental and cellular systems.
Summary
Each cell in our body has a specific biography that is defined by its pedigree relationship with other cells (lineage) and by its history of gene expression (trajectory). A fundamental question in cellular and developmental biology has been how the lineage and trajectory of a cell lead to its specification and differentiation. Remarkable progress in genome editing and single-cell sequencing has generated the opportunity to understand this process at global scales and single-cell resolution. We have recently developed methods to reconstruct the cellular ancestry and transcriptional trajectories of cells during embryogenesis. The resulting lineage and trajectory trees can be analyzed to gain comprehensive views of how cellular diversity arises and how differentiation leads to physiologically specialized cell types. To generate such global views of cellular development, we will: 1. Define the cellular diversity and gene expression trajectories during zebrafish embryogenesis and organogenesis. Trajectory trees will be generated from scRNA-seq data and analyzed to reconstruct the gene expression pathways underlying fate specification. 2. Reveal the relationships between lineage and transcriptional trajectories during fate specification. Lineage trees will be generated by marking cells via genome editing and combined with trajectory trees to reveal the cellular paths towards fate specification. 3. Discover the gene expression cascades that remodel cells into physiologically functional types. Cell biological modules will be identified by comparing gene enrichment in differentiation trajectories and reveal the specialized and shared mechanisms of differentiation. These studies will help provide the first comprehensive and global view of the trajectories and lineages underlying vertebrate development. Our focus is on the zebrafish model system, but the data and concepts developed in this project will be applicable to other developmental and cellular systems.
Max ERC Funding
2 411 440 €
Duration
Start date: 2020-01-01, End date: 2024-12-31
Project acronym CENDUP
Project Decoding the mechanisms of centrosome duplication
Researcher (PI) Pierre Goenczy
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS3, ERC-2008-AdG
Summary Centrosome duplication entails the formation of a single procentriole next to each centriole once per cell cycle. The mechanisms governing procentriole formation are poorly understood and constitute a fundamental open question in cell biology. We will launch an innovative multidisciplinary research program to gain significant insight into these mechanisms using C. elegans and human cells. This research program is also expected to have a significant impact by contributing important novel assays to the field. Six specific aims will be pursued: 1) SAS-6 as a ZYG-1 substrate: mechanisms of procentriole formation in C. elegans. We will test in vivo the consequence of SAS-6 phosphorylation by ZYG-1. 2) Biochemical and structural analysis of SAS-6-containing macromolecular complexes (SAMACs). We will isolate and characterize SAMACs from C. elegans embryos and human cells, and analyze their structure using single-particle electron microscopy. 3) Novel cell-free assay for procentriole formation in human cells. We will develop such an assay and use it to test whether SAMACs can direct procentriole formation and whether candidate proteins are needed at centrioles or in the cytoplasm. 4) Mapping interactions between centriolar proteins in live human cells. We will use chemical methods developed by our collaborators to probe interactions between HsSAS-6 and centriolar proteins in a time- and space-resolved manner. 5) Functional genomic and chemical genetic screens in human cells. We will conduct high-throughput fluorescence-based screens in human cells to identify novel genes required for procentriole formation and small molecule inhibitors of this process. 6) Mechanisms underlying differential centriolar maintenance in the germline. In C. elegans, we will characterize how the sas-1 locus is required for centriole maintenance during spermatogenesis, as well as analyze centriole elimination during oogenesis and identify components needed for this process
Summary
Centrosome duplication entails the formation of a single procentriole next to each centriole once per cell cycle. The mechanisms governing procentriole formation are poorly understood and constitute a fundamental open question in cell biology. We will launch an innovative multidisciplinary research program to gain significant insight into these mechanisms using C. elegans and human cells. This research program is also expected to have a significant impact by contributing important novel assays to the field. Six specific aims will be pursued: 1) SAS-6 as a ZYG-1 substrate: mechanisms of procentriole formation in C. elegans. We will test in vivo the consequence of SAS-6 phosphorylation by ZYG-1. 2) Biochemical and structural analysis of SAS-6-containing macromolecular complexes (SAMACs). We will isolate and characterize SAMACs from C. elegans embryos and human cells, and analyze their structure using single-particle electron microscopy. 3) Novel cell-free assay for procentriole formation in human cells. We will develop such an assay and use it to test whether SAMACs can direct procentriole formation and whether candidate proteins are needed at centrioles or in the cytoplasm. 4) Mapping interactions between centriolar proteins in live human cells. We will use chemical methods developed by our collaborators to probe interactions between HsSAS-6 and centriolar proteins in a time- and space-resolved manner. 5) Functional genomic and chemical genetic screens in human cells. We will conduct high-throughput fluorescence-based screens in human cells to identify novel genes required for procentriole formation and small molecule inhibitors of this process. 6) Mechanisms underlying differential centriolar maintenance in the germline. In C. elegans, we will characterize how the sas-1 locus is required for centriole maintenance during spermatogenesis, as well as analyze centriole elimination during oogenesis and identify components needed for this process
Max ERC Funding
2 004 155 €
Duration
Start date: 2009-04-01, End date: 2014-03-31
Project acronym CENFOR
Project Dissecting the mechanisms governing centriole formation
Researcher (PI) Pierre Goenczy
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS3, ERC-2013-ADG
Summary "Centrioles are critical for the formation of cilia, flagella and centrosomes, as well as for human health. The mechanisms governing centriole formation constitute a long-standing question in cell biology. We will pursue an innovative multidisciplinary research program to gain further insight into these mechanisms, using human cells, C. elegans and Trichonympha as model systems. This program is expected to also have a major impact by contributing a novel cell free assay to the field, thus paving the way towards making synthetic centrioles. Six specific aims will be pursued:
1) Deciphering HsSAS-6/STIL distribution and dynamics. We will use super-resolution microscopy, molecular counting, photoconversion and FCS to further characterize these two key components required for centriole formation in human cells.
2) The SAS-6 ring model as a tool to redirect centriole organization. Utilizing predictions from the SAS-6 ring model, we will assay the consequences for centrioles and cilia of altering the diameter and symmetry of the structure.
3) Determining the architecture of C. elegans centrioles. We will conduct molecular counting and cryo-ET of purified C. elegans centrioles to determine if they contain a spiral or a cartwheel, as well as identify SAS-6-interacting components.
4) Comprehensive 3D map and proteomics of Trichonympha centriole. We will obtain a ~35 Å 3D map of the complete T. agilis centriole, perform proteomic analysis to identify its constituents and test their function using RNAi.
5) Regulation of cartwheel height and centriole length. We will explore whether cartwheel height is set by SAS-6 proteins and perform screens in human cells to identify novel components regulating cartwheel height and centriole length.
6) Novel cell free assay for cartwheel assembly and centriole formation. Using SAS-6 proteins on a lipid monolayer as starting point, we will develop and utilize a cell-free assay to reconstitute cartwheel assembly and centriole format"
Summary
"Centrioles are critical for the formation of cilia, flagella and centrosomes, as well as for human health. The mechanisms governing centriole formation constitute a long-standing question in cell biology. We will pursue an innovative multidisciplinary research program to gain further insight into these mechanisms, using human cells, C. elegans and Trichonympha as model systems. This program is expected to also have a major impact by contributing a novel cell free assay to the field, thus paving the way towards making synthetic centrioles. Six specific aims will be pursued:
1) Deciphering HsSAS-6/STIL distribution and dynamics. We will use super-resolution microscopy, molecular counting, photoconversion and FCS to further characterize these two key components required for centriole formation in human cells.
2) The SAS-6 ring model as a tool to redirect centriole organization. Utilizing predictions from the SAS-6 ring model, we will assay the consequences for centrioles and cilia of altering the diameter and symmetry of the structure.
3) Determining the architecture of C. elegans centrioles. We will conduct molecular counting and cryo-ET of purified C. elegans centrioles to determine if they contain a spiral or a cartwheel, as well as identify SAS-6-interacting components.
4) Comprehensive 3D map and proteomics of Trichonympha centriole. We will obtain a ~35 Å 3D map of the complete T. agilis centriole, perform proteomic analysis to identify its constituents and test their function using RNAi.
5) Regulation of cartwheel height and centriole length. We will explore whether cartwheel height is set by SAS-6 proteins and perform screens in human cells to identify novel components regulating cartwheel height and centriole length.
6) Novel cell free assay for cartwheel assembly and centriole formation. Using SAS-6 proteins on a lipid monolayer as starting point, we will develop and utilize a cell-free assay to reconstitute cartwheel assembly and centriole format"
Max ERC Funding
2 499 270 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym CENGIN
Project Deciphering and engineering centriole assembly
Researcher (PI) Pierre Joerg GoeNCZY
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS3, ERC-2018-ADG
Summary Deciphering and engineering the assembly of cellular organelles is a key pursuit in biology. The centriole is an evolutionarily conserved organelle well suited for this goal, and which is crucial for cell signaling, motility and division. The centriole exhibits a striking 9-fold radial symmetry of microtubules around a likewise symmetrical cartwheel containing stacked ring-bearing structures. Components essential for generating this remarkable architecture from alga to man have been identified. A next critical step is to engineer assays to probe the dynamics of centriole assembly with molecular precision to fully understand how these components together build a functional organelle. Our ambitious research proposal aims at taking groundbreaking steps in this direction through four specific aims:
1) Reconstituting cartwheel ring assembly dynamics. We will use high-speed AFM (HS-AFM) to dissect the biophysics of SAS-6 ring polymer dynamics at the root of cartwheel assembly. We will also use HS-AFM to analyze monobodies against SAS-6, as well as engineer surfaces and DNA origamis to further dissect ring assembly.
2) Deciphering ring stacking mechanisms. We will use cryo-ET to identify SAS-6 features that direct stacking of ring structures and set cartwheel height. Moreover, we will develop an HS-AFM stacking assay and a reconstituted stacking assay from human cells.
3) Understanding peripheral element contributions to centriole biogenesis. We will dissect the function of the peripheral centriole pinhead protein Cep135/Bld10p, as well as identify and likewise dissect peripheral A-C linker proteins. Furthermore, we will further engineer the HS-AFM assay to include such peripheral components.
4) Dissecting de novo centriole assembly mechanisms. We will dissect de novo centriole formation in human cells and water fern. We will also explore whether de novo formation involves a phase separation mechanism and repurpose the HS-AFM assay to probe de novo organelle biogenes
Summary
Deciphering and engineering the assembly of cellular organelles is a key pursuit in biology. The centriole is an evolutionarily conserved organelle well suited for this goal, and which is crucial for cell signaling, motility and division. The centriole exhibits a striking 9-fold radial symmetry of microtubules around a likewise symmetrical cartwheel containing stacked ring-bearing structures. Components essential for generating this remarkable architecture from alga to man have been identified. A next critical step is to engineer assays to probe the dynamics of centriole assembly with molecular precision to fully understand how these components together build a functional organelle. Our ambitious research proposal aims at taking groundbreaking steps in this direction through four specific aims:
1) Reconstituting cartwheel ring assembly dynamics. We will use high-speed AFM (HS-AFM) to dissect the biophysics of SAS-6 ring polymer dynamics at the root of cartwheel assembly. We will also use HS-AFM to analyze monobodies against SAS-6, as well as engineer surfaces and DNA origamis to further dissect ring assembly.
2) Deciphering ring stacking mechanisms. We will use cryo-ET to identify SAS-6 features that direct stacking of ring structures and set cartwheel height. Moreover, we will develop an HS-AFM stacking assay and a reconstituted stacking assay from human cells.
3) Understanding peripheral element contributions to centriole biogenesis. We will dissect the function of the peripheral centriole pinhead protein Cep135/Bld10p, as well as identify and likewise dissect peripheral A-C linker proteins. Furthermore, we will further engineer the HS-AFM assay to include such peripheral components.
4) Dissecting de novo centriole assembly mechanisms. We will dissect de novo centriole formation in human cells and water fern. We will also explore whether de novo formation involves a phase separation mechanism and repurpose the HS-AFM assay to probe de novo organelle biogenes
Max ERC Funding
2 500 000 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym CFRFSS
Project Chromatin Fiber and Remodeling Factor Structural Studies
Researcher (PI) Timothy John Richmond
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Country Switzerland
Call Details Advanced Grant (AdG), LS1, ERC-2012-ADG_20120314
Summary "DNA in higher organisms is organized in a nucleoprotein complex called chromatin. The structure of chromatin is responsible for compacting DNA to fit within the nucleus and for governing its access in nuclear processes. Epigenetic information is encoded chiefly via chromatin modifications. Readout of the genetic code depends on chromatin remodeling, a process actively altering chromatin structure. An understanding of the hierarchical structure of chromatin and of structurally based, remodeling mechanisms will have enormous impact for developments in medicine.
Following our high resolution structure of the nucleosome core particle, the fundamental repeating unit of chromatin, we have endeavored to determine the structure of the chromatin fiber. We showed with our X-ray structure of a tetranucleosome how nucleosomes could be organized in the fiber. Further progress has been limited by structural polymorphism and crystal disorder, but new evidence on the in vivo spacing of nucleosomes in chromatin should stimulate more advances. Part A of this application describes how we would apply these new findings to our cryo-electron microscopy study of the chromatin fiber and to our crystallographic study of a tetranucleosome containing linker histone.
Recently, my laboratory succeeded in providing the first structurally based mechanism for nucleosome spacing by a chromatin remodeling factor. We combined the X-ray structure of ISW1a(ATPase) bound to DNA with cryo-EM structures of the factor bound to two different nucleosomes to build a model showing how this remodeler uses a dinucleosome, not a mononucleosome, as its substrate. Our results from a functional assay using ISW1a further justified this model. Part B of this application describes how we would proceed to the relevant cryo-EM and X-ray structures incorporating dinucleosomes. Our recombinant ISW1a allows us to study in addition the interaction of the ATPase domain with nucleosome substrates."
Summary
"DNA in higher organisms is organized in a nucleoprotein complex called chromatin. The structure of chromatin is responsible for compacting DNA to fit within the nucleus and for governing its access in nuclear processes. Epigenetic information is encoded chiefly via chromatin modifications. Readout of the genetic code depends on chromatin remodeling, a process actively altering chromatin structure. An understanding of the hierarchical structure of chromatin and of structurally based, remodeling mechanisms will have enormous impact for developments in medicine.
Following our high resolution structure of the nucleosome core particle, the fundamental repeating unit of chromatin, we have endeavored to determine the structure of the chromatin fiber. We showed with our X-ray structure of a tetranucleosome how nucleosomes could be organized in the fiber. Further progress has been limited by structural polymorphism and crystal disorder, but new evidence on the in vivo spacing of nucleosomes in chromatin should stimulate more advances. Part A of this application describes how we would apply these new findings to our cryo-electron microscopy study of the chromatin fiber and to our crystallographic study of a tetranucleosome containing linker histone.
Recently, my laboratory succeeded in providing the first structurally based mechanism for nucleosome spacing by a chromatin remodeling factor. We combined the X-ray structure of ISW1a(ATPase) bound to DNA with cryo-EM structures of the factor bound to two different nucleosomes to build a model showing how this remodeler uses a dinucleosome, not a mononucleosome, as its substrate. Our results from a functional assay using ISW1a further justified this model. Part B of this application describes how we would proceed to the relevant cryo-EM and X-ray structures incorporating dinucleosomes. Our recombinant ISW1a allows us to study in addition the interaction of the ATPase domain with nucleosome substrates."
Max ERC Funding
2 500 000 €
Duration
Start date: 2013-01-01, End date: 2017-12-31
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
Country Israel
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 CHROMATINSYS
Project Systematic Approach to Dissect the Interplay between Chromatin and Transcription
Researcher (PI) Nir Friedman
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Country Israel
Call Details Advanced Grant (AdG), LS2, ERC-2013-ADG
Summary Epigenetic mechanisms play an important role in regulating and maintaining the functionality of cells and have been implicated in a wide range of human diseases. Histone proteins that form the protein core of nucleosomes are subject to a bewildering array of covalent and structural modifications, which can repress, permit, or promote transcription. These modifications can be added and removed by specialized complexes that are recruited by other covalent modifications, by transcription factors, or by the transcriptional machinery. Advances in genomics led to comprehensive mapping of the ``epigenome'' in a range of tissues and organisms. These maps established the tight connection between histone modifications and transcription programs. These static charts, however, are less successful at uncovering the underlying mechanisms, logic, and function of histone modifications in establishing and maintaining transcriptional programs. Our premise is that we can answer these basic questions by observing the effect of genetic perturbations on the dynamics of both chromatin state and transcriptional activity. We aim to dissect the chromatin-transcription system in a systematic manner by building on our extensive experience in modeling and analysis, and a unique high-throughput experimental system we established in my lab.
We plan to use the budding yeast model organism, which allows for
efficient genetic and experimental manipulations. We will combine two technologies: (1) high-throughput measurements of single-cell
transcriptional output using fluorescence reporters; and (2) high-throughput immunoprecipitation sequencing assays to map chromatin state. Measuring with these the dynamics of response to stimuli under different genetic backgrounds and using advanced stochastic network models, we will chart detailed mechanisms that are opaque to current approaches and elucidate the general principles that govern the interplay between chromatin and transcription.
Summary
Epigenetic mechanisms play an important role in regulating and maintaining the functionality of cells and have been implicated in a wide range of human diseases. Histone proteins that form the protein core of nucleosomes are subject to a bewildering array of covalent and structural modifications, which can repress, permit, or promote transcription. These modifications can be added and removed by specialized complexes that are recruited by other covalent modifications, by transcription factors, or by the transcriptional machinery. Advances in genomics led to comprehensive mapping of the ``epigenome'' in a range of tissues and organisms. These maps established the tight connection between histone modifications and transcription programs. These static charts, however, are less successful at uncovering the underlying mechanisms, logic, and function of histone modifications in establishing and maintaining transcriptional programs. Our premise is that we can answer these basic questions by observing the effect of genetic perturbations on the dynamics of both chromatin state and transcriptional activity. We aim to dissect the chromatin-transcription system in a systematic manner by building on our extensive experience in modeling and analysis, and a unique high-throughput experimental system we established in my lab.
We plan to use the budding yeast model organism, which allows for
efficient genetic and experimental manipulations. We will combine two technologies: (1) high-throughput measurements of single-cell
transcriptional output using fluorescence reporters; and (2) high-throughput immunoprecipitation sequencing assays to map chromatin state. Measuring with these the dynamics of response to stimuli under different genetic backgrounds and using advanced stochastic network models, we will chart detailed mechanisms that are opaque to current approaches and elucidate the general principles that govern the interplay between chromatin and transcription.
Max ERC Funding
2 396 450 €
Duration
Start date: 2014-01-01, End date: 2018-12-31
Project acronym CloudRadioNet
Project Cloud Wireless Networks: An Information Theoretic Framework
Researcher (PI) Shlomo Shamai Shitz
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Country Israel
Call Details Advanced Grant (AdG), PE7, ERC-2015-AdG
Summary This five years research proposal is focused on the development of novel information theoretic concepts and techniques and their usage, as to identify the ultimate communications limits and potential of different cloud radio network structures, in which the central signal processing is migrated to the cloud (remote central units), via fronthaul/backhaul infrastructure links. Moreover, it is also directed to introduce and study the optimal or close to optimal strategies for those systems that are to be motivated by the developed theory. We plan to address wireless networks, having future cellular technology in mind, but the basic tools and approaches to be built and researched are relevant to other communication networks as well. Cloud communication networks motivate novel information theoretic views, and perspectives that put backhaul/fronthaul connections in the center, thus deviating considerably from standard theoretical studies of communications links and networks, which are applied to this domain. Our approach accounts for the fact that in such networks information theoretic separation concepts are no longer optimal, hence isolating simple basic components of the network is essentially suboptimal. The proposed view incorporates, in a unified way, under the general cover of information theory: Multi-terminal distributed networks; Basic and timely concepts of distributed coding and communications; Network communications and primarily network coding, Index coding, as associated with interference alignment and caching; Information-Estimation relations and signal processing, addressing the impact of distributed channel state information directly; A variety of fundamental concepts in optimization and random matrix theories. This path provides a natural theoretical framework directed towards better understanding the potential and limitation of cloud networks on one hand and paves the way to innovative communications design principles on the other.
Summary
This five years research proposal is focused on the development of novel information theoretic concepts and techniques and their usage, as to identify the ultimate communications limits and potential of different cloud radio network structures, in which the central signal processing is migrated to the cloud (remote central units), via fronthaul/backhaul infrastructure links. Moreover, it is also directed to introduce and study the optimal or close to optimal strategies for those systems that are to be motivated by the developed theory. We plan to address wireless networks, having future cellular technology in mind, but the basic tools and approaches to be built and researched are relevant to other communication networks as well. Cloud communication networks motivate novel information theoretic views, and perspectives that put backhaul/fronthaul connections in the center, thus deviating considerably from standard theoretical studies of communications links and networks, which are applied to this domain. Our approach accounts for the fact that in such networks information theoretic separation concepts are no longer optimal, hence isolating simple basic components of the network is essentially suboptimal. The proposed view incorporates, in a unified way, under the general cover of information theory: Multi-terminal distributed networks; Basic and timely concepts of distributed coding and communications; Network communications and primarily network coding, Index coding, as associated with interference alignment and caching; Information-Estimation relations and signal processing, addressing the impact of distributed channel state information directly; A variety of fundamental concepts in optimization and random matrix theories. This path provides a natural theoretical framework directed towards better understanding the potential and limitation of cloud networks on one hand and paves the way to innovative communications design principles on the other.
Max ERC Funding
1 981 782 €
Duration
Start date: 2016-07-01, End date: 2021-12-31
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
Country Israel
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 COLT-MDP
Project Computational Learning Theory: compact representation, efficient computation, and societal challenges in learning MDPs
Researcher (PI) Yishay Mansour
Host Institution (HI) TEL AVIV UNIVERSITY
Country Israel
Call Details Advanced Grant (AdG), PE6, ERC-2019-ADG
Summary Computational learning theory has been highly successful over the last three decades, both in providing deep mathematical theories and in influencing the practice of machine learning. One of the great recent successes of computational learning theory has been the study of online learning and multi-arm bandits. This line of research has been highly successful, both theoretically and practically, addressing many important applications. Unfortunately, the recent theoretical progress in Markov Decision Process and reinforcement learning has been slower.
Based on my fundamental contributions to reinforcement learning (e.g. policy gradient, sparse sampling and trajectory trees), to online learning and machine learning in general, I propose to take the theoretical and practical success of online learning to the “next level” by making significant breakthroughs in reinforcement learning. Our main aim is to advance the state of the art in the theory of reinforcement learning, and our research will revolve around three pillars: (1) compact representation, (2) efficient computation and (3) societal challenges, including fairness and privacy.
A successful project will greatly impact reinforcement learning in all its stages. Modelling: Introducing new compact representation models, will enhance our understanding how to structure complex problems, which would greatly extend the applicability of reinforcement learning. Efficient computation: New algorithmic methodologies will give new insight for overcoming computational and statistical barriers both for planning and learning. Learning: New learning paradigms would address fundamental issues of copping with uncertainties in complex control environments of reinforcement learning. Societal challenges: Allowing the community to understand, assess, address and overcome societal challenges is of the greatest importance to the acceptance of AI methodologies by the general public.
Summary
Computational learning theory has been highly successful over the last three decades, both in providing deep mathematical theories and in influencing the practice of machine learning. One of the great recent successes of computational learning theory has been the study of online learning and multi-arm bandits. This line of research has been highly successful, both theoretically and practically, addressing many important applications. Unfortunately, the recent theoretical progress in Markov Decision Process and reinforcement learning has been slower.
Based on my fundamental contributions to reinforcement learning (e.g. policy gradient, sparse sampling and trajectory trees), to online learning and machine learning in general, I propose to take the theoretical and practical success of online learning to the “next level” by making significant breakthroughs in reinforcement learning. Our main aim is to advance the state of the art in the theory of reinforcement learning, and our research will revolve around three pillars: (1) compact representation, (2) efficient computation and (3) societal challenges, including fairness and privacy.
A successful project will greatly impact reinforcement learning in all its stages. Modelling: Introducing new compact representation models, will enhance our understanding how to structure complex problems, which would greatly extend the applicability of reinforcement learning. Efficient computation: New algorithmic methodologies will give new insight for overcoming computational and statistical barriers both for planning and learning. Learning: New learning paradigms would address fundamental issues of copping with uncertainties in complex control environments of reinforcement learning. Societal challenges: Allowing the community to understand, assess, address and overcome societal challenges is of the greatest importance to the acceptance of AI methodologies by the general public.
Max ERC Funding
1 878 125 €
Duration
Start date: 2020-10-01, End date: 2025-09-30
Project acronym COMPECON
Project Complexity and Simplicity in Economic Mechanisms
Researcher (PI) Noam NISAN
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Country Israel
Call Details Advanced Grant (AdG), PE6, ERC-2016-ADG
Summary As more and more economic activity is moving to the Internet, familiar economic mechanisms are being deployed
at unprecedented scales of size, speed, and complexity. In many cases this new complexity becomes the defining
feature of the deployed economic mechanism and the quantitative difference becomes a key qualitative one.
A well-studied example of such situations is how the humble single-item auction suddenly becomes a
billion-times repeated online ad auction, or even becomes a combinatorial auction with exponentially
many possible outcomes. Similar complexity explosions occur with various markets, with information
dissemination, with pricing structures, and with many other economic mechanisms.
The aim of this proposal is to study the role and implications of such complexity and to start
developing a coherent economic theory that can handle it. We aim to identify various measures of
complexity that are crucial bottlenecks and study them. Examples of such complexities include the
amount of access to data, the length of the description of a mechanism, its communication requirements,
the cognitive complexity required from users, and, of course, the associated computational complexity.
On one hand we will attempt finding ways of effectively dealing with complexity when it is needed, and on
the other hand, attempt avoiding complexity, when possible, replacing it with ``simple'' alternatives
without incurring too large of a loss.
Summary
As more and more economic activity is moving to the Internet, familiar economic mechanisms are being deployed
at unprecedented scales of size, speed, and complexity. In many cases this new complexity becomes the defining
feature of the deployed economic mechanism and the quantitative difference becomes a key qualitative one.
A well-studied example of such situations is how the humble single-item auction suddenly becomes a
billion-times repeated online ad auction, or even becomes a combinatorial auction with exponentially
many possible outcomes. Similar complexity explosions occur with various markets, with information
dissemination, with pricing structures, and with many other economic mechanisms.
The aim of this proposal is to study the role and implications of such complexity and to start
developing a coherent economic theory that can handle it. We aim to identify various measures of
complexity that are crucial bottlenecks and study them. Examples of such complexities include the
amount of access to data, the length of the description of a mechanism, its communication requirements,
the cognitive complexity required from users, and, of course, the associated computational complexity.
On one hand we will attempt finding ways of effectively dealing with complexity when it is needed, and on
the other hand, attempt avoiding complexity, when possible, replacing it with ``simple'' alternatives
without incurring too large of a loss.
Max ERC Funding
2 026 706 €
Duration
Start date: 2017-05-01, End date: 2022-04-30
Project acronym CORALWARM
Project Corals and global warming: The Mediterranean versus the Red Sea
Researcher (PI) Zvy Dubinsky
Host Institution (HI) BAR ILAN UNIVERSITY
Country Israel
Call Details Advanced Grant (AdG), LS8, ERC-2009-AdG
Summary CoralWarm will generate for the first time projections of temperate and subtropical coral survival by integrating sublethal temperature increase effects on metabolic and skeletal processes in Mediterranean and Red Sea key species. CoralWarm unique approach is from the nano- to the macro-scale, correlating molecular events to environmental processes. This will show new pathways to future investigations on cellular mechanisms linking environmental factors to final phenotype, potentially improving prediction powers and paleoclimatological interpretation. Biological and chemical expertise will merge, producing new interdisciplinary approaches for ecophysiology and biomineralization. Field transplantations will be combined with controlled experiments under IPCC scenarios. Corals will be grown in aquaria, exposing the Mediterranean species native to cooler waters to higher temperatures, and the Red Sea ones to gradually increasing above ambient warming seawater. Virtually all state-of-the-art methods will be used, by uniquely combining the investigators expertise. Expected results include responses of algal symbionts photosynthesis, host, symbiont and holobiont respiration, biomineralization rates and patterns, including colony architecture, and reproduction to temperature and pH gradients and combinations. Integration of molecular aspects of potential replacement of symbiont clades, changes in skeletal crystallography, with biochemical and physiological aspects of temperature response, will lead to a novel mechanistic model predicting changes in coral ecology and survival prospect. High-temperature tolerant clades and species will be revealed, allowing future bioremediation actions and establishment of coral refuges, saving corals and coral reefs for future generations.
Summary
CoralWarm will generate for the first time projections of temperate and subtropical coral survival by integrating sublethal temperature increase effects on metabolic and skeletal processes in Mediterranean and Red Sea key species. CoralWarm unique approach is from the nano- to the macro-scale, correlating molecular events to environmental processes. This will show new pathways to future investigations on cellular mechanisms linking environmental factors to final phenotype, potentially improving prediction powers and paleoclimatological interpretation. Biological and chemical expertise will merge, producing new interdisciplinary approaches for ecophysiology and biomineralization. Field transplantations will be combined with controlled experiments under IPCC scenarios. Corals will be grown in aquaria, exposing the Mediterranean species native to cooler waters to higher temperatures, and the Red Sea ones to gradually increasing above ambient warming seawater. Virtually all state-of-the-art methods will be used, by uniquely combining the investigators expertise. Expected results include responses of algal symbionts photosynthesis, host, symbiont and holobiont respiration, biomineralization rates and patterns, including colony architecture, and reproduction to temperature and pH gradients and combinations. Integration of molecular aspects of potential replacement of symbiont clades, changes in skeletal crystallography, with biochemical and physiological aspects of temperature response, will lead to a novel mechanistic model predicting changes in coral ecology and survival prospect. High-temperature tolerant clades and species will be revealed, allowing future bioremediation actions and establishment of coral refuges, saving corals and coral reefs for future generations.
Max ERC Funding
3 332 032 €
Duration
Start date: 2010-06-01, End date: 2016-05-31
Project acronym CRISPR-EVOL
Project The eco-evolutionary costs and benefits of CRISPR-Cas systems, and their effect on genome diversity within populations
Researcher (PI) Uri Gophna
Host Institution (HI) TEL AVIV UNIVERSITY
Country Israel
Call Details Advanced Grant (AdG), LS8, ERC-2017-ADG
Summary CRISPR-Cas systems are microbial defense systems that provide prokaryotes with acquired and heritable DNA-based immunity against selfish genetic elements, primarily viruses. However, the full scope of benefits that these systems can provide, as well as their costs remain unknown. Specifically, it is unclear whether the benefits against viral infection outweigh the continual costs incurred even in the absence of parasitic elements, and whether CRISPR-Cas systems affect microbial genome diversity in nature.
Since CRISPR-Cas systems can impede lateral gene transfer, it is often assumed that they reduce genetic diversity. Conversely, our recent results suggest the exact opposite: that these systems generate a high level of genomic diversity within populations. We have recently combined genomics of environmental strains and experimental genetics to show that archaea frequently acquire CRISPR immune memory, known as spacers, from chromosomes of related species in the environment. The presence of these spacers reduces gene exchange between lineages, indicating that CRISPR-Cas contributes to diversification. We have also shown that such inter-species mating events induce the acquisition of spacers against a strain's own replicons, supporting a role for CRISPR-Cas systems in generating deletions in natural plasmids and unessential genomic loci, again increasing genome diversity within populations.
Here we aim to test our hypothesis that CRISPR-Cas systems increase within-population diversity, and quantify their benefits to both cells and populations, using large-scale genomics and experimental evolution. We will explore how these systems alter the patterns of recombination within and between species, and explore the potential involvement of CRISPR-associated proteins in cellular DNA repair.
This work will reveal the eco-evolutionary role of CRISPR-Cas systems in shaping microbial populations, and open new research avenues regarding additional roles beyond anti-viral defense
Summary
CRISPR-Cas systems are microbial defense systems that provide prokaryotes with acquired and heritable DNA-based immunity against selfish genetic elements, primarily viruses. However, the full scope of benefits that these systems can provide, as well as their costs remain unknown. Specifically, it is unclear whether the benefits against viral infection outweigh the continual costs incurred even in the absence of parasitic elements, and whether CRISPR-Cas systems affect microbial genome diversity in nature.
Since CRISPR-Cas systems can impede lateral gene transfer, it is often assumed that they reduce genetic diversity. Conversely, our recent results suggest the exact opposite: that these systems generate a high level of genomic diversity within populations. We have recently combined genomics of environmental strains and experimental genetics to show that archaea frequently acquire CRISPR immune memory, known as spacers, from chromosomes of related species in the environment. The presence of these spacers reduces gene exchange between lineages, indicating that CRISPR-Cas contributes to diversification. We have also shown that such inter-species mating events induce the acquisition of spacers against a strain's own replicons, supporting a role for CRISPR-Cas systems in generating deletions in natural plasmids and unessential genomic loci, again increasing genome diversity within populations.
Here we aim to test our hypothesis that CRISPR-Cas systems increase within-population diversity, and quantify their benefits to both cells and populations, using large-scale genomics and experimental evolution. We will explore how these systems alter the patterns of recombination within and between species, and explore the potential involvement of CRISPR-associated proteins in cellular DNA repair.
This work will reveal the eco-evolutionary role of CRISPR-Cas systems in shaping microbial populations, and open new research avenues regarding additional roles beyond anti-viral defense
Max ERC Funding
2 495 625 €
Duration
Start date: 2018-05-01, End date: 2023-04-30
Project acronym CROSSINGSCALES
Project Reverse Scale-Crossing Effects In Biology
Researcher (PI) Lucas PELKMANS
Host Institution (HI) UNIVERSITAT ZURICH
Country Switzerland
Call Details Advanced Grant (AdG), LS2, ERC-2019-ADG
Summary The central dogma in biology often invokes a bottom-up picture of life. However, at different biological scales, new properties in form and function arise that have a superseding causal impact on the behaviour of the lower-scale components from which these new properties emerge. These top-down or reverse scale-crossing effects must be taken into account in order to make predictions about spatiotemporally controlled single-cell fates, activities, levels of gene expression, or the functional outcome of cellular signalling. They can stem from the multicellular, the cellular, and the intracellular scale, and can be quantified using multiscale and multiplexed RNA and protein state imaging in combination with computer vision and data-driven modelling. The ability to comprehensively map these reverse causal effects across multiple scales has the potential to revolutionize most, if not all domains of biology and medicine. In this project, we will establish the importance of reverse causal effects in human induced pluripotent stem cells and early D. rerio embryos. To achieve this, we will develop a quantitative imaging method beyond the diffraction limit of light without compromising scalability in temporal and spatial dimensions. We will also develop a method that achieves scalable, transcriptome-wide image-based multiplexing of mRNA transcripts, and we will extend our computer vision approaches to higher resolution and to three spatial dimensions. These methods will be systematically applied to stem cell collectives grown in 2D and 3D, as well as to early embryos, achieving comprehensive quantification of nuclear and chromatin states, gene expression, subcellular organization, cellular states, and tissue-scale organization across millions of individual cells within the same dataset. These datasets will be used to quantify how, at different scales, new properties in form and function arise that have a superseding causal impact on the behaviour of the lower-scale components
Summary
The central dogma in biology often invokes a bottom-up picture of life. However, at different biological scales, new properties in form and function arise that have a superseding causal impact on the behaviour of the lower-scale components from which these new properties emerge. These top-down or reverse scale-crossing effects must be taken into account in order to make predictions about spatiotemporally controlled single-cell fates, activities, levels of gene expression, or the functional outcome of cellular signalling. They can stem from the multicellular, the cellular, and the intracellular scale, and can be quantified using multiscale and multiplexed RNA and protein state imaging in combination with computer vision and data-driven modelling. The ability to comprehensively map these reverse causal effects across multiple scales has the potential to revolutionize most, if not all domains of biology and medicine. In this project, we will establish the importance of reverse causal effects in human induced pluripotent stem cells and early D. rerio embryos. To achieve this, we will develop a quantitative imaging method beyond the diffraction limit of light without compromising scalability in temporal and spatial dimensions. We will also develop a method that achieves scalable, transcriptome-wide image-based multiplexing of mRNA transcripts, and we will extend our computer vision approaches to higher resolution and to three spatial dimensions. These methods will be systematically applied to stem cell collectives grown in 2D and 3D, as well as to early embryos, achieving comprehensive quantification of nuclear and chromatin states, gene expression, subcellular organization, cellular states, and tissue-scale organization across millions of individual cells within the same dataset. These datasets will be used to quantify how, at different scales, new properties in form and function arise that have a superseding causal impact on the behaviour of the lower-scale components
Max ERC Funding
2 411 075 €
Duration
Start date: 2020-09-01, End date: 2025-08-31
Project acronym CsnCRL
Project The molecular basis of CULLIN E3 ligase regulation by the COP9 signalosome
Researcher (PI) Nicolas Thoma
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Country Switzerland
Call Details Advanced Grant (AdG), LS1, ERC-2014-ADG
Summary Specificity in the ubiquitin-proteasome system is largely conferred by ubiquitin E3 ligases (E3s). Cullin-RING ligases (CRLs), constituting ~30% of all E3s in humans, mediate the ubiquitination of ~20% of the proteins degraded by the proteasome. CRLs are divided into seven families based on their cullin constituent. Each cullin binds a RING domain protein, and a vast repertoire of adaptor/substrate receptor modules, collectively creating more than 200 distinct CRLs. All CRLs are regulated by the COP9 signalosome (CSN), an eight-protein isopeptidase that removes the covalently attached activator, NEDD8, from the cullin. Independent of NEDD8 cleavage, CSN forms protective complexes with CRLs, which prevents destructive auto-ubiquitination.
The integrity of the CSN-CRL system is crucially important for the normal cell physiology. Based on our previous work on CRL structures (Fischer, et al., Nature 2014; Fischer, et al., Cell 2011) and that of isolated CSN (Lingaraju et al., Nature 2014), We now intend to provide the underlying molecular mechanism of CRL regulation by CSN. Structural insights at atomic resolution, combined with in vitro and in vivo functional studies are designed to reveal (i) how the signalosome deneddylates and maintains the bound ligases in an inactive state, (ii) how the multiple CSN subunits bind to structurally diverse CRLs, and (iii) how CSN is itself subject to regulation by post-translational modifications or additional further factors.
The ERC funding would allow my lab to pursue an ambitious interdisciplinary approach combining X-ray crystallography, cryo-electron microscopy, biochemistry and cell biology. This is expected to provide a unique molecular understanding of CSN action. Beyond ubiquitination, insight into this >13- subunit CSN-CRL assembly will allow examining general principles of multi-subunit complex action and reveal how the numerous, often essential, subunits contribute to complex function.
Summary
Specificity in the ubiquitin-proteasome system is largely conferred by ubiquitin E3 ligases (E3s). Cullin-RING ligases (CRLs), constituting ~30% of all E3s in humans, mediate the ubiquitination of ~20% of the proteins degraded by the proteasome. CRLs are divided into seven families based on their cullin constituent. Each cullin binds a RING domain protein, and a vast repertoire of adaptor/substrate receptor modules, collectively creating more than 200 distinct CRLs. All CRLs are regulated by the COP9 signalosome (CSN), an eight-protein isopeptidase that removes the covalently attached activator, NEDD8, from the cullin. Independent of NEDD8 cleavage, CSN forms protective complexes with CRLs, which prevents destructive auto-ubiquitination.
The integrity of the CSN-CRL system is crucially important for the normal cell physiology. Based on our previous work on CRL structures (Fischer, et al., Nature 2014; Fischer, et al., Cell 2011) and that of isolated CSN (Lingaraju et al., Nature 2014), We now intend to provide the underlying molecular mechanism of CRL regulation by CSN. Structural insights at atomic resolution, combined with in vitro and in vivo functional studies are designed to reveal (i) how the signalosome deneddylates and maintains the bound ligases in an inactive state, (ii) how the multiple CSN subunits bind to structurally diverse CRLs, and (iii) how CSN is itself subject to regulation by post-translational modifications or additional further factors.
The ERC funding would allow my lab to pursue an ambitious interdisciplinary approach combining X-ray crystallography, cryo-electron microscopy, biochemistry and cell biology. This is expected to provide a unique molecular understanding of CSN action. Beyond ubiquitination, insight into this >13- subunit CSN-CRL assembly will allow examining general principles of multi-subunit complex action and reveal how the numerous, often essential, subunits contribute to complex function.
Max ERC Funding
2 200 677 €
Duration
Start date: 2016-01-01, End date: 2021-02-28
Project acronym CUSTOMER
Project Customizable Embedded Real-Time Systems: Challenges and Key Techniques
Researcher (PI) Yi WANG
Host Institution (HI) UPPSALA UNIVERSITET
Country Sweden
Call Details Advanced Grant (AdG), PE6, ERC-2018-ADG
Summary Today, many industrial products are defined by software and therefore customizable: their functionalities implemented by software can be modified and extended by dynamic software updates on demand. This trend towards customizable products is rapidly expanding into all domains of IT, including Embedded Real-Time Systems (ERTS) deployed in Cyber-Physical Systems such as cars, medical devices etc. However, the current state-of-practice in safety-critical systems allows hardly any modifications once they are put in operation. The lack of techniques to preserve crucial safety conditions for customizable systems severely restricts the benefits of advances in software-defined systems engineering.
CUSTOMER is to provide the missing paradigm and technology for building and updating ERTS after deployment – subject to stringent timing constraints, dynamic workloads, and limited resources on complex platforms. CUSTOMER explores research areas crossing two fields: Real-Time Computing and Formal Verification to develop the key techniques enabling (1) dynamic updates of ERTS in the field, (2) incremental updates over the products life time and (3) safe updates by verification to avoid updates that may compromise system safety.
CUSTOMER will develop a unified model-based framework supported with tools for the design, modelling, verification, deployment and update of ERTS, aiming at advancing the research fields by establishing the missing scientific foundation for multiprocessor real-time computing and providing the next generation of design tools with significantly enhanced capability and scalability increased by orders of magnitude compared with state-of-the-art tools e.g. UPPAAL.
Summary
Today, many industrial products are defined by software and therefore customizable: their functionalities implemented by software can be modified and extended by dynamic software updates on demand. This trend towards customizable products is rapidly expanding into all domains of IT, including Embedded Real-Time Systems (ERTS) deployed in Cyber-Physical Systems such as cars, medical devices etc. However, the current state-of-practice in safety-critical systems allows hardly any modifications once they are put in operation. The lack of techniques to preserve crucial safety conditions for customizable systems severely restricts the benefits of advances in software-defined systems engineering.
CUSTOMER is to provide the missing paradigm and technology for building and updating ERTS after deployment – subject to stringent timing constraints, dynamic workloads, and limited resources on complex platforms. CUSTOMER explores research areas crossing two fields: Real-Time Computing and Formal Verification to develop the key techniques enabling (1) dynamic updates of ERTS in the field, (2) incremental updates over the products life time and (3) safe updates by verification to avoid updates that may compromise system safety.
CUSTOMER will develop a unified model-based framework supported with tools for the design, modelling, verification, deployment and update of ERTS, aiming at advancing the research fields by establishing the missing scientific foundation for multiprocessor real-time computing and providing the next generation of design tools with significantly enhanced capability and scalability increased by orders of magnitude compared with state-of-the-art tools e.g. UPPAAL.
Max ERC Funding
2 499 894 €
Duration
Start date: 2019-10-01, End date: 2024-09-30
Project acronym CyberCare
Project Integrated Sensing Architectures and Tools for Health Care
Researcher (PI) Giovanni De Micheli
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), PE6, ERC-2014-ADG
Summary This proposal addresses high-risk, high-reward research of integrated sensing and computing architectures, as well as of models, methods and tools for their design and operation. Such architectures provide the bridge between bio-systems and information processing systems, where a bio-system is an abstraction of a human in terms of biophysical parameters. Breakthroughs in data acquisition, processing and decision making support will enable new smart-health applications.
The essential research goals of this proposal are: biophysical data acquisition by novel programmable integrated sensor arrays and their design and test using a modular and structured architecture; data processing in situ and/or remotely using application-specific hardware and/or embedded software; a new robust synthesis methodology for data processing units based on a new logic structure; models, abstractions and software tools for reasoning about the acquired data, to validate health conditions and/or to provide remedies (i.e., therapy). The results of this research will be embodied in a demonstrator showing the effectiveness of these combined technologies in first-aid medical care.
The outcome of this research will have a deep and broad impact on health care, because it will improve diagnosis and therapy in a variety of cases. Namely, it will boost the quality and quantity of the acquired biophysical data, possibly in real time, by leveraging multiple sensing modalities and dedicated computing architectures. The use of formal methods for design, data evaluation and decision making support will enhance the quality of the diagnostic platforms and will ease their qualification and adoption. Moreover, the integration of sensing and electronics and their in-field programmability will reduce production cost and lower the barrier of adoption, thus providing for better and more affordable health care means.
Summary
This proposal addresses high-risk, high-reward research of integrated sensing and computing architectures, as well as of models, methods and tools for their design and operation. Such architectures provide the bridge between bio-systems and information processing systems, where a bio-system is an abstraction of a human in terms of biophysical parameters. Breakthroughs in data acquisition, processing and decision making support will enable new smart-health applications.
The essential research goals of this proposal are: biophysical data acquisition by novel programmable integrated sensor arrays and their design and test using a modular and structured architecture; data processing in situ and/or remotely using application-specific hardware and/or embedded software; a new robust synthesis methodology for data processing units based on a new logic structure; models, abstractions and software tools for reasoning about the acquired data, to validate health conditions and/or to provide remedies (i.e., therapy). The results of this research will be embodied in a demonstrator showing the effectiveness of these combined technologies in first-aid medical care.
The outcome of this research will have a deep and broad impact on health care, because it will improve diagnosis and therapy in a variety of cases. Namely, it will boost the quality and quantity of the acquired biophysical data, possibly in real time, by leveraging multiple sensing modalities and dedicated computing architectures. The use of formal methods for design, data evaluation and decision making support will enhance the quality of the diagnostic platforms and will ease their qualification and adoption. Moreover, the integration of sensing and electronics and their in-field programmability will reduce production cost and lower the barrier of adoption, thus providing for better and more affordable health care means.
Max ERC Funding
2 086 740 €
Duration
Start date: 2016-01-01, End date: 2021-06-30
Project acronym CyberGenetics
Project Cybergenetics: Theory and Design Tools for Biomolecular Control Systems
Researcher (PI) Mustafa KHAMMASH
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Country Switzerland
Call Details Advanced Grant (AdG), PE7, ERC-2016-ADG
Summary We propose to develop a new theory and design tools for the estimation and real-time control of living cells. The control systems designed using these tools will precisely and robustly steer the dynamic behavior of living cells in real time to achieve desired objectives. Cells would be controlled either collectively at the population level, or individually as single cells. The control systems achieving this regulation will be realized either on a digital computer that is interfaced with living cells, or using de novo genetic circuits that are introduced into the cells where they are designed to function as molecular control systems. Our methods will explicitly confront the numerous challenges brought about by the special environment of the cell including nonlinearity, stochasticity, cell-to-cell variability, metabolic burden, etc. The theory and methods developed in this project will thus enable the systematic, rational, and effective feedback control of living cells at the gene level, and will lay the foundation for a new corresponding body of knowledge which we call ``Cybergenetics''. It will also open new research directions in the areas of control theory and estimation.
We also propose to design three cybergenetic control systems, each addressing an important application in biotechnology or therapeutics. In the first, the controller will use light and nutrient supply to precisely regulate gene expression and cell growth in E. coli to achieve high protein and low biomass production rates. The second involves multiple feedback controllers regulating in parallel a large number of single stem cells, and leading to their differentiation to desired fates, e.g. beta cells, with potential for therapeutic applications. Finally, we will engineer into living cells dynamic molecular control systems. Such controllers can be used to monitor physiological variables and secrete biological effectors in a feedback fashion for the treatment of diseases like Type 1 diabetes.
Summary
We propose to develop a new theory and design tools for the estimation and real-time control of living cells. The control systems designed using these tools will precisely and robustly steer the dynamic behavior of living cells in real time to achieve desired objectives. Cells would be controlled either collectively at the population level, or individually as single cells. The control systems achieving this regulation will be realized either on a digital computer that is interfaced with living cells, or using de novo genetic circuits that are introduced into the cells where they are designed to function as molecular control systems. Our methods will explicitly confront the numerous challenges brought about by the special environment of the cell including nonlinearity, stochasticity, cell-to-cell variability, metabolic burden, etc. The theory and methods developed in this project will thus enable the systematic, rational, and effective feedback control of living cells at the gene level, and will lay the foundation for a new corresponding body of knowledge which we call ``Cybergenetics''. It will also open new research directions in the areas of control theory and estimation.
We also propose to design three cybergenetic control systems, each addressing an important application in biotechnology or therapeutics. In the first, the controller will use light and nutrient supply to precisely regulate gene expression and cell growth in E. coli to achieve high protein and low biomass production rates. The second involves multiple feedback controllers regulating in parallel a large number of single stem cells, and leading to their differentiation to desired fates, e.g. beta cells, with potential for therapeutic applications. Finally, we will engineer into living cells dynamic molecular control systems. Such controllers can be used to monitor physiological variables and secrete biological effectors in a feedback fashion for the treatment of diseases like Type 1 diabetes.
Max ERC Funding
2 499 887 €
Duration
Start date: 2017-08-01, End date: 2022-07-31
Project acronym CYCLODE
Project Cyclical and Linear Timing Modes in Development
Researcher (PI) Helge GROSSHANS
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Country Switzerland
Call Details Advanced Grant (AdG), LS3, ERC-2016-ADG
Summary Organismal development requires proper timing of events such as cell fate choices, but the mechanisms that control temporal patterning remain poorly understood. In particular, we know little of the cyclical timers, or ‘clocks’, that control recurring events such as vertebrate segmentation or nematode molting. Furthermore, it is unknown how cyclical timers are coordinated with the global, or linear, timing of development, e.g. to ensure an appropriate number of cyclical repeats. We propose to elucidate the components, wiring, and properties of a prototypic developmental clock by studying developmental timing in the roundworm C. elegans. We build on our recent discovery that nearly 20% of the worm’s transcriptome oscillates during larval development – an apparent manifestation of a clock that times the various recurring events that encompass each larval stage. Our aims are i) to identify components of this clock using genetic screens, ii) to gain insight into the system’s architecture and properties by employing specific perturbations such as food deprivation, and iii) to understand the coupling of this cyclic clock to the linear heterochronic timer through genetic manipulations. To achieve our ambitious goals, we will develop tools for mRNA sequencing of individual worms and for their developmental tracking and microchamber-based imaging. These important advances will increase temporal resolution, enhance signal-to-noise ratio, and achieve live tracking of oscillations in vivo. Our combination of genetic, genomic, imaging, and computational approaches will provide a detailed understanding of this clock, and biological timing mechanisms in general. As heterochronic genes and rhythmic gene expression are also important for controlling stem cell fates, we foresee that the results gained will additionally reveal regulatory mechanisms of stem cells, thus advancing our fundamental understanding of animal development and future applications in regenerative medicine.
Summary
Organismal development requires proper timing of events such as cell fate choices, but the mechanisms that control temporal patterning remain poorly understood. In particular, we know little of the cyclical timers, or ‘clocks’, that control recurring events such as vertebrate segmentation or nematode molting. Furthermore, it is unknown how cyclical timers are coordinated with the global, or linear, timing of development, e.g. to ensure an appropriate number of cyclical repeats. We propose to elucidate the components, wiring, and properties of a prototypic developmental clock by studying developmental timing in the roundworm C. elegans. We build on our recent discovery that nearly 20% of the worm’s transcriptome oscillates during larval development – an apparent manifestation of a clock that times the various recurring events that encompass each larval stage. Our aims are i) to identify components of this clock using genetic screens, ii) to gain insight into the system’s architecture and properties by employing specific perturbations such as food deprivation, and iii) to understand the coupling of this cyclic clock to the linear heterochronic timer through genetic manipulations. To achieve our ambitious goals, we will develop tools for mRNA sequencing of individual worms and for their developmental tracking and microchamber-based imaging. These important advances will increase temporal resolution, enhance signal-to-noise ratio, and achieve live tracking of oscillations in vivo. Our combination of genetic, genomic, imaging, and computational approaches will provide a detailed understanding of this clock, and biological timing mechanisms in general. As heterochronic genes and rhythmic gene expression are also important for controlling stem cell fates, we foresee that the results gained will additionally reveal regulatory mechanisms of stem cells, thus advancing our fundamental understanding of animal development and future applications in regenerative medicine.
Max ERC Funding
2 358 625 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym DEATHSWITCHING
Project Identifying genes and pathways that drive molecular switches and back-up mechanisms between apoptosis and autophagy
Researcher (PI) Adi Kimchi
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Country Israel
Call Details Advanced Grant (AdG), LS3, ERC-2012-ADG_20120314
Summary A cell’s decision to die is governed by multiple input signals received from a complex network of programmed cell death (PCD) pathways, including apoptosis and programmed necrosis. Additionally, under some conditions, autophagy, whose function is mainly pro-survival, may act as a back-up death pathway. We propose to apply new approaches to study the molecular basis of two important questions that await resolution in the field: a) how the cell switches from a pro-survival autophagic response to an apoptotic response and b) whether and how pro-survival autophagy is converted to a death mechanism when apoptosis is blocked. To address the first issue, we will screen for direct physical interactions between autophagic and apoptotic proteins, using the protein fragment complementation assay. Validated pairs will be studied in depth to identify built-in molecular switches that activate apoptosis when autophagy fails to restore homeostasis. As a pilot case to address the concept of molecular ‘sensors’ and ‘switches’, we will focus on the previously identified Atg12/Bcl-2 interaction. In the second line of research we will categorize autophagy-dependent cell death triggers into those that directly result from autophagy-dependent degradation, either by excessive self-digestion or by selective protein degradation, and those that utilize the autophagy machinery to activate programmed necrosis. We will identify the genes regulating these scenarios by whole genome RNAi screens for increased cell survival. In parallel, we will use a cell library of annotated fluorescent-tagged proteins for measuring selective protein degradation. These will be the starting point for identification of the molecular pathways that convert survival autophagy to a death program. Finally, we will explore the physiological relevance of back-up death mechanisms and the newly identified molecular mechanisms to developmental PCD during the cavitation process in early stages of embryogenesis.
Summary
A cell’s decision to die is governed by multiple input signals received from a complex network of programmed cell death (PCD) pathways, including apoptosis and programmed necrosis. Additionally, under some conditions, autophagy, whose function is mainly pro-survival, may act as a back-up death pathway. We propose to apply new approaches to study the molecular basis of two important questions that await resolution in the field: a) how the cell switches from a pro-survival autophagic response to an apoptotic response and b) whether and how pro-survival autophagy is converted to a death mechanism when apoptosis is blocked. To address the first issue, we will screen for direct physical interactions between autophagic and apoptotic proteins, using the protein fragment complementation assay. Validated pairs will be studied in depth to identify built-in molecular switches that activate apoptosis when autophagy fails to restore homeostasis. As a pilot case to address the concept of molecular ‘sensors’ and ‘switches’, we will focus on the previously identified Atg12/Bcl-2 interaction. In the second line of research we will categorize autophagy-dependent cell death triggers into those that directly result from autophagy-dependent degradation, either by excessive self-digestion or by selective protein degradation, and those that utilize the autophagy machinery to activate programmed necrosis. We will identify the genes regulating these scenarios by whole genome RNAi screens for increased cell survival. In parallel, we will use a cell library of annotated fluorescent-tagged proteins for measuring selective protein degradation. These will be the starting point for identification of the molecular pathways that convert survival autophagy to a death program. Finally, we will explore the physiological relevance of back-up death mechanisms and the newly identified molecular mechanisms to developmental PCD during the cavitation process in early stages of embryogenesis.
Max ERC Funding
2 500 000 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym DeepInternal
Project Going Deep and Blind with Internal Statistics
Researcher (PI) Michal IRANI
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Country Israel
Call Details Advanced Grant (AdG), PE6, ERC-2017-ADG
Summary Unsupervised visual inference can often be performed by exploiting the internal redundancy inside a single visual datum (an image or a video). The strong repetition of patches inside a single image/video provides a powerful data-specific prior for solving a variety of vision tasks in a “blind” manner: (i) Blind in the sense that sophisticated unsupervised inferences can be made with no prior examples or training; (ii) Blind in the sense that complex ill-posed Inverse-Problems can be solved, even when the forward degradation is unknown.
While the above fully unsupervised approach achieved impressive results, it relies on internal data alone, hence cannot enjoy the “wisdom of the crowd” which Deep-Learning (DL) so wisely extracts from external collections of images, yielding state-of-the-art (SOTA) results. Nevertheless, DL requires huge amounts of training data, which restricts its applicability. Moreover, some internal image-specific information, which is clearly visible, remains unexploited by today's DL methods. One such example is shown in Fig.1.
We propose to combine the power of these two complementary approaches – unsupervised Internal Data Recurrence, with Deep Learning, to obtain the best of both worlds. If successful, this will have several important outcomes including:
• A wide range of low-level & high-level inferences (image & video).
• A continuum between Internal & External training – a platform to explore theoretical and practical tradeoffs between amount of available training data and optimal Internal-vs-External training.
• Enable totally unsupervised DL when no training data are available.
• Enable supervised DL with modest amounts of training data.
• New applications, disciplines and domains, which are enabled by the unified approach.
• A platform for substantial progress in video analysis (which has been lagging behind so far due to the strong reliance on exhaustive supervised training data).
Summary
Unsupervised visual inference can often be performed by exploiting the internal redundancy inside a single visual datum (an image or a video). The strong repetition of patches inside a single image/video provides a powerful data-specific prior for solving a variety of vision tasks in a “blind” manner: (i) Blind in the sense that sophisticated unsupervised inferences can be made with no prior examples or training; (ii) Blind in the sense that complex ill-posed Inverse-Problems can be solved, even when the forward degradation is unknown.
While the above fully unsupervised approach achieved impressive results, it relies on internal data alone, hence cannot enjoy the “wisdom of the crowd” which Deep-Learning (DL) so wisely extracts from external collections of images, yielding state-of-the-art (SOTA) results. Nevertheless, DL requires huge amounts of training data, which restricts its applicability. Moreover, some internal image-specific information, which is clearly visible, remains unexploited by today's DL methods. One such example is shown in Fig.1.
We propose to combine the power of these two complementary approaches – unsupervised Internal Data Recurrence, with Deep Learning, to obtain the best of both worlds. If successful, this will have several important outcomes including:
• A wide range of low-level & high-level inferences (image & video).
• A continuum between Internal & External training – a platform to explore theoretical and practical tradeoffs between amount of available training data and optimal Internal-vs-External training.
• Enable totally unsupervised DL when no training data are available.
• Enable supervised DL with modest amounts of training data.
• New applications, disciplines and domains, which are enabled by the unified approach.
• A platform for substantial progress in video analysis (which has been lagging behind so far due to the strong reliance on exhaustive supervised training data).
Max ERC Funding
2 466 940 €
Duration
Start date: 2018-05-01, End date: 2023-10-31
Project acronym DEPICT
Project Design principles and controllability of protein circuits
Researcher (PI) Uri Alon
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Country Israel
Call Details Advanced Grant (AdG), LS2, ERC-2009-AdG
Summary Cells use circuits of interacting proteins to respond to their environment. In the past decades, molecular biology has provided detailed knowledge on the proteins in these circuits and their interactions. To fully understand circuit function requires, in addition to molecular knowledge, new concepts that explain how multiple components work together to perform systems level functions. Our lab has been a leader in defining such concepts, based on combined experimental and theoretical study of well characterized circuits in bacteria and human cells. In this proposal we aim to find novel principles on how circuits resist fluctuations and errors, and how they can be controlled by drugs: (1) Why do key regulatory systems use bifunctional enzymes that catalyze antagonistic reactions (e.g. both kinase and phosphatase)? We will test the role of bifunctional enzymes in making circuits robust to variations in protein levels. (2) Why are some genes regulated by a repressor and others by an activator? We will test this in the context of reduction of errors in transcription control. (3) Are there principles that describe how drugs combine to affect protein dynamics in human cells? We will use a novel dynamic proteomics approach developed in our lab to explore how protein dynamics can be controlled by drug combinations. This research will define principles that unite our understanding of seemingly distinct biological systems, and explain their particular design in terms of systems-level functions. This understanding will help form the basis for a future medicine that rationally controls the state of the cell based on a detailed blueprint of their circuit design, and quantitative principles for the effects of drugs on this circuitry.
Summary
Cells use circuits of interacting proteins to respond to their environment. In the past decades, molecular biology has provided detailed knowledge on the proteins in these circuits and their interactions. To fully understand circuit function requires, in addition to molecular knowledge, new concepts that explain how multiple components work together to perform systems level functions. Our lab has been a leader in defining such concepts, based on combined experimental and theoretical study of well characterized circuits in bacteria and human cells. In this proposal we aim to find novel principles on how circuits resist fluctuations and errors, and how they can be controlled by drugs: (1) Why do key regulatory systems use bifunctional enzymes that catalyze antagonistic reactions (e.g. both kinase and phosphatase)? We will test the role of bifunctional enzymes in making circuits robust to variations in protein levels. (2) Why are some genes regulated by a repressor and others by an activator? We will test this in the context of reduction of errors in transcription control. (3) Are there principles that describe how drugs combine to affect protein dynamics in human cells? We will use a novel dynamic proteomics approach developed in our lab to explore how protein dynamics can be controlled by drug combinations. This research will define principles that unite our understanding of seemingly distinct biological systems, and explain their particular design in terms of systems-level functions. This understanding will help form the basis for a future medicine that rationally controls the state of the cell based on a detailed blueprint of their circuit design, and quantitative principles for the effects of drugs on this circuitry.
Max ERC Funding
2 261 440 €
Duration
Start date: 2010-03-01, End date: 2015-02-28
Project acronym Descent
Project Control of Action Diversification by Descending Motor CircuitsControl of action diversification by descending motor circuits
Researcher (PI) Silvia Arber
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Country Switzerland
Call Details Advanced Grant (AdG), LS5, ERC-2015-AdG
Summary Movement is the behavioral output of the nervous system. Animals carry out an enormous repertoire of distinct actions, spanning from seemingly simple repetitive tasks like walking to much more complex movements such as forelimb manipulation tasks. An important question is how neuronal circuits are organized and function to choose, maintain, adjust and terminate these many distinct motor behaviors. Recent technological advances in neuroscience have made it possible to begin to unravel the links between the organization of specific neuronal circuit elements in the CNS and the control of movement, a topic that will be central to this research program.
While past work proposes that supraspinal centers in the brainstem are instrumental to the control of action diversification, little is known about how brainstem circuits translate movement intention to body control, how competing motor programs are selected, and how behavioral state influences movement control. The goal of this research project is to unravel the circuit blueprint of mouse descending motor pathways at a fine-scale level and to probe the intersection between revealed circuit organization and their behavioral function at many levels. The focus will be on studies on the interactions between brainstem neurons and spinal circuits to determine how initiation, duration, termination and selection of motor programs are implemented through specific neuronal subpopulations. Mapping descending connectivity matrices of motor circuits will serve as entry point and we will make use of state-of-the art intersectional technology including mouse genetics, viral approaches, in vivo neuronal recordings and activity manipulations of specific neuronal populations during behavior. Together, our project will elucidate the circuit organization and function of the descending motor output system and thereby uncover principles of how the nervous system generates diverse actions.
Summary
Movement is the behavioral output of the nervous system. Animals carry out an enormous repertoire of distinct actions, spanning from seemingly simple repetitive tasks like walking to much more complex movements such as forelimb manipulation tasks. An important question is how neuronal circuits are organized and function to choose, maintain, adjust and terminate these many distinct motor behaviors. Recent technological advances in neuroscience have made it possible to begin to unravel the links between the organization of specific neuronal circuit elements in the CNS and the control of movement, a topic that will be central to this research program.
While past work proposes that supraspinal centers in the brainstem are instrumental to the control of action diversification, little is known about how brainstem circuits translate movement intention to body control, how competing motor programs are selected, and how behavioral state influences movement control. The goal of this research project is to unravel the circuit blueprint of mouse descending motor pathways at a fine-scale level and to probe the intersection between revealed circuit organization and their behavioral function at many levels. The focus will be on studies on the interactions between brainstem neurons and spinal circuits to determine how initiation, duration, termination and selection of motor programs are implemented through specific neuronal subpopulations. Mapping descending connectivity matrices of motor circuits will serve as entry point and we will make use of state-of-the art intersectional technology including mouse genetics, viral approaches, in vivo neuronal recordings and activity manipulations of specific neuronal populations during behavior. Together, our project will elucidate the circuit organization and function of the descending motor output system and thereby uncover principles of how the nervous system generates diverse actions.
Max ERC Funding
2 500 000 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym DG-PESP-CS
Project Deterministic Generation of Polarization Entangled single Photons Cluster States
Researcher (PI) David Gershoni
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Country Israel
Call Details Advanced Grant (AdG), PE2, ERC-2015-AdG
Summary Measurement based quantum computing is one of the most fault-tolerant architectures proposed for quantum information processing. It opens the possibility of performing quantum computing tasks using linear optical systems. An efficient route for measurement based quantum computing utilizes highly entangled states of photons, called cluster states. Propagation and processing quantum information is made possible this way using only single qubit measurements. It is highly resilient to qubit losses. In addition, single qubit measurements of polarization qubits is easily performed with high fidelity using standard optical tools. These features make photonic clusters excellent platforms for quantum information processing.
Constructing photonic cluster states, however, is a formidable challenge, attracting vast amounts of research efforts. While in principle it is possible to build up cluster states using interferometry, such a method is of a probabilistic nature and entails a large overhead of resources. The use of entangled photon pairs reduces this overhead by a small factor only.
We outline a novel route for constructing a deterministic source of photonic cluster states using a device based on semiconductor quantum dot. Our proposal follows a suggestion by Lindner and Rudolph. We use repeated optical excitations of a long lived coherent spin confined in a single semiconductor quantum dot and demonstrate for the first time practical realization of their proposal. Our preliminary demonstration presents a breakthrough in quantum technology since deterministic source of photonic cluster, reduces the resources needed quantum information processing. It may have revolutionary prospects for technological applications as well as to our fundamental understanding of quantum systems.
We propose to capitalize on this recent breakthrough and concentrate on R&D which will further advance this forefront field of science and technology by utilizing the horizons that it opens.
Summary
Measurement based quantum computing is one of the most fault-tolerant architectures proposed for quantum information processing. It opens the possibility of performing quantum computing tasks using linear optical systems. An efficient route for measurement based quantum computing utilizes highly entangled states of photons, called cluster states. Propagation and processing quantum information is made possible this way using only single qubit measurements. It is highly resilient to qubit losses. In addition, single qubit measurements of polarization qubits is easily performed with high fidelity using standard optical tools. These features make photonic clusters excellent platforms for quantum information processing.
Constructing photonic cluster states, however, is a formidable challenge, attracting vast amounts of research efforts. While in principle it is possible to build up cluster states using interferometry, such a method is of a probabilistic nature and entails a large overhead of resources. The use of entangled photon pairs reduces this overhead by a small factor only.
We outline a novel route for constructing a deterministic source of photonic cluster states using a device based on semiconductor quantum dot. Our proposal follows a suggestion by Lindner and Rudolph. We use repeated optical excitations of a long lived coherent spin confined in a single semiconductor quantum dot and demonstrate for the first time practical realization of their proposal. Our preliminary demonstration presents a breakthrough in quantum technology since deterministic source of photonic cluster, reduces the resources needed quantum information processing. It may have revolutionary prospects for technological applications as well as to our fundamental understanding of quantum systems.
We propose to capitalize on this recent breakthrough and concentrate on R&D which will further advance this forefront field of science and technology by utilizing the horizons that it opens.
Max ERC Funding
2 502 974 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
Project acronym DHISP
Project Dorsal Horn Interneurons in Sensory Processing
Researcher (PI) Hanns Ulrich Zeilhofer
Host Institution (HI) University of Zurich
Country Switzerland
Call Details Advanced Grant (AdG), LS5, ERC-2009-AdG
Summary Chronic pain syndromes are to a large extent due to maladaptive plastic changes in the CNS. A CNS area particularly relevant for such changes is the spinal dorsal horn, where inputs from nociceptive and non-nociceptive fibers undergo their first synaptic integration. This area harbors a sophisticated network of interneurons, which function as a gate-control unit for incoming sensory signals. Several different types of interneurons can be distinguished based e.g. on their neurotransmitter and neuropeptide content. Despite more than 40 years of research, our knowledge about the integration of these neurons in dorsal horn circuits and their contribution to sensory processing is still very limited. This proposal aims at a comprehensive characterization of the dorsal horn neuronal network under normal conditions and in chronic pain states with a focus on inhibitory interneurons. A genome-wide analysis of the gene expression profile shall be made from defined dorsal horn interneurons genetically tagged with fluorescent markers and isolated by fluorescence activated cell sorting. A functional characterization of the connectivity of these neurons in spinal cord slices and of their role in in vivo sensory processing shall be achieved with optogenetic tools (channelrhodopsin-2), which permit activation of these neurons with light. Finally, behavioral analyses shall be made in mice after diphteria toxin-mediated ablation of defined interneuron types. All three approaches shall be applied to naïve mice and to mice with inflammatory or neuropathic pain. The results from these studies will improve our understanding of the malfunctioning of sensory processing in chronic pain states and will provide the basis for novel approaches to the prevention or reversal of chronic pain states.
Summary
Chronic pain syndromes are to a large extent due to maladaptive plastic changes in the CNS. A CNS area particularly relevant for such changes is the spinal dorsal horn, where inputs from nociceptive and non-nociceptive fibers undergo their first synaptic integration. This area harbors a sophisticated network of interneurons, which function as a gate-control unit for incoming sensory signals. Several different types of interneurons can be distinguished based e.g. on their neurotransmitter and neuropeptide content. Despite more than 40 years of research, our knowledge about the integration of these neurons in dorsal horn circuits and their contribution to sensory processing is still very limited. This proposal aims at a comprehensive characterization of the dorsal horn neuronal network under normal conditions and in chronic pain states with a focus on inhibitory interneurons. A genome-wide analysis of the gene expression profile shall be made from defined dorsal horn interneurons genetically tagged with fluorescent markers and isolated by fluorescence activated cell sorting. A functional characterization of the connectivity of these neurons in spinal cord slices and of their role in in vivo sensory processing shall be achieved with optogenetic tools (channelrhodopsin-2), which permit activation of these neurons with light. Finally, behavioral analyses shall be made in mice after diphteria toxin-mediated ablation of defined interneuron types. All three approaches shall be applied to naïve mice and to mice with inflammatory or neuropathic pain. The results from these studies will improve our understanding of the malfunctioning of sensory processing in chronic pain states and will provide the basis for novel approaches to the prevention or reversal of chronic pain states.
Max ERC Funding
2 467 000 €
Duration
Start date: 2010-05-01, End date: 2016-04-30
Project acronym DNAccess
Project Overcoming chromatin restricted DNA access
Researcher (PI) dirk SCHUBELER
Host Institution (HI) FRIEDRICH MIESCHER INSTITUTE FOR BIOMEDICAL RESEARCH FONDATION
Country Switzerland
Call Details Advanced Grant (AdG), LS2, ERC-2019-ADG
Summary Differential gene expression programs account for the diversity of cell types in our body. They are controlled by intricate networks of sequence-specific transcription factors (TFs) that operate in the context of chromatin. Chromatin is itself an essential component of this process, and is part of the system that selectively restricts DNA access for TFs. As a consequence, most TFs only bind to a small subset of their motif occurrences, in a way we currently do not fully understand. Our inability to predict binding of a TF based on its cognate motif and location in the genome is a serious obstacle towards predictive models of gene regulation.
DNAccess has the ambitious goal to define in vivo the sensitivities of TFs to nucleosomes and their reliance on chromatin remodeling enzymes for binding. Using novel genomics and genome editing tools we will: (a) systematically vary TF motifs, presence of nucleosomes and their modifications at a defined chromosomal locus and quantify resulting TF binding; (b) explore genome-wide the ability of ectopic TFs to engage with a chromatinized genome in the absence of host cofactor engagement; (c) dissect chromatin remodeler dependent TF binding in order to define temporal order and subcomplex function and (d) deconstruct remodeler recruitment by mutating TF interaction domains.
DNAccess will build a highly integrated setup to comprehensively characterize how nucleosomes, their modifications and mobility restricts genome access. We will characterize existing chromatin barriers and identify how TFs overcome them. This represents a crucial step towards a comprehensive understanding of the role of chromatin in gene regulation, and will advance our understanding of how specificity is generated in large eukaryotic genomes.
Summary
Differential gene expression programs account for the diversity of cell types in our body. They are controlled by intricate networks of sequence-specific transcription factors (TFs) that operate in the context of chromatin. Chromatin is itself an essential component of this process, and is part of the system that selectively restricts DNA access for TFs. As a consequence, most TFs only bind to a small subset of their motif occurrences, in a way we currently do not fully understand. Our inability to predict binding of a TF based on its cognate motif and location in the genome is a serious obstacle towards predictive models of gene regulation.
DNAccess has the ambitious goal to define in vivo the sensitivities of TFs to nucleosomes and their reliance on chromatin remodeling enzymes for binding. Using novel genomics and genome editing tools we will: (a) systematically vary TF motifs, presence of nucleosomes and their modifications at a defined chromosomal locus and quantify resulting TF binding; (b) explore genome-wide the ability of ectopic TFs to engage with a chromatinized genome in the absence of host cofactor engagement; (c) dissect chromatin remodeler dependent TF binding in order to define temporal order and subcomplex function and (d) deconstruct remodeler recruitment by mutating TF interaction domains.
DNAccess will build a highly integrated setup to comprehensively characterize how nucleosomes, their modifications and mobility restricts genome access. We will characterize existing chromatin barriers and identify how TFs overcome them. This represents a crucial step towards a comprehensive understanding of the role of chromatin in gene regulation, and will advance our understanding of how specificity is generated in large eukaryotic genomes.
Max ERC Funding
2 302 500 €
Duration
Start date: 2021-03-01, End date: 2026-02-28
Project acronym DOPPLER
Project Domain-optimised parallelisation by polymorphic language embeddings and rewritings
Researcher (PI) Martin Maria Anton Nikolaus Odersky
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), PE6, ERC-2010-AdG_20100224
Summary Concurrent and parallel programming are becoming indispensable for
exploiting modern hardware. Because possible speed increases of single
processors have reached their limit, increasing transistor count will
yield more, but not necessarily faster cores, and this for the
foreseeable future. This means that, from now on, parallelism in
software will have to double every 18 months to keep up with
hardware. This problem has been identified as the ``Popular Parallel
Programming'' grand challenge by the computer architecture community.
The proposed project will research new ways to solve this challenge.
We start with a set of domain-specific languages which naturally admit
a high degree of parallelism. The domain specific languages are
integrated in a common host language using polymorphic language
embeddings. Such embeddings provide a high degree of abstraction,
which gives considerable freedom in their actual representation and
implementation. The new direction taken by this proposal is to combine
polymorphic embeddings with optimizing rewritings in a staged
compilation process. This can lead to highly parallel and efficient
implementations on a variety of heterogeneous hardware.
Summary
Concurrent and parallel programming are becoming indispensable for
exploiting modern hardware. Because possible speed increases of single
processors have reached their limit, increasing transistor count will
yield more, but not necessarily faster cores, and this for the
foreseeable future. This means that, from now on, parallelism in
software will have to double every 18 months to keep up with
hardware. This problem has been identified as the ``Popular Parallel
Programming'' grand challenge by the computer architecture community.
The proposed project will research new ways to solve this challenge.
We start with a set of domain-specific languages which naturally admit
a high degree of parallelism. The domain specific languages are
integrated in a common host language using polymorphic language
embeddings. Such embeddings provide a high degree of abstraction,
which gives considerable freedom in their actual representation and
implementation. The new direction taken by this proposal is to combine
polymorphic embeddings with optimizing rewritings in a staged
compilation process. This can lead to highly parallel and efficient
implementations on a variety of heterogeneous hardware.
Max ERC Funding
2 392 400 €
Duration
Start date: 2011-04-01, End date: 2016-03-31
Project acronym DROSOPHILASIGNALING
Project Signaling Pathways Controlling Patterning, Growth and Final Size of Drosophila Limbs
Researcher (PI) Konrad Basler
Host Institution (HI) University of Zurich
Country Switzerland
Call Details Advanced Grant (AdG), LS3, ERC-2008-AdG
Summary Developmental biology seeks not only to learn more about the fundamental processes of growth and pattern per se, but to understand how they synergize to enable the morphogenesis of multicellular organisms. Our goal is to perform real-time analyses of these developmental processes in an intact developing organ. By applying a vital imaging approach, we can circumvent the normal limitations of inferring cellular dynamics from static images or molecular data, and obtain the real dynamic view of growth and patterning. The wing imaginal disc of Drosophila, which starts out as a simple epithelial structure and gives rise to a precisely structured adult limb, will serve as an ideal model system. This system has the combined advantages of relative simplicity and genetic tractability. We will create several innovations that expand the current toolkit and thus facilitate the detailed dissection of growth and patterning. A key early step will be to develop novel reporters to dynamically and faithfully monitor signaling cascades involved in growth and patterning, such as the Dpp and Hippo pathways. We will also implement quantification techniques that are currently being set up in collaboration with an experimental physicist, to deduce, and alter, the mechanical forces that develop in the cells of a growing tissue. The large amount of quantitative data that will be generated allow us derive computational models of the individual pathways and their interaction. The focus of the study will be to answer the following questions: 1) Is the Hippo pathway regulated spatially and temporally, and by what signaling pathways? 2) Do mechanical forces play a pivotal controlling role in organ morphogenesis? 3) What are the global effects on growth, when pathways controlling patterning, cell competition or compensatory proliferation are perturbed? The proposed project will bring the approaches taken to define the mechanisms underlying and controlling growth and patterning to the next level.
Summary
Developmental biology seeks not only to learn more about the fundamental processes of growth and pattern per se, but to understand how they synergize to enable the morphogenesis of multicellular organisms. Our goal is to perform real-time analyses of these developmental processes in an intact developing organ. By applying a vital imaging approach, we can circumvent the normal limitations of inferring cellular dynamics from static images or molecular data, and obtain the real dynamic view of growth and patterning. The wing imaginal disc of Drosophila, which starts out as a simple epithelial structure and gives rise to a precisely structured adult limb, will serve as an ideal model system. This system has the combined advantages of relative simplicity and genetic tractability. We will create several innovations that expand the current toolkit and thus facilitate the detailed dissection of growth and patterning. A key early step will be to develop novel reporters to dynamically and faithfully monitor signaling cascades involved in growth and patterning, such as the Dpp and Hippo pathways. We will also implement quantification techniques that are currently being set up in collaboration with an experimental physicist, to deduce, and alter, the mechanical forces that develop in the cells of a growing tissue. The large amount of quantitative data that will be generated allow us derive computational models of the individual pathways and their interaction. The focus of the study will be to answer the following questions: 1) Is the Hippo pathway regulated spatially and temporally, and by what signaling pathways? 2) Do mechanical forces play a pivotal controlling role in organ morphogenesis? 3) What are the global effects on growth, when pathways controlling patterning, cell competition or compensatory proliferation are perturbed? The proposed project will bring the approaches taken to define the mechanisms underlying and controlling growth and patterning to the next level.
Max ERC Funding
2 310 000 €
Duration
Start date: 2009-02-01, End date: 2014-01-31
Project acronym DrosoSpiro
Project The Drosophila-Spiroplasma interaction as a model to dissect the molecular mechanisms underlying insect endosymbiosis
Researcher (PI) Bruno Lemaitre
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS8, ERC-2013-ADG
Summary Virtually every species of insect harbors facultative bacterial endosymbionts that are transmitted from females to their offspring, often in the egg cytoplasm. These symbionts play crucial roles in the biology of their hosts. Many manipulate host reproduction in order to spread within host populations. Others increase the fitness of their hosts under certain conditions. For example, increasing tolerance to heat or protecting their hosts against natural enemies. Over the past decade, our understanding of insect endosymbionts has shifted from seeing them as fascinating oddities to being ubiquitous and central to the biology of their hosts, including many of high economic and medical importance. However, in spite of growing interest in endosymbionts, very little is known about the molecular mechanisms underlying most endosymbiont-insect interactions. For instance, the basis of the main phenotypes caused by endosymbionts, including diverse reproductive manipulations or symbiont-protective immunity, remains largely enigmatic. The goal of the present application is to fill this gap by dissecting the interaction between Drosophila and its native endosymbiont Spiroplasma poulsonii. This project will use a broad range of approaches ranging from molecular genetic to genomics to dissect the molecular mechanisms underlying key features of the symbiosis, including vertical transmission, male killing, regulation of symbiont growth, and symbiont-mediated protection against parasitic wasps. We believe that the fundamental knowledge generated on the Drosophila-Spiroplasma interaction will serve as a paradigm for other endosymbiont-insect interactions that are less amenable to genetic studies.
Summary
Virtually every species of insect harbors facultative bacterial endosymbionts that are transmitted from females to their offspring, often in the egg cytoplasm. These symbionts play crucial roles in the biology of their hosts. Many manipulate host reproduction in order to spread within host populations. Others increase the fitness of their hosts under certain conditions. For example, increasing tolerance to heat or protecting their hosts against natural enemies. Over the past decade, our understanding of insect endosymbionts has shifted from seeing them as fascinating oddities to being ubiquitous and central to the biology of their hosts, including many of high economic and medical importance. However, in spite of growing interest in endosymbionts, very little is known about the molecular mechanisms underlying most endosymbiont-insect interactions. For instance, the basis of the main phenotypes caused by endosymbionts, including diverse reproductive manipulations or symbiont-protective immunity, remains largely enigmatic. The goal of the present application is to fill this gap by dissecting the interaction between Drosophila and its native endosymbiont Spiroplasma poulsonii. This project will use a broad range of approaches ranging from molecular genetic to genomics to dissect the molecular mechanisms underlying key features of the symbiosis, including vertical transmission, male killing, regulation of symbiont growth, and symbiont-mediated protection against parasitic wasps. We believe that the fundamental knowledge generated on the Drosophila-Spiroplasma interaction will serve as a paradigm for other endosymbiont-insect interactions that are less amenable to genetic studies.
Max ERC Funding
1 963 926 €
Duration
Start date: 2014-05-01, End date: 2019-04-30
Project acronym DURABLERESISTANCE
Project Durable resistance against fungal plant pathogens
Researcher (PI) Beat Keller
Host Institution (HI) University of Zurich
Country Switzerland
Call Details Advanced Grant (AdG), LS9, ERC-2009-AdG
Summary Plants and their pathogens are in a constant process of co-evolution. Consequently, many of the known defense genes of plants against fungal pathogens are rapidly loosing effectiveness under agricultural conditions. However, there are examples for durable resistance. It is one of the main research questions in plant biology to determine the genetic basis of such naturally occurring resistance and to understand the underlying biochemical and molecular cause for durability. This durability is characterized by the apparent inability of the pathogen to adapt to the resistance mechanism. The molecular understanding of durable resistance will contribute to future attempts to develop such resistance by design. We want to use two approaches towards understanding and developing durable resistance: the first one is based on the naturally occurring durable resistance gene Lr34 against rust and mildew diseases in wheat. This gene was recently isolated in our group and it encodes a putative ABC type of transporter protein, providing a possible link between non-host and durable resistance. Its function in resistance will be studied by genetic and biochemical approaches in the crop plant wheat, as there is no Lr34-type of resistance characterized in any other plant. However, there is a close Lr34-homolog in rice and its function will be investigated in this diploid system. The second approach will be based on natural diversity found in a specific resistance gene, conferring strong, but not durable resistance. This diversity will be used for a designed improvement of durability by developing new proteins or protein combinations to which the pathogen can not adapt. We will use the 15 naturally occurring alleles of the Pm3 powdery mildew resistance genes to identify the structural basis of specific interactions. Based on this characterization, we will develop intragenic or gene combination pyramiding strategies to obtain more broad-spectrum and more durable resistance.
Summary
Plants and their pathogens are in a constant process of co-evolution. Consequently, many of the known defense genes of plants against fungal pathogens are rapidly loosing effectiveness under agricultural conditions. However, there are examples for durable resistance. It is one of the main research questions in plant biology to determine the genetic basis of such naturally occurring resistance and to understand the underlying biochemical and molecular cause for durability. This durability is characterized by the apparent inability of the pathogen to adapt to the resistance mechanism. The molecular understanding of durable resistance will contribute to future attempts to develop such resistance by design. We want to use two approaches towards understanding and developing durable resistance: the first one is based on the naturally occurring durable resistance gene Lr34 against rust and mildew diseases in wheat. This gene was recently isolated in our group and it encodes a putative ABC type of transporter protein, providing a possible link between non-host and durable resistance. Its function in resistance will be studied by genetic and biochemical approaches in the crop plant wheat, as there is no Lr34-type of resistance characterized in any other plant. However, there is a close Lr34-homolog in rice and its function will be investigated in this diploid system. The second approach will be based on natural diversity found in a specific resistance gene, conferring strong, but not durable resistance. This diversity will be used for a designed improvement of durability by developing new proteins or protein combinations to which the pathogen can not adapt. We will use the 15 naturally occurring alleles of the Pm3 powdery mildew resistance genes to identify the structural basis of specific interactions. Based on this characterization, we will develop intragenic or gene combination pyramiding strategies to obtain more broad-spectrum and more durable resistance.
Max ERC Funding
2 100 000 €
Duration
Start date: 2010-04-01, End date: 2015-03-31
Project acronym ECAP
Project Genetic/epigenetic basis of ethnic differences in cancer predisposition
Researcher (PI) Gian-Paolo Dotto
Host Institution (HI) UNIVERSITE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS4, ERC-2013-ADG
Summary "Integration of large scale genetic and epigenetic analysis needs to be coupled with well defined biological hypotheses that can be experimentally tested. This project is aimed at developing a novel integrated approach to understand genetic and epigenetic predisposition to cancer with skin as model system.
The Caucasian (West European) and Asian (East Asian) populations differ substantially in their predisposition to skin cancer, specifically Squamous Cell Carcinoma (SCC). The underlying mechanisms are poorly understood. As in other organs, skin SCC results from changes in both epithelial and mesenchymal compartments. We will be focusing on two key gene regulatory networks of cells of the two compartments (keratinocytes and dermal fibroblasts), with a key role in skin SCC. The ""keratinocyte network"" has Notch/p53/p63 as key nodes, while the ""dermal fibroblast network"" had Notch and AP1 family members. We will pursue two main goals :
1) We will test the hypothesis that a linkage can be established between specific genetic and epigenetic marks in the Caucasian versus Asian populations and differences in expression and function of ""keratinocyte and/or dermal fibroblast network genes"".
2) We will test the hypothesis that keratinocytes and/or dermal fibroblasts of Caucasian versus Asian individuals differ in their tumor yielding capability, and that these differences in cancer forming capability are due to differences in either ""keratinocyte or dermal fibroblast network genes"".
The applicant is a world leader in epithelial signaling and cancer biology, and is heading interdisciplinary research efforts that bridge the basic and clinical sciences. Together with his bioinformatician and clinician collaborators, he is in an excellent position to attain the high goals of the proposal. The approach has not been attempted before, is only possible within the frame of an advanced ERC grant, and has substantial basic as well as translational/clinical implications."
Summary
"Integration of large scale genetic and epigenetic analysis needs to be coupled with well defined biological hypotheses that can be experimentally tested. This project is aimed at developing a novel integrated approach to understand genetic and epigenetic predisposition to cancer with skin as model system.
The Caucasian (West European) and Asian (East Asian) populations differ substantially in their predisposition to skin cancer, specifically Squamous Cell Carcinoma (SCC). The underlying mechanisms are poorly understood. As in other organs, skin SCC results from changes in both epithelial and mesenchymal compartments. We will be focusing on two key gene regulatory networks of cells of the two compartments (keratinocytes and dermal fibroblasts), with a key role in skin SCC. The ""keratinocyte network"" has Notch/p53/p63 as key nodes, while the ""dermal fibroblast network"" had Notch and AP1 family members. We will pursue two main goals :
1) We will test the hypothesis that a linkage can be established between specific genetic and epigenetic marks in the Caucasian versus Asian populations and differences in expression and function of ""keratinocyte and/or dermal fibroblast network genes"".
2) We will test the hypothesis that keratinocytes and/or dermal fibroblasts of Caucasian versus Asian individuals differ in their tumor yielding capability, and that these differences in cancer forming capability are due to differences in either ""keratinocyte or dermal fibroblast network genes"".
The applicant is a world leader in epithelial signaling and cancer biology, and is heading interdisciplinary research efforts that bridge the basic and clinical sciences. Together with his bioinformatician and clinician collaborators, he is in an excellent position to attain the high goals of the proposal. The approach has not been attempted before, is only possible within the frame of an advanced ERC grant, and has substantial basic as well as translational/clinical implications."
Max ERC Funding
2 495 425 €
Duration
Start date: 2014-02-01, End date: 2020-01-31
Project acronym ECC SCIENG
Project Error-correcting codes and their applications in Science and Engineering
Researcher (PI) Mohammad Amin Shokrollahi
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), PE6, ERC-2008-AdG
Summary Error correcting codes are combinatorial objects which have traditionally been used to enhance the transmission of data on unreliable media. They have experienced a phenomenal growth since their birth some fifty years ago. Today, everyday tasks such as listening to a CD, accessing the hard disk of an electronic device, talking on a wireless phone, or downloading files from the Internet are impossible without the use of error-correcting codes. Though traditional communication still occupies centerstage in the realm of applied coding theory, emerging applications are changing the rules of the game, and calling for a new type of coding theory capable of addressing future needs. These are not limited to physical applications, however. In fact, coding theory is an integral part of solutions offered by researchers outside traditional physical communication to solve fundamental problems of interest, such as the complexity of computation, reliable transfer of bulk data, cryptographic protocols, self correcting software, signal processing, or even computational biology.While research in the past fifty years has put traditional coding theory on firm theoretical grounds, emerging applications are in need of new tools and methods to design, analyze, and implement coding technologies capable of dealing with future needs. This is the main concern of the present proposal. To strike the right balance between length and impact we have identified five areas of research that span the full spectrum of coding theory ranging from fundamental theoretical aspects to practical applications. We set out to develop new theoretical and practical models for the design and analysis of codes, and explore new application areas hitherto untouched. A unique feature of this proposal is our choice of the tools, ranging from classical areas of algebra, combinatorics, and probability theory, to ideas and methods from theoretical computer science.
Summary
Error correcting codes are combinatorial objects which have traditionally been used to enhance the transmission of data on unreliable media. They have experienced a phenomenal growth since their birth some fifty years ago. Today, everyday tasks such as listening to a CD, accessing the hard disk of an electronic device, talking on a wireless phone, or downloading files from the Internet are impossible without the use of error-correcting codes. Though traditional communication still occupies centerstage in the realm of applied coding theory, emerging applications are changing the rules of the game, and calling for a new type of coding theory capable of addressing future needs. These are not limited to physical applications, however. In fact, coding theory is an integral part of solutions offered by researchers outside traditional physical communication to solve fundamental problems of interest, such as the complexity of computation, reliable transfer of bulk data, cryptographic protocols, self correcting software, signal processing, or even computational biology.While research in the past fifty years has put traditional coding theory on firm theoretical grounds, emerging applications are in need of new tools and methods to design, analyze, and implement coding technologies capable of dealing with future needs. This is the main concern of the present proposal. To strike the right balance between length and impact we have identified five areas of research that span the full spectrum of coding theory ranging from fundamental theoretical aspects to practical applications. We set out to develop new theoretical and practical models for the design and analysis of codes, and explore new application areas hitherto untouched. A unique feature of this proposal is our choice of the tools, ranging from classical areas of algebra, combinatorics, and probability theory, to ideas and methods from theoretical computer science.
Max ERC Funding
1 959 998 €
Duration
Start date: 2009-04-01, End date: 2013-03-31
Project acronym ECOEVODRTB
Project Linking within-host and between-host evolution of multidrug-resistant Mycobacterium tuberculosis
Researcher (PI) Sebastien GAGNEUX
Host Institution (HI) SCHWEIZERISCHES TROPEN- UND PUBLIC HEALTH-INSTITUT
Country Switzerland
Call Details Advanced Grant (AdG), LS8, ERC-2019-ADG
Summary Mycobacterium tuberculosis (Mtb), the etiologic agent of tuberculosis (TB), is the main cause of human deaths due to infection in general, and to antimicrobial resistance in particular. Little is known on the within-host evolution of Mtb. Theory predicts that in chronic infections like TB, short-sighted evolution operates in the light of a trade-off between virulence and transmission. This trade-off is particularly relevant in the context of drug resistance, given the fitness costs of resistance. However, the role of short-sighted evolution in TB has never been explored empirically. Theory and model systems further predict that phenotypic drug tolerance facilitates the emergence of drug resistance, but the relevance of phenotypic drug tolerance for drug resistance evolution in the clinic has not been established for TB or any other bacterial disease. To address these and related questions, I propose to build on my recent work on the transmission of drug-resistant Mtb with a new focus on the within-patient evolution of drug-resistant Mtb and its link to between-patient evolution during transmission.
Specifically, I shall:
1) Define the genomic characteristics and evolutionary forces shaping multidrug-resistant Mtb populations in individual patients over time and across different body compartments;
2) Compare the genomic and phenotypic properties of multidrug-resistant Mtb populations in individual patients to those circulating within the corresponding patient population;
3) Determine the effect of suboptimal patient treatment and phenotypic drug tolerance on drug resistance evolution in Mtb inside patients.
By combining population genomics of Mtb sampled sequentially and from surgical specimens with experimental evolution and phenotypic characterization of clinical and experimentally evolved strains, this project will generate new insights relevant to both basic science and global public health.
Summary
Mycobacterium tuberculosis (Mtb), the etiologic agent of tuberculosis (TB), is the main cause of human deaths due to infection in general, and to antimicrobial resistance in particular. Little is known on the within-host evolution of Mtb. Theory predicts that in chronic infections like TB, short-sighted evolution operates in the light of a trade-off between virulence and transmission. This trade-off is particularly relevant in the context of drug resistance, given the fitness costs of resistance. However, the role of short-sighted evolution in TB has never been explored empirically. Theory and model systems further predict that phenotypic drug tolerance facilitates the emergence of drug resistance, but the relevance of phenotypic drug tolerance for drug resistance evolution in the clinic has not been established for TB or any other bacterial disease. To address these and related questions, I propose to build on my recent work on the transmission of drug-resistant Mtb with a new focus on the within-patient evolution of drug-resistant Mtb and its link to between-patient evolution during transmission.
Specifically, I shall:
1) Define the genomic characteristics and evolutionary forces shaping multidrug-resistant Mtb populations in individual patients over time and across different body compartments;
2) Compare the genomic and phenotypic properties of multidrug-resistant Mtb populations in individual patients to those circulating within the corresponding patient population;
3) Determine the effect of suboptimal patient treatment and phenotypic drug tolerance on drug resistance evolution in Mtb inside patients.
By combining population genomics of Mtb sampled sequentially and from surgical specimens with experimental evolution and phenotypic characterization of clinical and experimentally evolved strains, this project will generate new insights relevant to both basic science and global public health.
Max ERC Funding
2 500 000 €
Duration
Start date: 2020-08-01, End date: 2025-07-31
Project acronym EcoImmuneCosts
Project Immunity in Ecology and Evolution: 'Hidden' costs of disease, immune function and their consequences for Darwinian fitness
Researcher (PI) Dennis Lennart HASSELQUIST
Host Institution (HI) LUNDS UNIVERSITET
Country Sweden
Call Details Advanced Grant (AdG), LS8, ERC-2016-ADG
Summary Eco-immunology targets one of the great challenges in biology and medicine - how the immune system has evolved to optimize protection and minimize immunopathology (incl. autoimmune) costs. A primary target of my proposal is to study low-virulent pathogens causing mild infections, which for long have been considered harmless. Recent research suggests that this notion is false and that seemingly harmless pathogens entail delayed (‘hidden’) fitness costs. However, the mechanisms mediating these costs are still unknown. I will experimentally test if accelerated telomere degradation is a causative mechanism through which small immune costs can accumulate and be translated into senescence and reduced Darwinian fitness. Another key target is immune costs, which may be ‘hidden’ because of sexually antagonistic effects, and I will study how this may affect immune gene variation, immune costs and Darwinian fitness. These aspects are central for advancing our understanding of the evolution of disease resistance and immune function, incl. immune over-reactions (autoimmunity).
My project exploits a comprehensive 32-year study of great reed warblers to analyze selection patterns in the wild (Fig. 1a), and uses established captive songbird set-ups to conduct carefully designed experiments. The exceptional quality of the long-term data set, together with cutting-edge techniques to measure and manipulate parasite infection, telomere length, oxidative stress and immune gene diversity, provides exciting opportunities to conduct research that previously was unfeasible, pushing the rapidly growing field of eco-immunology (Fig. 1b) to new frontiers. The work integrates theory and methods of evolutionary ecology, immunology and molecular biology, and has broad significance including for e.g. epidemiology and ageing research. I envision my research to change how we look upon causes, consequences (and precautions) of mild infectious, autoimmune and degenerative diseases.
Summary
Eco-immunology targets one of the great challenges in biology and medicine - how the immune system has evolved to optimize protection and minimize immunopathology (incl. autoimmune) costs. A primary target of my proposal is to study low-virulent pathogens causing mild infections, which for long have been considered harmless. Recent research suggests that this notion is false and that seemingly harmless pathogens entail delayed (‘hidden’) fitness costs. However, the mechanisms mediating these costs are still unknown. I will experimentally test if accelerated telomere degradation is a causative mechanism through which small immune costs can accumulate and be translated into senescence and reduced Darwinian fitness. Another key target is immune costs, which may be ‘hidden’ because of sexually antagonistic effects, and I will study how this may affect immune gene variation, immune costs and Darwinian fitness. These aspects are central for advancing our understanding of the evolution of disease resistance and immune function, incl. immune over-reactions (autoimmunity).
My project exploits a comprehensive 32-year study of great reed warblers to analyze selection patterns in the wild (Fig. 1a), and uses established captive songbird set-ups to conduct carefully designed experiments. The exceptional quality of the long-term data set, together with cutting-edge techniques to measure and manipulate parasite infection, telomere length, oxidative stress and immune gene diversity, provides exciting opportunities to conduct research that previously was unfeasible, pushing the rapidly growing field of eco-immunology (Fig. 1b) to new frontiers. The work integrates theory and methods of evolutionary ecology, immunology and molecular biology, and has broad significance including for e.g. epidemiology and ageing research. I envision my research to change how we look upon causes, consequences (and precautions) of mild infectious, autoimmune and degenerative diseases.
Max ERC Funding
2 500 000 €
Duration
Start date: 2017-08-01, End date: 2023-01-31
Project acronym Ecol of interactions
Project Developing the predictive ecology of plant-animal interactions across space and time
Researcher (PI) Catherine GRAHAM
Host Institution (HI) EIDGENOSSICHEN FORSCHUNGSANSTALT FUR WALD SCHNEE UND LANDSCHAFT
Country Switzerland
Call Details Advanced Grant (AdG), LS8, ERC-2017-ADG
Summary In the face of the alarming pace of recent environmental change we lack the tools to accurately predict how biodiversity and ecosystem services will respond. One key gap in knowledge that limits our predictive ability is uncertainty concerning how the biotic interactions will change. Developing a predictive science of species interactions requires integrating evolutionary, biogeographic and ecological mechanisms acting at different spatial and temporal scales. We will use a hierarchical cross-scale approach, combining phylogeography, network ecology, statistical modelling and experiments, to disentangle the mechanisms governing species richness and mutualistic interactions in tropical hummingbirds and their food plants. Hummingbirds and their food plants are an excellent model system because they are highly diverse, highly specialized, and logistically feasible to study. Our objectives are to (1) evaluate the influence of factors, such as trait-matching, environmental conditions and relatedness, on network structure; (2) quantify how and why interaction beta-diversity (i.e., reflecting the change in both species composition, and in interacting partners) changes across elevation gradients in each of three biogeographic regions with distinct evolutionary histories (mountain regions in Costa Rica, Ecuador, Brazil); (3) evaluate the importance of multiple factors, such as trait-matching, environmental conditions, relatedness and abundance, on species interactions and network structure; and (4) develop a predictive model of species interactions and evaluate its performance using cross-validation and experimentation. Together, these tasks will provide new insight into one of the central enigmas in ecology, namely, why species diversity and its interaction architecture change across space and time. We will also be able predict how species interactions will change from present to the future, which is essential for the conservation of biodiversity and ecosystem services.
Summary
In the face of the alarming pace of recent environmental change we lack the tools to accurately predict how biodiversity and ecosystem services will respond. One key gap in knowledge that limits our predictive ability is uncertainty concerning how the biotic interactions will change. Developing a predictive science of species interactions requires integrating evolutionary, biogeographic and ecological mechanisms acting at different spatial and temporal scales. We will use a hierarchical cross-scale approach, combining phylogeography, network ecology, statistical modelling and experiments, to disentangle the mechanisms governing species richness and mutualistic interactions in tropical hummingbirds and their food plants. Hummingbirds and their food plants are an excellent model system because they are highly diverse, highly specialized, and logistically feasible to study. Our objectives are to (1) evaluate the influence of factors, such as trait-matching, environmental conditions and relatedness, on network structure; (2) quantify how and why interaction beta-diversity (i.e., reflecting the change in both species composition, and in interacting partners) changes across elevation gradients in each of three biogeographic regions with distinct evolutionary histories (mountain regions in Costa Rica, Ecuador, Brazil); (3) evaluate the importance of multiple factors, such as trait-matching, environmental conditions, relatedness and abundance, on species interactions and network structure; and (4) develop a predictive model of species interactions and evaluate its performance using cross-validation and experimentation. Together, these tasks will provide new insight into one of the central enigmas in ecology, namely, why species diversity and its interaction architecture change across space and time. We will also be able predict how species interactions will change from present to the future, which is essential for the conservation of biodiversity and ecosystem services.
Max ERC Funding
2 499 930 €
Duration
Start date: 2018-10-01, End date: 2023-09-30
Project acronym ElectroGene
Project Electrogenetics – Shaping Electrogenetic Interfaces for Closed-Loop Voltage-Controlled Gene Expression
Researcher (PI) Martin Fussenegger
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Country Switzerland
Call Details Advanced Grant (AdG), LS9, ERC-2017-ADG
Summary Man and man-made electronic systems share the same ecosystem, and yet work radically differently. Human metabolism uses ion gradients across insulated membranes to simultaneously process slow analog chemical reactions and communicate information in multicellular systems via soluble/volatile molecular signals. By contrast, electronic systems use multicore central processing units to control the flow of electrons through insulated metal wires with gigahertz frequency and communicate information across networks via wired/wireless connections. With the advent of the internet of things, networks of interconnected electronic devices will reach the processing complexity of living systems, yet they remain largely incompatible with biological systems. Wearable electronics can profile physical parameters such as steps and heartbeat, and Google’s proposal to develop glucose-monitoring contact lenses has triggered a wave of interest in harnessing the full potential of bioelectronics for medical applications. Yet this vision remains limited to diagnostics. Capitalizing on our mind-controlled and smartphone-adjustable optogenetic drug-dosing devices, ElectroGene will establish the foundations of electrogenetics, the science of creating electro-genetic interfaces that enable direct two-way communication between electronic devices and living cells. ElectroGene consists of three pillars, (i) voltage-triggered gene expression, (ii) genetically programmed electronics and (iii) wireless-powered implants providing closed-loop bioelectronic control, which allow real-time monitoring of metabolic conditions (diagnosis), enable remote-controlled production and dosing of protein therapeutics by implanted designer cells (treatment), and manage closed-loop control between cells and electronics, thus linking diagnosis and therapy to block disease onset (prevention). ElectroGene design principles and devices will be validated in proof-of-concept preclinical studies for the treatment of diabetes.
Summary
Man and man-made electronic systems share the same ecosystem, and yet work radically differently. Human metabolism uses ion gradients across insulated membranes to simultaneously process slow analog chemical reactions and communicate information in multicellular systems via soluble/volatile molecular signals. By contrast, electronic systems use multicore central processing units to control the flow of electrons through insulated metal wires with gigahertz frequency and communicate information across networks via wired/wireless connections. With the advent of the internet of things, networks of interconnected electronic devices will reach the processing complexity of living systems, yet they remain largely incompatible with biological systems. Wearable electronics can profile physical parameters such as steps and heartbeat, and Google’s proposal to develop glucose-monitoring contact lenses has triggered a wave of interest in harnessing the full potential of bioelectronics for medical applications. Yet this vision remains limited to diagnostics. Capitalizing on our mind-controlled and smartphone-adjustable optogenetic drug-dosing devices, ElectroGene will establish the foundations of electrogenetics, the science of creating electro-genetic interfaces that enable direct two-way communication between electronic devices and living cells. ElectroGene consists of three pillars, (i) voltage-triggered gene expression, (ii) genetically programmed electronics and (iii) wireless-powered implants providing closed-loop bioelectronic control, which allow real-time monitoring of metabolic conditions (diagnosis), enable remote-controlled production and dosing of protein therapeutics by implanted designer cells (treatment), and manage closed-loop control between cells and electronics, thus linking diagnosis and therapy to block disease onset (prevention). ElectroGene design principles and devices will be validated in proof-of-concept preclinical studies for the treatment of diabetes.
Max ERC Funding
2 500 000 €
Duration
Start date: 2018-11-01, End date: 2023-10-31
Project acronym ELEGANSFUSION
Project Mechanisms of cell fusion in eukaryotes
Researcher (PI) Benjamin Podbilewicz
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Country Israel
Call Details Advanced Grant (AdG), LS3, ERC-2010-AdG_20100317
Summary Membrane fusion is a universal process essential inside cells (endoplasmic) and between cells in fertilization and organ formation (exoplasmic). With the exception of SNARE-mediated endoplasmic fusion the proteins that mediate cellular fusion (fusogens) are unknown. Despite many years of research, little is known about the mechanism of cell-cell fusion. Our studies of developmental cell fusion in the nematode C. elegans have led to the discovery of the first family of eukaryotic fusogens (FF). These fusogens, EFF-1 and AFF-1, are type I membrane glycoproteins that are essential for cell fusion and can fuse cells when ectopically expressed on the membranes of C. elegans and heterologous cells.
Our main goals are:
(1) To determine the physicochemical mechanism of cell membrane fusion mediated by FF proteins.
(2) To find the missing fusogens that act in cell fusion events across all kingdoms of life.
We hypothesize that FF proteins fuse membranes by a mechanism analogous to viral or endoplasmic fusogens and that unidentified fusogens fuse cells following the same principles as FF proteins.
Our specific aims are:
AIM 1 Determine the mechanism of FF-mediated cell fusion: A paradigm for cell membrane fusion
AIM 2 Find the sperm-egg fusion proteins (fusogens) in C. elegans
AIM 3 Identify the myoblast fusogens in mammals
AIM 4 Test fusogens using functional cell fusion assays in heterologous systems
Identifying critical domains required for FF fusion, intermediates in membrane remodeling, and atomic structures of FF proteins will advance the fundamental understanding of the mechanisms of eukaryotic cell fusion. We propose to find the Holy Grail of fertilization and mammalian myoblast fusion. We estimate that this project, if successful, will bring a breakthrough to the sperm-egg and muscle fusion fields with potential applications in basic and applied biomedical sciences.
Summary
Membrane fusion is a universal process essential inside cells (endoplasmic) and between cells in fertilization and organ formation (exoplasmic). With the exception of SNARE-mediated endoplasmic fusion the proteins that mediate cellular fusion (fusogens) are unknown. Despite many years of research, little is known about the mechanism of cell-cell fusion. Our studies of developmental cell fusion in the nematode C. elegans have led to the discovery of the first family of eukaryotic fusogens (FF). These fusogens, EFF-1 and AFF-1, are type I membrane glycoproteins that are essential for cell fusion and can fuse cells when ectopically expressed on the membranes of C. elegans and heterologous cells.
Our main goals are:
(1) To determine the physicochemical mechanism of cell membrane fusion mediated by FF proteins.
(2) To find the missing fusogens that act in cell fusion events across all kingdoms of life.
We hypothesize that FF proteins fuse membranes by a mechanism analogous to viral or endoplasmic fusogens and that unidentified fusogens fuse cells following the same principles as FF proteins.
Our specific aims are:
AIM 1 Determine the mechanism of FF-mediated cell fusion: A paradigm for cell membrane fusion
AIM 2 Find the sperm-egg fusion proteins (fusogens) in C. elegans
AIM 3 Identify the myoblast fusogens in mammals
AIM 4 Test fusogens using functional cell fusion assays in heterologous systems
Identifying critical domains required for FF fusion, intermediates in membrane remodeling, and atomic structures of FF proteins will advance the fundamental understanding of the mechanisms of eukaryotic cell fusion. We propose to find the Holy Grail of fertilization and mammalian myoblast fusion. We estimate that this project, if successful, will bring a breakthrough to the sperm-egg and muscle fusion fields with potential applications in basic and applied biomedical sciences.
Max ERC Funding
2 380 000 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym Epiherigans
Project Writing, reading and managing stress with H3K9me
Researcher (PI) Susan GASSER
Host Institution (HI) UNIVERSITE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS2, ERC-2016-ADG
Summary Epigenetic inheritance is the transmission of information, generally in the form of DNA methylation or post-translational modifications on histones that regulate the availability of underlying genetic information for transcription. RNA itself feeds back to contribute to histone modification. Sequence accessibility is both a matter of folding the chromatin fibre to alter access to recognition motifs, and the local concentration of factors needed for efficient transcriptional initiation, elongation, termination or mRNA stability. In heterochromatin we find a subset of regulatory factors in carefully balanced concentrations that are maintained in part by the segregation of active and inactive domains. Histone H3 K9 methylation is key to this compartmentation.
C. elegans provides an ideal system in which to study chromatin-based gene repression. We have demonstrated that histone H3 K9 methylation is the essential signal for the sequestration of heterochromatin at the nuclear envelope in C. elegans. The recognition of H3K9me1/2/3 by an inner nuclear envelope-bound chromodomain protein, CEC-4, actively sequesters heterochromatin in embryos, and contributes redundantly in adult tissues.
Epiherigans has the ambitious goal to determine definitively what targets H3K9 methylation, and identify its physiological roles. We will examine how this mark contributes to the epigenetic recognition of repeat vs non-repeat sequence, and mediates a stress-induced response to oxidative damage. We will examine the link between these and the spatial clustering of heterochromatic domains. Epiherigans will develop an integrated approach to identify in vivo the factors that distinguish repeats from non-repeats, self from non-self within genomes and will examine how H3K9me contributes to a persistent ROS or DNA damage stress response. It represents a crucial step towards understanding of how our genomes use heterochromatin to modulate, stabilize and transmit chromatin organization.
Summary
Epigenetic inheritance is the transmission of information, generally in the form of DNA methylation or post-translational modifications on histones that regulate the availability of underlying genetic information for transcription. RNA itself feeds back to contribute to histone modification. Sequence accessibility is both a matter of folding the chromatin fibre to alter access to recognition motifs, and the local concentration of factors needed for efficient transcriptional initiation, elongation, termination or mRNA stability. In heterochromatin we find a subset of regulatory factors in carefully balanced concentrations that are maintained in part by the segregation of active and inactive domains. Histone H3 K9 methylation is key to this compartmentation.
C. elegans provides an ideal system in which to study chromatin-based gene repression. We have demonstrated that histone H3 K9 methylation is the essential signal for the sequestration of heterochromatin at the nuclear envelope in C. elegans. The recognition of H3K9me1/2/3 by an inner nuclear envelope-bound chromodomain protein, CEC-4, actively sequesters heterochromatin in embryos, and contributes redundantly in adult tissues.
Epiherigans has the ambitious goal to determine definitively what targets H3K9 methylation, and identify its physiological roles. We will examine how this mark contributes to the epigenetic recognition of repeat vs non-repeat sequence, and mediates a stress-induced response to oxidative damage. We will examine the link between these and the spatial clustering of heterochromatic domains. Epiherigans will develop an integrated approach to identify in vivo the factors that distinguish repeats from non-repeats, self from non-self within genomes and will examine how H3K9me contributes to a persistent ROS or DNA damage stress response. It represents a crucial step towards understanding of how our genomes use heterochromatin to modulate, stabilize and transmit chromatin organization.
Max ERC Funding
2 500 000 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym EUKARYOTIC RIBOSOME
Project Structural studies of the eukaryotic ribosome by X-ray crystallography
Researcher (PI) Nenad Ban
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Country Switzerland
Call Details Advanced Grant (AdG), LS1, ERC-2009-AdG
Summary The ribosome is a large cellular organelle that plays a central role in the process of protein synthesis in all organisms. Currently, structural information at atomic resolution exists only for bacterial ribosomes and some of their functional complexes. Eukaryotic ribosomes are larger and significantly more complex than their bacterial counterparts. They consist of two unequal subunits with a combined molecular weight of approximately 4 million Daltons and contain 70-80 different protein molecules and four different RNAs. Currently the only structural information on eukaryotic ribosomes is available from cryo electron microscopic reconstructions in the nanometer resolution range, which is insufficient to derive information about the function of the eukaryotic ribosome at the atomic level. The aim of this proposal is to use X-ray crystallography to obtain structural and functional information on the eukaryotic ribosome and its functional complexes at high resolution. The key targets of the structural work will be: i) the structure of the small ribosomal subunit, ii) the structure of the large ribosomal subunit, and iii) structures of complexes involved in the initiation of protein synthesis. Besides the obvious fundamental importance of this research for understanding protein synthesis in eukaryotes the proposed studies will also be the prerequisite for understanding the structural basis of the regulation of protein synthesis in normal cells and how it is perturbed in various diseases. Finally, comparing the structures of bacterial and eukaryotic ribosomes is important for understanding the specificity of various clinically used antibiotics for the bacterial ribosome.
Summary
The ribosome is a large cellular organelle that plays a central role in the process of protein synthesis in all organisms. Currently, structural information at atomic resolution exists only for bacterial ribosomes and some of their functional complexes. Eukaryotic ribosomes are larger and significantly more complex than their bacterial counterparts. They consist of two unequal subunits with a combined molecular weight of approximately 4 million Daltons and contain 70-80 different protein molecules and four different RNAs. Currently the only structural information on eukaryotic ribosomes is available from cryo electron microscopic reconstructions in the nanometer resolution range, which is insufficient to derive information about the function of the eukaryotic ribosome at the atomic level. The aim of this proposal is to use X-ray crystallography to obtain structural and functional information on the eukaryotic ribosome and its functional complexes at high resolution. The key targets of the structural work will be: i) the structure of the small ribosomal subunit, ii) the structure of the large ribosomal subunit, and iii) structures of complexes involved in the initiation of protein synthesis. Besides the obvious fundamental importance of this research for understanding protein synthesis in eukaryotes the proposed studies will also be the prerequisite for understanding the structural basis of the regulation of protein synthesis in normal cells and how it is perturbed in various diseases. Finally, comparing the structures of bacterial and eukaryotic ribosomes is important for understanding the specificity of various clinically used antibiotics for the bacterial ribosome.
Max ERC Funding
2 446 725 €
Duration
Start date: 2010-07-01, End date: 2015-06-30
Project acronym EVOMORPHYS
Project Identifying how Evolution exploits physical properties of tissues to generate the complexity and diversity of Life
Researcher (PI) Michel Charles MILINKOVITCH
Host Institution (HI) UNIVERSITE DE GENEVE
Country Switzerland
Call Details Advanced Grant (AdG), LS8, ERC-2018-ADG
Summary My project focuses on answering one fundamental question: what are the drivers of Life’s morphological complexity and diversity? I claim that this question can only be addressed by a Newtonian-Darwinian synthesis that considers how Evolution exploits the physical properties of living matter. I will investigate how the evolutionary process explores the phase space of possible interactions between physical (mechanics, reaction-diffusion) and biological (cell signalling, proliferation, migration) processes and generates configurations that compute functional phenotypes. In particular, I will combine experiments in biology and physics, as well as mathematical models and Artificial-Life (ALife) numerical simulations. The latter will be based on physics’ first principles, symmetry-breaking processes and a genetic algorithm. First, I will investigate how geometry affects signalling by (i) imaging the embryonic development of colour patterns and skin geometries of multiple squamate species with various scale-to-colour pattern correspondences, (ii) generating CRISPR/Cas9 scaleless mutants in two lizard species to study the effect of skin 3D geometry on colour patterning, and (iii) performing ALife experiments to explore how the evolutionary process can modify signalling events and exploit geometry to generate new patterns. Second, I will analyse how growth can affect geometry by (i) performing in-silico experiments where coupling between growth and morphogenesis is systematically explored and (ii) evaluating how much the in-silico model captures morphologies generated with physics laboratory experiments using 3D layered polymeric gels. Third, I will build a Newtonian-Darwinian framework by coupling geometry, signalling, growth and mechanics in extensive open-ended ALife experiments. The results of the EVOMORPHYS project will constitute a novel framework for understanding how Evolution exploits physics to generate the morphological diversity and complexity of Life forms.
Summary
My project focuses on answering one fundamental question: what are the drivers of Life’s morphological complexity and diversity? I claim that this question can only be addressed by a Newtonian-Darwinian synthesis that considers how Evolution exploits the physical properties of living matter. I will investigate how the evolutionary process explores the phase space of possible interactions between physical (mechanics, reaction-diffusion) and biological (cell signalling, proliferation, migration) processes and generates configurations that compute functional phenotypes. In particular, I will combine experiments in biology and physics, as well as mathematical models and Artificial-Life (ALife) numerical simulations. The latter will be based on physics’ first principles, symmetry-breaking processes and a genetic algorithm. First, I will investigate how geometry affects signalling by (i) imaging the embryonic development of colour patterns and skin geometries of multiple squamate species with various scale-to-colour pattern correspondences, (ii) generating CRISPR/Cas9 scaleless mutants in two lizard species to study the effect of skin 3D geometry on colour patterning, and (iii) performing ALife experiments to explore how the evolutionary process can modify signalling events and exploit geometry to generate new patterns. Second, I will analyse how growth can affect geometry by (i) performing in-silico experiments where coupling between growth and morphogenesis is systematically explored and (ii) evaluating how much the in-silico model captures morphologies generated with physics laboratory experiments using 3D layered polymeric gels. Third, I will build a Newtonian-Darwinian framework by coupling geometry, signalling, growth and mechanics in extensive open-ended ALife experiments. The results of the EVOMORPHYS project will constitute a novel framework for understanding how Evolution exploits physics to generate the morphological diversity and complexity of Life forms.
Max ERC Funding
2 499 070 €
Duration
Start date: 2019-08-01, End date: 2024-07-31
Project acronym F-Addict
Project Convergence of positive and negative reinforcement in fentanyl addiction
Researcher (PI) Christian LueSCHER
Host Institution (HI) UNIVERSITE DE GENEVE
Country Switzerland
Call Details Advanced Grant (AdG), LS5, ERC-2019-ADG
Summary F-Addict strives to unravel the neural circuits driving compulsion in fentanyl addiction. We ask the question how fentanyl causes fast transition from medical or recreational controlled drug use to compulsive consumption. About a third of opioid users eventually loose control, which increases the risk of death by overdose; a number that is even higher for fentanyl and definitely exceeds the transition observed with psychostimulants. The neural correlate of this difference remains elusive. We posit that repetitive withdrawal leads to strong negative reinforcement, which in conjunction with inherent positive reinforcement favors the transition to compulsion. F-Addict will uncover the synaptic processes and neuronal population activity leading to addiction in a mouse model of oral fentanyl self-administration. Much preliminary data implicate activity in the mesolimbic dopamine system and upstream subcortical regions (paraventricular thalamus/habenula/basolateral amygdala) in positive and negative reinforcement, respectively. In addition, top down control, in particular by the orbitofrontal cortex may drive compulsive drug use. The proposed project will harness advanced circuit investigations for an innovative, original perspective: how does positive and negative reinforcement in fentanyl addiction contrast with current circuit models of addiction that are based on psychostimulants? In a translational spirit, F-Addict will also examine the effects of oral substitution with methadone and buprenorphine, recognized therapies for opioid addiction. Much preliminary data provides proof of feasibility and principle. We are confident that our approach at the frontiers of modern neurosciences carries the potential for groundbreaking results to answer a timely question. Unraveling the neural basis of fentanyl addiction will enhance the molecular understanding of circuit modulation to shape future therapies facing the still growing opioid epidemic.
Summary
F-Addict strives to unravel the neural circuits driving compulsion in fentanyl addiction. We ask the question how fentanyl causes fast transition from medical or recreational controlled drug use to compulsive consumption. About a third of opioid users eventually loose control, which increases the risk of death by overdose; a number that is even higher for fentanyl and definitely exceeds the transition observed with psychostimulants. The neural correlate of this difference remains elusive. We posit that repetitive withdrawal leads to strong negative reinforcement, which in conjunction with inherent positive reinforcement favors the transition to compulsion. F-Addict will uncover the synaptic processes and neuronal population activity leading to addiction in a mouse model of oral fentanyl self-administration. Much preliminary data implicate activity in the mesolimbic dopamine system and upstream subcortical regions (paraventricular thalamus/habenula/basolateral amygdala) in positive and negative reinforcement, respectively. In addition, top down control, in particular by the orbitofrontal cortex may drive compulsive drug use. The proposed project will harness advanced circuit investigations for an innovative, original perspective: how does positive and negative reinforcement in fentanyl addiction contrast with current circuit models of addiction that are based on psychostimulants? In a translational spirit, F-Addict will also examine the effects of oral substitution with methadone and buprenorphine, recognized therapies for opioid addiction. Much preliminary data provides proof of feasibility and principle. We are confident that our approach at the frontiers of modern neurosciences carries the potential for groundbreaking results to answer a timely question. Unraveling the neural basis of fentanyl addiction will enhance the molecular understanding of circuit modulation to shape future therapies facing the still growing opioid epidemic.
Max ERC Funding
2 499 748 €
Duration
Start date: 2020-06-01, End date: 2025-05-31
Project acronym FICModFun
Project FIC-Mediated Post-Translational Modifications at the
Pathogen-Host Interface: Elucidating Structure, Function and Role in Infection
Researcher (PI) Christoph Georg Fritz Dehio
Host Institution (HI) UNIVERSITAT BASEL
Country Switzerland
Call Details Advanced Grant (AdG), LS6, ERC-2013-ADG
Summary The ubiquitous FIC domain catalyzes post-translational modifications (PTMs) of target proteins; i.e.
adenylylation (=AMPylation) and, more rarely, uridylylation and phosphocholination. Fic proteins are
thought to play critical roles in intrinsic signaling processes of prokaryotes and eukaryotes; however, a
subset encoded by bacterial pathogens is translocated via dedicated secretion systems into the cytoplasm of
mammalian host cells. Some of these host-targeted Fic proteins modify small GTPases leading to collapse of
the actin cytoskeleton and other drastic cellular changes. Recently, we described a large set of functionally
diverse homologues in pathogens of the genus Bartonella that are required for their “stealth attack” strategy
and persistent course of infection [1, 2]. Our preliminary functional analysis of some of these host-targeted
Fic proteins of Bartonella demonstrated adenylylation activity towards novel host targets (e.g. tubulin and
vimentin). Moreover, in addition to the canonical adenylylation activity they may also display a competing
kinase activity resulting from altered ATP binding to the FIC active site. Finally, we described a conserved
mechanism of FIC active site auto- inhibition that is relieved by a single amino acid exchange [1], thus
facilitating functional analysis of any Fic protein of interest. Despite this recent progress only a few Fic
proteins have been functionally characterized to date; our understanding of the functional plasticity of the
FIC domain in mediating diverse target PTMs and their specific roles in infection thus remains limited.
In this project, we aim to study the vast repertoire of host-targeted Fic proteins of Bartonella to: 1)
identify novel target proteins and types of PTMs; 2) study their physiological consequences and molecular
mechanisms of action; and 3) analyze structure-function relationships critical for FIC-mediated PTMs and infer from these data determinants of target specificity, type of PTM and mode of regulation. At the forefront of infection biology research, this project is ground-breaking as (i) we will identify a
plethora of novel host target PTMs that are critical for a “stealth attack” infection strategy and thus will open
new avenues for investigating fundamental mechanisms of persistent infection; and (ii), we will unveil the
molecular basis of the remarkable functional versatility of the structurally conserved FIC domain.
Summary
The ubiquitous FIC domain catalyzes post-translational modifications (PTMs) of target proteins; i.e.
adenylylation (=AMPylation) and, more rarely, uridylylation and phosphocholination. Fic proteins are
thought to play critical roles in intrinsic signaling processes of prokaryotes and eukaryotes; however, a
subset encoded by bacterial pathogens is translocated via dedicated secretion systems into the cytoplasm of
mammalian host cells. Some of these host-targeted Fic proteins modify small GTPases leading to collapse of
the actin cytoskeleton and other drastic cellular changes. Recently, we described a large set of functionally
diverse homologues in pathogens of the genus Bartonella that are required for their “stealth attack” strategy
and persistent course of infection [1, 2]. Our preliminary functional analysis of some of these host-targeted
Fic proteins of Bartonella demonstrated adenylylation activity towards novel host targets (e.g. tubulin and
vimentin). Moreover, in addition to the canonical adenylylation activity they may also display a competing
kinase activity resulting from altered ATP binding to the FIC active site. Finally, we described a conserved
mechanism of FIC active site auto- inhibition that is relieved by a single amino acid exchange [1], thus
facilitating functional analysis of any Fic protein of interest. Despite this recent progress only a few Fic
proteins have been functionally characterized to date; our understanding of the functional plasticity of the
FIC domain in mediating diverse target PTMs and their specific roles in infection thus remains limited.
In this project, we aim to study the vast repertoire of host-targeted Fic proteins of Bartonella to: 1)
identify novel target proteins and types of PTMs; 2) study their physiological consequences and molecular
mechanisms of action; and 3) analyze structure-function relationships critical for FIC-mediated PTMs and infer from these data determinants of target specificity, type of PTM and mode of regulation. At the forefront of infection biology research, this project is ground-breaking as (i) we will identify a
plethora of novel host target PTMs that are critical for a “stealth attack” infection strategy and thus will open
new avenues for investigating fundamental mechanisms of persistent infection; and (ii), we will unveil the
molecular basis of the remarkable functional versatility of the structurally conserved FIC domain.
Max ERC Funding
1 699 858 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym FilAtmo
Project Laser Filamentation for Probing and Controlling Atmospheric Processes
Researcher (PI) Jean-Pierre, Louis Wolf
Host Institution (HI) UNIVERSITE DE GENEVE
Country Switzerland
Call Details Advanced Grant (AdG), PE2, ERC-2011-ADG_20110209
Summary The prevention of damaging weather phenomena like floods, hail and lightning strikes has been a dream for centuries. We propose a highly innovative approach relying on laser filaments for both triggering and guiding lightning and produce water condensation in the atmosphere. Filaments are self-sustained light strings of typ. 100 um diameter and hundreds of meters length in air, bear very high intensities and are electrically conductive through molecular ionization.
The filamentation process in air was considered until recently as resulting from the dynamic balance between the optical Kerr effect and defocusing by the self-generated plasma. Our unexpected discovery, last year, that filaments are governed by negative higher-order Kerr effect (HOKE), opened both basic physical questions about the stabilization mechanism and new opportunities to optimize the envisioned applications to lightning triggering and cloud condensation.
We propose first to study in the laboratory the physical origin of the alternated signs of HOKE in gases, which are suspected to stem from populated bound states. Coherently controlling these bound states in rare gases and air will allow us to tailor the HOKE inversion, and consequently to control the filament process itself. Optimal pulse shapes will then be sought by adaptive (closed loop) techniques to maximize the plasma density and lifetime in filaments for lightning control applications. Similar coherent control approaches will be performed for optimizing the complex photochemistry that leads to water vapor condensation in the atmosphere.
We will then apply the optimal pulse shapes to real scale field experiments. To this end we intend to use the mobile TW laser from the Teramobile consortium, which we are part of, in order to perform two extensive campaigns for real-scale lightning control (in Lugano) and haze/cloud generation (in Geneva). These experiments will constitute the first coherent manipulation of atmospheric process.
Summary
The prevention of damaging weather phenomena like floods, hail and lightning strikes has been a dream for centuries. We propose a highly innovative approach relying on laser filaments for both triggering and guiding lightning and produce water condensation in the atmosphere. Filaments are self-sustained light strings of typ. 100 um diameter and hundreds of meters length in air, bear very high intensities and are electrically conductive through molecular ionization.
The filamentation process in air was considered until recently as resulting from the dynamic balance between the optical Kerr effect and defocusing by the self-generated plasma. Our unexpected discovery, last year, that filaments are governed by negative higher-order Kerr effect (HOKE), opened both basic physical questions about the stabilization mechanism and new opportunities to optimize the envisioned applications to lightning triggering and cloud condensation.
We propose first to study in the laboratory the physical origin of the alternated signs of HOKE in gases, which are suspected to stem from populated bound states. Coherently controlling these bound states in rare gases and air will allow us to tailor the HOKE inversion, and consequently to control the filament process itself. Optimal pulse shapes will then be sought by adaptive (closed loop) techniques to maximize the plasma density and lifetime in filaments for lightning control applications. Similar coherent control approaches will be performed for optimizing the complex photochemistry that leads to water vapor condensation in the atmosphere.
We will then apply the optimal pulse shapes to real scale field experiments. To this end we intend to use the mobile TW laser from the Teramobile consortium, which we are part of, in order to perform two extensive campaigns for real-scale lightning control (in Lugano) and haze/cloud generation (in Geneva). These experiments will constitute the first coherent manipulation of atmospheric process.
Max ERC Funding
2 403 425 €
Duration
Start date: 2012-07-01, End date: 2017-06-30
Project acronym FLAY
Project Flavor Anomalies and the origin of the Yukawa couplings
Researcher (PI) Gino ISIDORI
Host Institution (HI) UNIVERSITAT ZURICH
Country Switzerland
Call Details Advanced Grant (AdG), PE2, ERC-2018-ADG
Summary Recent experimental results in flavor physics exhibit deviations from the Standard Model predictions that are growing with time, both as far as statistical significance and as far as internal consistency. Understanding the origin of this phenomenon, the so-called “flavor anomalies”, is of paramount importance for a deeper understanding of fundamental interactions. As recently shown by the PI and collaborators, this phenomenon is likely to be intimately related to the long-standing “flavor problem”, or the origin of the hierarchical pattern of quark and lepton mass matrices observed in Nature. The goal of this project is to shed light on both these issues, providing a solution to old and recent puzzles in flavor physics. We propose to address these questions via an original bottom-up approach, based on Effective Field Theory methods and simplified models, combined with new top-down ideas about the ultraviolet completion of the Standard Model. On the phenomenological side, the proposed bottom-up approach will allow us to exploit with the highest accuracy all the available and expected experimental data. It will allow us to take into account both low- and high-energy observables, as well as both quark and lepton sectors. These results will constitute the basis for the theoretical investigation of a new class of Standard Model extensions not considered so far. The latter are based on new ideas, such as flavor non-universal gauge interactions, that imply a change of paradigm in theoretical high-energy physics: the origin of the flavor hierarchies plays a central role in revealing the ultraviolet completion of the Standard Model. Combining a bottom-up approach to flavor-physics data with top-down ideas on the origin of the flavor hierarchies, this project has the potential to lead to a major advancement in fundamental physics.
Summary
Recent experimental results in flavor physics exhibit deviations from the Standard Model predictions that are growing with time, both as far as statistical significance and as far as internal consistency. Understanding the origin of this phenomenon, the so-called “flavor anomalies”, is of paramount importance for a deeper understanding of fundamental interactions. As recently shown by the PI and collaborators, this phenomenon is likely to be intimately related to the long-standing “flavor problem”, or the origin of the hierarchical pattern of quark and lepton mass matrices observed in Nature. The goal of this project is to shed light on both these issues, providing a solution to old and recent puzzles in flavor physics. We propose to address these questions via an original bottom-up approach, based on Effective Field Theory methods and simplified models, combined with new top-down ideas about the ultraviolet completion of the Standard Model. On the phenomenological side, the proposed bottom-up approach will allow us to exploit with the highest accuracy all the available and expected experimental data. It will allow us to take into account both low- and high-energy observables, as well as both quark and lepton sectors. These results will constitute the basis for the theoretical investigation of a new class of Standard Model extensions not considered so far. The latter are based on new ideas, such as flavor non-universal gauge interactions, that imply a change of paradigm in theoretical high-energy physics: the origin of the flavor hierarchies plays a central role in revealing the ultraviolet completion of the Standard Model. Combining a bottom-up approach to flavor-physics data with top-down ideas on the origin of the flavor hierarchies, this project has the potential to lead to a major advancement in fundamental physics.
Max ERC Funding
2 318 750 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym FRONTIERS OF RNAI-II
Project High resolution and chemical genetic approaches to RNA silencing mechanisms
Researcher (PI) Olivier Robert Georges Voinnet
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Country Switzerland
Call Details Advanced Grant (AdG), LS2, ERC-2012-ADG_20120314
Summary In eukaryotes, silencing small (s)RNAs, including micro (mi)RNAs and small interfering (si)RNAs, regulate many aspects of biology, including cell differentiation, development, hormonal responses, or defense. In particular, many plant and metazoan miRNAs play crucial roles in embryonic/post-embryonic development; the precise timing and localization of their expression is thus crucial to their action. Hence, specific miRNA repertoires underlie specific cell identities, and deviations from such repertoires often have deleterious consequences such as cancer. Many miRNAs also help organisms to adapt to stress, thus, given their importance in virtually all aspects of biology, understanding how, when and where miRNAs exert their actions is of paramount importance. To date, however, the few approaches to miRNA-mediated silencing in whole organisms have not taken into account the exquisite definition, in space and time, of their biogenesis and action, leading to an inaccurate view of the biology of these molecules at the systems level.
Using the root system of the model plant Arabidopsis thaliana, we propose to explore, at single-cell and subcellular resolution levels, the biology of the main miRNA effector protein, ARGONAUTE 1 (AGO1) in intact tissues. Using a combination of state-of the-art technologies for single-cell forward genetics, protein purification and RNA/polysome profiling, we will establish a functional spatiotemporal map of the root AGO1-sRNAome and identify cell-specific modifiers of sRNA biogenesis and action. As a complement to the above approaches, chemical genetics will isolate small molecules allowing direct and specific manipulation of AGO1-dependent sRNA pathways in planta. RNA silencing modifier compounds will also accelerate forward and reverse approaches of RNA silencing in plants with sensitized genetic backgrounds, and uncover novel connections between miRNA/siRNA and physiological or metabolic pathways.
Summary
In eukaryotes, silencing small (s)RNAs, including micro (mi)RNAs and small interfering (si)RNAs, regulate many aspects of biology, including cell differentiation, development, hormonal responses, or defense. In particular, many plant and metazoan miRNAs play crucial roles in embryonic/post-embryonic development; the precise timing and localization of their expression is thus crucial to their action. Hence, specific miRNA repertoires underlie specific cell identities, and deviations from such repertoires often have deleterious consequences such as cancer. Many miRNAs also help organisms to adapt to stress, thus, given their importance in virtually all aspects of biology, understanding how, when and where miRNAs exert their actions is of paramount importance. To date, however, the few approaches to miRNA-mediated silencing in whole organisms have not taken into account the exquisite definition, in space and time, of their biogenesis and action, leading to an inaccurate view of the biology of these molecules at the systems level.
Using the root system of the model plant Arabidopsis thaliana, we propose to explore, at single-cell and subcellular resolution levels, the biology of the main miRNA effector protein, ARGONAUTE 1 (AGO1) in intact tissues. Using a combination of state-of the-art technologies for single-cell forward genetics, protein purification and RNA/polysome profiling, we will establish a functional spatiotemporal map of the root AGO1-sRNAome and identify cell-specific modifiers of sRNA biogenesis and action. As a complement to the above approaches, chemical genetics will isolate small molecules allowing direct and specific manipulation of AGO1-dependent sRNA pathways in planta. RNA silencing modifier compounds will also accelerate forward and reverse approaches of RNA silencing in plants with sensitized genetic backgrounds, and uncover novel connections between miRNA/siRNA and physiological or metabolic pathways.
Max ERC Funding
2 251 600 €
Duration
Start date: 2013-07-01, End date: 2018-06-30
Project acronym FUN-SP
Project A functional framework for sparse, non-gaussian signal processing and bioimaging
Researcher (PI) Michael Unser
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), PE7, ERC-2010-AdG_20100224
Summary "In recent years, the research focus in signal processing has shifted away from the classical linear paradigm, which is intimately linked with the theory of stationary Gaussian processes. Instead of considering Fourier transforms and performing quadratic optimization, researchers are presently favoring wavelet-like representations and have adopted ”sparsity” as design paradigm.
Our ambition is to develop a unifying operator-based framework for signal processing that would provide the ``sparse"" counterpart of the classical theory, which is currently missing. To that end, we shall specify and investigate sparse stochastic processes that are continuously-defined and ruled by differential equations, and construct corresponding wavelet-like sparsifying transforms. Our hope is to be able to rigorously connect non-quadratic regularization and sparsity-constrained optimization to well-defined continuous-domain statistical models. We also want to develop a novel Lie-group formalism for the design of steerable, signal-adapted wavelet transforms with improved invariance and sparsifying properties, both in 2-D and 3-D.
We shall use these tools to define new reversible image representations in terms of singular points (contours and keypoints) and to develop novel algorithms for 3-D biomedical image analysis. In close collaboration with imaging scientists, we shall apply our framework to the development of new 3-D reconstruction algorithms for emerging bioimaging modalities such as fluorescence deconvolution microscopy, digital holography microscopy, X-ray phase-contrast microscopy, and advanced MRI."
Summary
"In recent years, the research focus in signal processing has shifted away from the classical linear paradigm, which is intimately linked with the theory of stationary Gaussian processes. Instead of considering Fourier transforms and performing quadratic optimization, researchers are presently favoring wavelet-like representations and have adopted ”sparsity” as design paradigm.
Our ambition is to develop a unifying operator-based framework for signal processing that would provide the ``sparse"" counterpart of the classical theory, which is currently missing. To that end, we shall specify and investigate sparse stochastic processes that are continuously-defined and ruled by differential equations, and construct corresponding wavelet-like sparsifying transforms. Our hope is to be able to rigorously connect non-quadratic regularization and sparsity-constrained optimization to well-defined continuous-domain statistical models. We also want to develop a novel Lie-group formalism for the design of steerable, signal-adapted wavelet transforms with improved invariance and sparsifying properties, both in 2-D and 3-D.
We shall use these tools to define new reversible image representations in terms of singular points (contours and keypoints) and to develop novel algorithms for 3-D biomedical image analysis. In close collaboration with imaging scientists, we shall apply our framework to the development of new 3-D reconstruction algorithms for emerging bioimaging modalities such as fluorescence deconvolution microscopy, digital holography microscopy, X-ray phase-contrast microscopy, and advanced MRI."
Max ERC Funding
2 106 994 €
Duration
Start date: 2011-04-01, End date: 2016-03-31
Project acronym GANGS
Project Gangs, Gangsters, and Ganglands: Towards a Global Comparative Ethnography
Researcher (PI) Dennis RODGERS
Host Institution (HI) FONDATION POUR L INSTITUT DE HAUTES ETUDES INTERNATIONALES ET DU DEVELOPPEMENT
Country Switzerland
Call Details Advanced Grant (AdG), SH3, ERC-2017-ADG
Summary Gangs occupy a key position in the global imaginary of violence, widely perceived and represented as primary sources of brutality and insecurity. This can be related to the fact that they are one of a small number of truly global phenomena, found in almost every society across both time and space. At the same time, however, as almost 100 years of gang research have highlighted, the phenomenon can vary significantly in form, dynamics, and consequences. While there have been many insightful studies of gangs, the overwhelming majority have focused on a single group or location, and we still lack a proper sense of what kinds of gang dynamics might be general, and which ones are specific to particular times and places. The GANGS project will develop a systematic comparative investigation of global gang dynamics, to better understand why they emerge, how they evolve over time, whether they are associated with particular urban configurations, how and why individuals join gangs, and what impact this has on their potential futures. It will draw on original ethnographic research carried out in multiple locations, adopting an explicitly tripartite focus on “Gangs”, “Gangsters”, and “Ganglands” in order to better explore the interplay between group, individual, and contextual factors. The first will consider the organisational dynamics of gangs, the second will focus on individual gang members and their trajectories before, during, and after their involvement in a gang, while the third will reflect on the contexts within which gangs emerge and evolve. Research will combine innovative collaborative ethnography in Nicaragua, South Africa, and France, a ground-breaking comparison of 35 individual gang member life histories from across Africa, Asia, Europe, North and South America, and unique joint ethnographic investigations into the political economy of three gang-affected cities in Nicaragua, France, and South Africa.
Summary
Gangs occupy a key position in the global imaginary of violence, widely perceived and represented as primary sources of brutality and insecurity. This can be related to the fact that they are one of a small number of truly global phenomena, found in almost every society across both time and space. At the same time, however, as almost 100 years of gang research have highlighted, the phenomenon can vary significantly in form, dynamics, and consequences. While there have been many insightful studies of gangs, the overwhelming majority have focused on a single group or location, and we still lack a proper sense of what kinds of gang dynamics might be general, and which ones are specific to particular times and places. The GANGS project will develop a systematic comparative investigation of global gang dynamics, to better understand why they emerge, how they evolve over time, whether they are associated with particular urban configurations, how and why individuals join gangs, and what impact this has on their potential futures. It will draw on original ethnographic research carried out in multiple locations, adopting an explicitly tripartite focus on “Gangs”, “Gangsters”, and “Ganglands” in order to better explore the interplay between group, individual, and contextual factors. The first will consider the organisational dynamics of gangs, the second will focus on individual gang members and their trajectories before, during, and after their involvement in a gang, while the third will reflect on the contexts within which gangs emerge and evolve. Research will combine innovative collaborative ethnography in Nicaragua, South Africa, and France, a ground-breaking comparison of 35 individual gang member life histories from across Africa, Asia, Europe, North and South America, and unique joint ethnographic investigations into the political economy of three gang-affected cities in Nicaragua, France, and South Africa.
Max ERC Funding
2 498 079 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym GAPWAVE ICS
Project Waveguide-type semiconductor integrated circuits (ICs) in gaps between conducting surfaces with texture – architecture, electromagnetic modeling and micromachining
Researcher (PI) Per-Simon Kildal
Host Institution (HI) CHALMERS TEKNISKA HOEGSKOLA AB
Country Sweden
Call Details Advanced Grant (AdG), PE7, ERC-2012-ADG_20120216
Summary In order to explore and exploit the frequency range from 30 GHz up to THz, new types of transmission lines and semiconductor architectures are needed. Conventional microwave technologies that are commonly used below 30 GHz become either too lossy or are too expensive to manufacture, and technologies used in the optical regime are not usable either. The intermediate frequency band is therefore often referred to as the THz gap, indicating the lack of commercialize-able technologies there.
Professor Kildal has invented a fundamentally new regime of transmission line, referred to as gap waveguides. The basis is newly discovered local waves appearing in the gap between two conducting surfaces, controlled by a texture in one or both of the surfaces. The gap waveguide has been verified below 20 GHz, but it will be more advantageous in the THz gap. The texture will for THz applications be of submillimeter or micrometer scale, realizable by micromachining or etching. Also, there is no need for a dielectric substrate, and there is no need for conductive contact between the two surfaces. Therefore, such gap waveguides and circuits for the THz gap can be manufactured with low cost.
The vision is that the topology of this new regime of gap waveguides will facilitate integration of semiconductor devices, and may lay the foundation for new architectures of transistors and other integrated circuits, being located inside the gap encapsulated by the conductive surfaces themselves. In order to reach this vision new and efficient numerical electromagnetic methods and modeling tools need to be developed, taking advantage of the particular gap waveguide geometry, and being able to connect to or replace the charge transport models for the transistors in the doped semiconductors themselves.
The gap waveguide technology can get a tremendous impact on exploring higher frequencies in radio astronomy, communications, and imaging for medical as well as security applications.
Summary
In order to explore and exploit the frequency range from 30 GHz up to THz, new types of transmission lines and semiconductor architectures are needed. Conventional microwave technologies that are commonly used below 30 GHz become either too lossy or are too expensive to manufacture, and technologies used in the optical regime are not usable either. The intermediate frequency band is therefore often referred to as the THz gap, indicating the lack of commercialize-able technologies there.
Professor Kildal has invented a fundamentally new regime of transmission line, referred to as gap waveguides. The basis is newly discovered local waves appearing in the gap between two conducting surfaces, controlled by a texture in one or both of the surfaces. The gap waveguide has been verified below 20 GHz, but it will be more advantageous in the THz gap. The texture will for THz applications be of submillimeter or micrometer scale, realizable by micromachining or etching. Also, there is no need for a dielectric substrate, and there is no need for conductive contact between the two surfaces. Therefore, such gap waveguides and circuits for the THz gap can be manufactured with low cost.
The vision is that the topology of this new regime of gap waveguides will facilitate integration of semiconductor devices, and may lay the foundation for new architectures of transistors and other integrated circuits, being located inside the gap encapsulated by the conductive surfaces themselves. In order to reach this vision new and efficient numerical electromagnetic methods and modeling tools need to be developed, taking advantage of the particular gap waveguide geometry, and being able to connect to or replace the charge transport models for the transistors in the doped semiconductors themselves.
The gap waveguide technology can get a tremendous impact on exploring higher frequencies in radio astronomy, communications, and imaging for medical as well as security applications.
Max ERC Funding
1 659 302 €
Duration
Start date: 2013-05-01, End date: 2017-04-30
Project acronym GENCON
Project The evolutionary implications of genetic conflict
Researcher (PI) Goeran Arnqvist
Host Institution (HI) UPPSALA UNIVERSITET
Country Sweden
Call Details Advanced Grant (AdG), LS8, ERC-2011-ADG_20110310
Summary The study of genetic conflict is developing at an almost explosive rate. The recognition that genes or alleles residing in individuals of the two sexes may have conflicting interests is transforming evolutionary biology and, likewise, conflict between genes showing different modes of transmission may fundamentally affect adaptive evolution. The research proposed here will push the frontiers of genetic conflict research and establish new domains. It is aimed at exploring the novel possibility that conflict between mitochondrial and nuclear genes have far-reaching implications for adaptive evolution and at advancing our understanding of the biological consequences of sexual conflict. The project consists of several interrelated parts and will employ insects as model systems. First, I will assess to what extent genetic variation in fitness is sexually antagonistic and what life history traits contribute to sexually antagonistic variation. Second, I will elucidate the genomics of metabolic rate and measure selection on metabolic phenotypes. Third, I will test whether sexually antagonistic epistatic interactions between mitochondrial and nuclear genes generate conflict over metabolic rate. Fourth, I will test the hypothesis that sexual conflict contribute to the evolution of primary and secondary sexual traits. Fifth, I will shed light on the complicated evolutionary interplay between sexual conflict and mating system evolution. I will employ an innovative research strategy, ‘experimental genomics’, in which genomic data is used to guide experimental evolutionary work with distinct genotypes. The research outlined here will collectively provide an unprecedented wealth of information into the role of genetic conflict in several horizons of adaptive evolution, ranging from DNA sequence evolution over life history evolution to speciation, and will set the standard for a new generation of insightful studies aimed at bridging the gap between phenotypic selection and genomics.
Summary
The study of genetic conflict is developing at an almost explosive rate. The recognition that genes or alleles residing in individuals of the two sexes may have conflicting interests is transforming evolutionary biology and, likewise, conflict between genes showing different modes of transmission may fundamentally affect adaptive evolution. The research proposed here will push the frontiers of genetic conflict research and establish new domains. It is aimed at exploring the novel possibility that conflict between mitochondrial and nuclear genes have far-reaching implications for adaptive evolution and at advancing our understanding of the biological consequences of sexual conflict. The project consists of several interrelated parts and will employ insects as model systems. First, I will assess to what extent genetic variation in fitness is sexually antagonistic and what life history traits contribute to sexually antagonistic variation. Second, I will elucidate the genomics of metabolic rate and measure selection on metabolic phenotypes. Third, I will test whether sexually antagonistic epistatic interactions between mitochondrial and nuclear genes generate conflict over metabolic rate. Fourth, I will test the hypothesis that sexual conflict contribute to the evolution of primary and secondary sexual traits. Fifth, I will shed light on the complicated evolutionary interplay between sexual conflict and mating system evolution. I will employ an innovative research strategy, ‘experimental genomics’, in which genomic data is used to guide experimental evolutionary work with distinct genotypes. The research outlined here will collectively provide an unprecedented wealth of information into the role of genetic conflict in several horizons of adaptive evolution, ranging from DNA sequence evolution over life history evolution to speciation, and will set the standard for a new generation of insightful studies aimed at bridging the gap between phenotypic selection and genomics.
Max ERC Funding
2 497 442 €
Duration
Start date: 2012-05-01, End date: 2017-04-30
Project acronym GENE TARGET T2D
Project General and targeted approaches to unravel the molecular causes of type 2 diabetes
Researcher (PI) Leif Christer Groop
Host Institution (HI) MAX IV Laboratory, Lund University
Country Sweden
Call Details Advanced Grant (AdG), LS4, ERC-2010-AdG_20100317
Summary Type 2 diabetes (T2D) affects worldwide at present about 250 million patients and an estimated 380 million in 2025. This epidemic has been ascribed to a collision between genes and an affluent society. Genetics of T2D has during recent years identified > 30 variants increasing susceptibility to T2D. Yet, these variants explain only 15% of the heritability of T2D. One reason could be that whole genome association studies can only detect common variants whereas identification of rare variants with stronger effects would require sequencing. A large part of this application is devoted to sequencing of affected family members from unique large pedigrees traced back to common ancestors around 1600. The advantage of using families is that identified variants can be tested for segregation with the trait. Genetic variants can influence expression of a gene in an allele specific manner. This will be explored by combining exome sequencing with sequencing of RNA from human islets.
Impaired effects of the incretin hormones GLP-1 and GIP on the pancreatic islets represent central defects in T2D. Variants in the TCF7L2 and GIPR genes contribute to these defects. I will here explore the molecular mechanisms by which TCF7L2, the strongest T2D gene, causes T2D. GIP has unprecedented effects not only on islet function but also on body composition, blood flow and vascular complications in T2D. This application explores these effects and will test whether manipulation of GIP can mimic the normalization of glucose tolerance seen after gastric bypass surgery.
Taken together, these general and targeted approaches are expected not only to provide new insights into the causes of T2D but also contribute with vital information for development of new treatments for T2D.
Summary
Type 2 diabetes (T2D) affects worldwide at present about 250 million patients and an estimated 380 million in 2025. This epidemic has been ascribed to a collision between genes and an affluent society. Genetics of T2D has during recent years identified > 30 variants increasing susceptibility to T2D. Yet, these variants explain only 15% of the heritability of T2D. One reason could be that whole genome association studies can only detect common variants whereas identification of rare variants with stronger effects would require sequencing. A large part of this application is devoted to sequencing of affected family members from unique large pedigrees traced back to common ancestors around 1600. The advantage of using families is that identified variants can be tested for segregation with the trait. Genetic variants can influence expression of a gene in an allele specific manner. This will be explored by combining exome sequencing with sequencing of RNA from human islets.
Impaired effects of the incretin hormones GLP-1 and GIP on the pancreatic islets represent central defects in T2D. Variants in the TCF7L2 and GIPR genes contribute to these defects. I will here explore the molecular mechanisms by which TCF7L2, the strongest T2D gene, causes T2D. GIP has unprecedented effects not only on islet function but also on body composition, blood flow and vascular complications in T2D. This application explores these effects and will test whether manipulation of GIP can mimic the normalization of glucose tolerance seen after gastric bypass surgery.
Taken together, these general and targeted approaches are expected not only to provide new insights into the causes of T2D but also contribute with vital information for development of new treatments for T2D.
Max ERC Funding
2 499 480 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym GENEWELL
Project Genetics and epigenetics of animal welfare
Researcher (PI) Per Ole Stokmann Jensen
Host Institution (HI) LINKOPINGS UNIVERSITET
Country Sweden
Call Details Advanced Grant (AdG), LS9, ERC-2012-ADG_20120314
Summary Animal welfare is a topic of highest societal and scientific priority. Here, I propose to use genomic and epigenetic tools to provide a new perspective on the biology of animal welfare. This will reveal mechanisms involved in modulating stress responses. Groundbreaking aspects include new insights into how environmental conditions shape the orchestration of the genome by means of epigenetic mechanisms, and how this in turn modulates coping patterns of animals. The flexible epigenome comprises the interface between the environment and the genome. It is involved in both short- and long-term, including transgenerational, adaptations of animals. Hence, populations may adapt to environmental conditions over generations, using epigenetic mechanisms. The project will primarily be based on chickens, but will also be extended to a novel species, the dog. We will generate congenic chicken strains, where interesting alleles and epialleles will be fixed against a common background of either RJF or domestic genotypes. In these, we will apply a broad phenotyping strategy, to characterize the effects on different welfare relevant behaviors. Furthermore, we will characterize how environmental stress affects the epigenome of birds, and tissue samples from more than 500 birds from an intercross between RJF and White Leghorn layers will be used to perform an extensive meth-QTL-analysis. This will reveal environmental and genetic mechanisms affecting gene-specific methylation. The dog is another highly interesting species in the context of behavior genetics, because of its high inter-breed variation in behavior, and its compact and sequenced genome. We will set up a large-scale F2-intercross experiment and phenotype about 400 dogs in standardized behavioral tests. All individuals will be genotyped on about 1000 genetic markers, and this will be used for performing an extensive QTL-analysis in order to find new loci and alleles associated with personalities and coping patterns.
Summary
Animal welfare is a topic of highest societal and scientific priority. Here, I propose to use genomic and epigenetic tools to provide a new perspective on the biology of animal welfare. This will reveal mechanisms involved in modulating stress responses. Groundbreaking aspects include new insights into how environmental conditions shape the orchestration of the genome by means of epigenetic mechanisms, and how this in turn modulates coping patterns of animals. The flexible epigenome comprises the interface between the environment and the genome. It is involved in both short- and long-term, including transgenerational, adaptations of animals. Hence, populations may adapt to environmental conditions over generations, using epigenetic mechanisms. The project will primarily be based on chickens, but will also be extended to a novel species, the dog. We will generate congenic chicken strains, where interesting alleles and epialleles will be fixed against a common background of either RJF or domestic genotypes. In these, we will apply a broad phenotyping strategy, to characterize the effects on different welfare relevant behaviors. Furthermore, we will characterize how environmental stress affects the epigenome of birds, and tissue samples from more than 500 birds from an intercross between RJF and White Leghorn layers will be used to perform an extensive meth-QTL-analysis. This will reveal environmental and genetic mechanisms affecting gene-specific methylation. The dog is another highly interesting species in the context of behavior genetics, because of its high inter-breed variation in behavior, and its compact and sequenced genome. We will set up a large-scale F2-intercross experiment and phenotype about 400 dogs in standardized behavioral tests. All individuals will be genotyped on about 1000 genetic markers, and this will be used for performing an extensive QTL-analysis in order to find new loci and alleles associated with personalities and coping patterns.
Max ERC Funding
2 499 828 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym GlobalBioIm
Project Global integrative framework for Computational Bio-Imaging
Researcher (PI) Michael Unser
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), PE7, ERC-2015-AdG
Summary A powerful strategy for increasing the quality and resolution of medical and biological images is to acquire larger quantities of data (Fourier samples for MRI, projections for X-ray imaging) and to jointly reconstruct the complete signal by correctly reallocating the measurements in 3D space/time and integrating all the information available. The underlying image sequence is reconstructed globally as the result of a very large-scale optimization that exploits the redundancy of the signal (spatio-temporal correlation, sparsity) to improve the solution. Due to recent advances in the field, we are arguing that such a “bigger data” integration is now within reach and that our team is ideally qualified to lead the way. A successful outcome will profoundly impact the design of future bioimaging systems.
We are proposing a unifying framework for the development of such next-generation reconstruction algorithms with a clear separation between the physical (forward model) and signal-related (regularization, incorporation of prior constraints) aspects of the problem. The pillars of our formulation are: an operator algebra with a corresponding set of fast linear solvers; an advanced statistical framework for the principled derivation of reconstruction methods; and learning schemes for parameter optimization and self-tuning. These core technologies will be incorporated into a modular software library featuring the key components for the implementation and testing of iterative reconstruction algorithms. We shall apply our framework to improve upon the state of the art in the following modalities: 1) phase-contrast X-ray tomography in full 3D; 2) structured illumination microscopy; 3) single-particle analysis in cryo-electron tomography; 4) a novel multipose fluorescence microscopy; 5) real-time MRI, and 6) a new multimodal digital microscope. In all instances, we shall work in close collaboration with the imaging scientists who are in charge of the instrumentation.
Summary
A powerful strategy for increasing the quality and resolution of medical and biological images is to acquire larger quantities of data (Fourier samples for MRI, projections for X-ray imaging) and to jointly reconstruct the complete signal by correctly reallocating the measurements in 3D space/time and integrating all the information available. The underlying image sequence is reconstructed globally as the result of a very large-scale optimization that exploits the redundancy of the signal (spatio-temporal correlation, sparsity) to improve the solution. Due to recent advances in the field, we are arguing that such a “bigger data” integration is now within reach and that our team is ideally qualified to lead the way. A successful outcome will profoundly impact the design of future bioimaging systems.
We are proposing a unifying framework for the development of such next-generation reconstruction algorithms with a clear separation between the physical (forward model) and signal-related (regularization, incorporation of prior constraints) aspects of the problem. The pillars of our formulation are: an operator algebra with a corresponding set of fast linear solvers; an advanced statistical framework for the principled derivation of reconstruction methods; and learning schemes for parameter optimization and self-tuning. These core technologies will be incorporated into a modular software library featuring the key components for the implementation and testing of iterative reconstruction algorithms. We shall apply our framework to improve upon the state of the art in the following modalities: 1) phase-contrast X-ray tomography in full 3D; 2) structured illumination microscopy; 3) single-particle analysis in cryo-electron tomography; 4) a novel multipose fluorescence microscopy; 5) real-time MRI, and 6) a new multimodal digital microscope. In all instances, we shall work in close collaboration with the imaging scientists who are in charge of the instrumentation.
Max ERC Funding
2 499 515 €
Duration
Start date: 2016-10-01, End date: 2022-03-31
Project acronym GROWTHCONTROL
Project Dissecting the transcriptional mechanisms controlling growth during normal development and cancer
Researcher (PI) Anssi Jussi Nikolai Taipale
Host Institution (HI) KAROLINSKA INSTITUTET
Country Sweden
Call Details Advanced Grant (AdG), LS2, ERC-2008-AdG
Summary The main scientific questions addressed in this proposal relate to the understanding of molecular mechanisms of growth control and cancer through the combined use of high-throughput technologies and computational biology. We aim to create a systems-level understanding of the cell cycle, and its regulation by physiological growth factors and oncogenes through the use high-throughput biology to identify all or the majority of genes that are essential for cell cycle progression, and by combining this dataset with computationally predicted and experimentally validated target genes of growth factors and oncogenic pathways. In my opinion, such systems biology approach is critical for understanding of growth control, as organ-specific growth control has proven particularly refractory to genetic dissection. Much of what we know about physiological mechanisms controlling cellular growth in mammals has been revealed by human cancer genetics. These studies have revealed that a large number of genes can contribute to aberrant cell growth; there are more than 300 genes that have been linked to cancer, and mutations found in cancer are often cell type specific ( oncogene preference , i.e. PTCH mutations in medulloblastoma, APC in colon cancer, TMPRSS2-ERG in prostate cancer), suggesting that different pathways in different cell lineages are coupled to the cell cycle machinery. We have preliminary evidence that hedgehog (Hh) and Wnt signals are directly coupled to expression of N-myc and c-Myc genes, but only in tissues and cell-types that display a proliferative response to these factors. Both classical molecular and developmental biology as well as high throughput and systems biological methods will be used for dissection of the molecular mechanism of this selectivity. If successful, these experiments would establish a principle explaining why particular mutations are extremely common in some tumor types but not found at all in others.
Summary
The main scientific questions addressed in this proposal relate to the understanding of molecular mechanisms of growth control and cancer through the combined use of high-throughput technologies and computational biology. We aim to create a systems-level understanding of the cell cycle, and its regulation by physiological growth factors and oncogenes through the use high-throughput biology to identify all or the majority of genes that are essential for cell cycle progression, and by combining this dataset with computationally predicted and experimentally validated target genes of growth factors and oncogenic pathways. In my opinion, such systems biology approach is critical for understanding of growth control, as organ-specific growth control has proven particularly refractory to genetic dissection. Much of what we know about physiological mechanisms controlling cellular growth in mammals has been revealed by human cancer genetics. These studies have revealed that a large number of genes can contribute to aberrant cell growth; there are more than 300 genes that have been linked to cancer, and mutations found in cancer are often cell type specific ( oncogene preference , i.e. PTCH mutations in medulloblastoma, APC in colon cancer, TMPRSS2-ERG in prostate cancer), suggesting that different pathways in different cell lineages are coupled to the cell cycle machinery. We have preliminary evidence that hedgehog (Hh) and Wnt signals are directly coupled to expression of N-myc and c-Myc genes, but only in tissues and cell-types that display a proliferative response to these factors. Both classical molecular and developmental biology as well as high throughput and systems biological methods will be used for dissection of the molecular mechanism of this selectivity. If successful, these experiments would establish a principle explaining why particular mutations are extremely common in some tumor types but not found at all in others.
Max ERC Funding
2 200 000 €
Duration
Start date: 2009-03-01, End date: 2014-02-28
Project acronym GUTDROSO
Project Gut immunity and homeostasis in Drosophila
Researcher (PI) Bruno Lemaitre
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS6, ERC-2008-AdG
Summary The gut is the major interface between microbes and their animal hosts and constitutes the main entry route for pathogens. As a consequence gut cells must be armed with efficient immune defenses to combat invasion and colonisation by pathogens. However, the gut also harbors a flora of commensal bacteria, with potentially beneficial effects for the host, which must be tolerated without a chronic, and harmful, immune response. In recent years Drosophila has emerged as a powerful model to dissect host-pathogen interactions, leading to the paradigm of antimicrobial peptide regulation by the Toll and Imd signaling pathways. The strength of this model derives from the availability of powerful and cost effective genetic and genomic tools as well as the high degree of similarities to vertebrate innate immunity. However, in spite of growing interest in gut mucosal immunity generally, very little is known about the immune response of the Drosophila gut. Using powerful new tools and those developed in the study of the systemic response, we propose to raise our understanding of Drosophila gut immunity to the same level as that of systemic immunity within the next five years. This project will involve integrated approaches to dissect not only the gut immune response but also gut homeostasis in the presence of commensal microbiota, as well as strategies used by entomopathogens to circumvent these defenses. We believe that the fundamental knowledge generated on Drosophila gut immunity will serve as a paradigm of epithelial immune reactivity and have a wider impact on our comprehension of animal defense mechanisms.
Summary
The gut is the major interface between microbes and their animal hosts and constitutes the main entry route for pathogens. As a consequence gut cells must be armed with efficient immune defenses to combat invasion and colonisation by pathogens. However, the gut also harbors a flora of commensal bacteria, with potentially beneficial effects for the host, which must be tolerated without a chronic, and harmful, immune response. In recent years Drosophila has emerged as a powerful model to dissect host-pathogen interactions, leading to the paradigm of antimicrobial peptide regulation by the Toll and Imd signaling pathways. The strength of this model derives from the availability of powerful and cost effective genetic and genomic tools as well as the high degree of similarities to vertebrate innate immunity. However, in spite of growing interest in gut mucosal immunity generally, very little is known about the immune response of the Drosophila gut. Using powerful new tools and those developed in the study of the systemic response, we propose to raise our understanding of Drosophila gut immunity to the same level as that of systemic immunity within the next five years. This project will involve integrated approaches to dissect not only the gut immune response but also gut homeostasis in the presence of commensal microbiota, as well as strategies used by entomopathogens to circumvent these defenses. We believe that the fundamental knowledge generated on Drosophila gut immunity will serve as a paradigm of epithelial immune reactivity and have a wider impact on our comprehension of animal defense mechanisms.
Max ERC Funding
1 485 627 €
Duration
Start date: 2009-04-01, End date: 2014-03-31
Project acronym Hallmarks-to-Therapy
Project Intersecting hallmarks of cancer: mechanisms of and interplay between invasion and angiogenesis, guiding new strategies for cancer therapy
Researcher (PI) Douglas Hanahan
Host Institution (HI) ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS4, ERC-2012-ADG_20120314
Summary Acquisition of the hallmark capability for invasion and in turn metastasis is for most human cancers the defining event in progression to life threatening disease. Its determinants are remarkably complex. Genetically engineered mice can model human cancers, with tumors arising in specific organs, reflecting onco-genomic and histopathological features of particular tumor types. This project will use four mouse models to characterize newly implicated determinants of invasive tumor growth. We have observed that genetic polymorphisms can govern predisposition to invasive cancer. Additionally, therapeutic targeting of another hallmark capability – tumor angiogenesis – has revealed adaptive resistance, whereby late-stage tumors, faced with the inability to grow en masse supported by angiogenesis, switch instead to grow diffusively, by invading adjacent tissue; this phenomenon may underlay the limited benefit seen with anti-angiogenic therapies in the clinic. There are three interconnected goals:
(1) Polymorphic regulation of tumor invasion. We will investigate the mechanisms and functional importance of candidate genes resident within a genetic modifier locus on mouse Chr 17 that can alternatively suppress or facilitate invasive tumor growth dependent on constitutional genetic background.
(2) Adaptive induction of invasion. We will elucidate the determinants of the invasive growth capability that is induced in response to potent inhibition of angiogenesis.
(3) Testing mechanism-based therapeutic co-targeting of the capabilities for invasion and angiogenesis.
We will use functional genetic, genomic profiling, and pharmacological approaches to assess these two new modes of regulating invasive growth, and then apply the knowledge in preclinical trials aiming to lay the groundwork for future clinical trials in which these intersecting hallmark capabilities are coordinately disrupted, with promise for more enduring therapeutic responses and benefit to cancer patients.
Summary
Acquisition of the hallmark capability for invasion and in turn metastasis is for most human cancers the defining event in progression to life threatening disease. Its determinants are remarkably complex. Genetically engineered mice can model human cancers, with tumors arising in specific organs, reflecting onco-genomic and histopathological features of particular tumor types. This project will use four mouse models to characterize newly implicated determinants of invasive tumor growth. We have observed that genetic polymorphisms can govern predisposition to invasive cancer. Additionally, therapeutic targeting of another hallmark capability – tumor angiogenesis – has revealed adaptive resistance, whereby late-stage tumors, faced with the inability to grow en masse supported by angiogenesis, switch instead to grow diffusively, by invading adjacent tissue; this phenomenon may underlay the limited benefit seen with anti-angiogenic therapies in the clinic. There are three interconnected goals:
(1) Polymorphic regulation of tumor invasion. We will investigate the mechanisms and functional importance of candidate genes resident within a genetic modifier locus on mouse Chr 17 that can alternatively suppress or facilitate invasive tumor growth dependent on constitutional genetic background.
(2) Adaptive induction of invasion. We will elucidate the determinants of the invasive growth capability that is induced in response to potent inhibition of angiogenesis.
(3) Testing mechanism-based therapeutic co-targeting of the capabilities for invasion and angiogenesis.
We will use functional genetic, genomic profiling, and pharmacological approaches to assess these two new modes of regulating invasive growth, and then apply the knowledge in preclinical trials aiming to lay the groundwork for future clinical trials in which these intersecting hallmark capabilities are coordinately disrupted, with promise for more enduring therapeutic responses and benefit to cancer patients.
Max ERC Funding
2 500 000 €
Duration
Start date: 2013-09-01, End date: 2018-08-31
Project acronym HHMM-Neonates
Project DEVELOPMENT OF HEALTHY HOST-MICROBIAL MUTUALISM IN EARLY LIFE
Researcher (PI) Andrew J. Macpherson
Host Institution (HI) UNIVERSITAET BERN
Country Switzerland
Call Details Advanced Grant (AdG), LS6, ERC-2016-ADG
Summary Background
Humans and other animals harbour enormous microbial consortia, especially in the lower intestine. My group has now shown that effects of the microbiota on host are far earlier and more pervasive than previously appreciated, starting even before birth from exposure to defined maternal microbial metabolites.
Concept
There is a critical window for development of immunity and metabolism in early life. This shapes infectious resistance, lymphocyte repertoire development and the likelihood of later autoimmune or inflammatory disease. We will determine the molecular mechanisms of how the maternal microbiota prepares the newborn for the critical fetal/suckling/early-independent-nutrition transitions. The core hypothesis is that generally pervasive effects of maternal microbial influences, so-far investigated only for innate immunity and metabolism of germ-free offspring, can be defined in terms of a clear portfolio of maternal microbial molecular signatures and epigenetic marks as the newborn develops with its own microbiota.
Approach
Interdependence of microbial ⇄ host interactions during gestation and lactation will be dissected using reversible colonisation systems under axenic and precisely controlled gnotobiotic conditions. The flow and identity of maternal microbial metabolites driving development and shaping incoming colonisation shall be determined from high-resolution metabolomics and host strain combinations that reveal in vivo signalling and epigenetic marks.
Significance
The project will reveal mechanisms of the earliest phases of mammalian adaptation to a microbiota, the epigenetic effects of maternal microbial metabolites and the resulting potential protection from metabolic disease or immunopathology. Conversely, there are profound effects of early life adaptation on the dynamics of microbial colonisation and the potential blooms and extinctions for the incoming microbiota: the project will define the different mechanisms involved.
Summary
Background
Humans and other animals harbour enormous microbial consortia, especially in the lower intestine. My group has now shown that effects of the microbiota on host are far earlier and more pervasive than previously appreciated, starting even before birth from exposure to defined maternal microbial metabolites.
Concept
There is a critical window for development of immunity and metabolism in early life. This shapes infectious resistance, lymphocyte repertoire development and the likelihood of later autoimmune or inflammatory disease. We will determine the molecular mechanisms of how the maternal microbiota prepares the newborn for the critical fetal/suckling/early-independent-nutrition transitions. The core hypothesis is that generally pervasive effects of maternal microbial influences, so-far investigated only for innate immunity and metabolism of germ-free offspring, can be defined in terms of a clear portfolio of maternal microbial molecular signatures and epigenetic marks as the newborn develops with its own microbiota.
Approach
Interdependence of microbial ⇄ host interactions during gestation and lactation will be dissected using reversible colonisation systems under axenic and precisely controlled gnotobiotic conditions. The flow and identity of maternal microbial metabolites driving development and shaping incoming colonisation shall be determined from high-resolution metabolomics and host strain combinations that reveal in vivo signalling and epigenetic marks.
Significance
The project will reveal mechanisms of the earliest phases of mammalian adaptation to a microbiota, the epigenetic effects of maternal microbial metabolites and the resulting potential protection from metabolic disease or immunopathology. Conversely, there are profound effects of early life adaptation on the dynamics of microbial colonisation and the potential blooms and extinctions for the incoming microbiota: the project will define the different mechanisms involved.
Max ERC Funding
2 500 000 €
Duration
Start date: 2017-09-01, End date: 2023-08-31
Project acronym HLA-DR15 in MS
Project Functional Role of the HLA-DR15 Haplotype in Multiple Sclerosis
Researcher (PI) Roland Michael Gunnar Martin
Host Institution (HI) University of Zurich
Country Switzerland
Call Details Advanced Grant (AdG), LS6, ERC-2013-ADG
Summary Multiple sclerosis (MS) is a prototypic CD4+ T cell-mediated autoimmune disease that damages the central nervous system. MS affects young adults and women twice as often as men. Neurological deficits cause substantial disability at an early age with high socioeconomic impact.
Both a complex genetic trait and environmental factors are involved in MS etiology. Similar to other autoimmune diseases it has been known for almost 40 years that certain HLA-class II genes, in MS the two DR15 alleles DRB1*15:01 and DRB5*01:01, confer by far most of the genetic risk. Despite this clear role remarkably little is known about the functional contribution of these genes to MS pathogenesis, and this holds also true for all other T cell-mediated autoimmune diseases. It is assumed that the DR15 alleles present peptides from organ-specific self-proteins to T cells and select an autoreactive CD4+ T cell repertoire that can be activated by certain environmental triggers. Interestingly, the effects of the three known environmental risk factors in MS, Epstein Barr virus (EBV), low vitamin D3 and smoking, are all amplified by DR15.
This core issue of research on autoimmune diseases and also MS, how disease-associated HLA-class II molecules contribute to disease development at the functional level, will be studied with state-of-the-art methodologies and a series of novel approaches. These will include in silico modeling approaches, studies of self-peptides, T cell receptor (TCR) repertoire and HLA-DR/peptide complexes, clonally expanded T cells from MS brain tissue and hypothesis-open methods such as combinatorial chemistry and tissue-derived cDNA libraries to identify target antigens. Finally, translational studies will investigate the relationship between the above aspects and MS disease heterogeneity and explore antigen-specific tolerization in proof-of concept clinical trials in MS.
Summary
Multiple sclerosis (MS) is a prototypic CD4+ T cell-mediated autoimmune disease that damages the central nervous system. MS affects young adults and women twice as often as men. Neurological deficits cause substantial disability at an early age with high socioeconomic impact.
Both a complex genetic trait and environmental factors are involved in MS etiology. Similar to other autoimmune diseases it has been known for almost 40 years that certain HLA-class II genes, in MS the two DR15 alleles DRB1*15:01 and DRB5*01:01, confer by far most of the genetic risk. Despite this clear role remarkably little is known about the functional contribution of these genes to MS pathogenesis, and this holds also true for all other T cell-mediated autoimmune diseases. It is assumed that the DR15 alleles present peptides from organ-specific self-proteins to T cells and select an autoreactive CD4+ T cell repertoire that can be activated by certain environmental triggers. Interestingly, the effects of the three known environmental risk factors in MS, Epstein Barr virus (EBV), low vitamin D3 and smoking, are all amplified by DR15.
This core issue of research on autoimmune diseases and also MS, how disease-associated HLA-class II molecules contribute to disease development at the functional level, will be studied with state-of-the-art methodologies and a series of novel approaches. These will include in silico modeling approaches, studies of self-peptides, T cell receptor (TCR) repertoire and HLA-DR/peptide complexes, clonally expanded T cells from MS brain tissue and hypothesis-open methods such as combinatorial chemistry and tissue-derived cDNA libraries to identify target antigens. Finally, translational studies will investigate the relationship between the above aspects and MS disease heterogeneity and explore antigen-specific tolerization in proof-of concept clinical trials in MS.
Max ERC Funding
2 368 068 €
Duration
Start date: 2015-01-01, End date: 2019-12-31
Project acronym HOPSEP
Project Harnessing Oxygenic Photosynthesis for Sustainable Energy Production
Researcher (PI) Nathan Nelson
Host Institution (HI) TEL AVIV UNIVERSITY
Country Israel
Call Details Advanced Grant (AdG), LS9, ERC-2011-ADG_20110310
Summary Oxygenic photosynthesis, that takes place in cyanobacteria algae and plants, provides most of the food and fuel on earth. The light stage of this process is driven by two photosystems. Photosystem II (PSII) that oxidizes water to O2 and 4 H+ and photosystem I (PSI) which in the light provides the most negative redox potential in nature that can drive numerous reactions including CO2 assimilation and hydrogen (H2) production. The structure of most of the complexes involved in oxygenic photosynthesis was solved in several laboratories including our own. Utilizing our plant PSI crystals we were able to generate a light dependent electric potential of up to 100 V. We will develop this system for designing biological based photoelectric devices. Recently, we discovered a marine phage that carries an operon encoding all PSI subunits. Generation, in synechocystis, of a phage-like PSI enabled the mutated complex to accept electrons from additional sources like respiratory cytochromes. This way a novel photorespiration, where PSI can substitute for cytochrome oxidase, is created. The wild type and mutant synechocystis PSI were crystallized and solved, confirming the suggested structural consequences. Moreover, several structural alterations in the mesophilic PSI were recorded. We designed a hydrogen producing bioreactor where the novel photorespiration will enable to utilize the organic material of the cell as an electron source for H2 production. We propose that in conjunction of engineering a Cyanobacterium strain with a temperature sensitive PSII, enhancing rates in its respiratory chain an efficient and sustainable hydrogen production can be achieved.
Summary
Oxygenic photosynthesis, that takes place in cyanobacteria algae and plants, provides most of the food and fuel on earth. The light stage of this process is driven by two photosystems. Photosystem II (PSII) that oxidizes water to O2 and 4 H+ and photosystem I (PSI) which in the light provides the most negative redox potential in nature that can drive numerous reactions including CO2 assimilation and hydrogen (H2) production. The structure of most of the complexes involved in oxygenic photosynthesis was solved in several laboratories including our own. Utilizing our plant PSI crystals we were able to generate a light dependent electric potential of up to 100 V. We will develop this system for designing biological based photoelectric devices. Recently, we discovered a marine phage that carries an operon encoding all PSI subunits. Generation, in synechocystis, of a phage-like PSI enabled the mutated complex to accept electrons from additional sources like respiratory cytochromes. This way a novel photorespiration, where PSI can substitute for cytochrome oxidase, is created. The wild type and mutant synechocystis PSI were crystallized and solved, confirming the suggested structural consequences. Moreover, several structural alterations in the mesophilic PSI were recorded. We designed a hydrogen producing bioreactor where the novel photorespiration will enable to utilize the organic material of the cell as an electron source for H2 production. We propose that in conjunction of engineering a Cyanobacterium strain with a temperature sensitive PSII, enhancing rates in its respiratory chain an efficient and sustainable hydrogen production can be achieved.
Max ERC Funding
2 487 000 €
Duration
Start date: 2012-01-01, End date: 2017-12-31
Project acronym HOWPER
Project An open or closed process: Determining the global scheme of perception
Researcher (PI) Ehud AHISSAR
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Country Israel
Call Details Advanced Grant (AdG), LS5, ERC-2017-ADG
Summary Despite decades of intensive research, there is no agreement about the general scheme of perception: Is the external object a trigger for a brain-internal process (open-loop perception, OLP) or is the object included in brain dynamics during the entire perceptual process (closed-loop perception, CLP)? HOWPER is designed to provide a definite answer to this question in the cases of human touch and vision. What enables this critical test is our development of an explicit CLP hypothesis, which will be contrasted, via specific testable predictions, with the OLP scheme. In the event that CLP is validated, HOWPER will introduce a radical paradigm shift in the study of perception, since almost all current experiments are guided, implicitly or explicitly, by the OLP scheme. If OLP is confirmed, HOWPER will provide the first formal affirmation for its superiority over CLP.
Our approach in this novel paradigm is based on a triangle of interactive efforts comprising theory, analytical experiments, and synthetic experiments. The theoretical effort (WP1) will be based on the core theoretical framework already developed in our lab. The analytical experiments (WP2) will involve human perceivers. The synthetic experiments (WP3) will be performed on synthesized artificial perceivers. The fourth WP will exploit our novel rat-machine hybrid model for testing the neural applicability of the insights gained in the other WPs, whereas the fifth WP will translate our insights into novel visual-to-tactile sensory substitution algorithms.
HOWPER is expected to either revolutionize or significantly advance the field of human perception, to greatly improve visual to tactile sensory substitution approaches and to contribute novel biomimetic algorithms for autonomous robotic agents.
Summary
Despite decades of intensive research, there is no agreement about the general scheme of perception: Is the external object a trigger for a brain-internal process (open-loop perception, OLP) or is the object included in brain dynamics during the entire perceptual process (closed-loop perception, CLP)? HOWPER is designed to provide a definite answer to this question in the cases of human touch and vision. What enables this critical test is our development of an explicit CLP hypothesis, which will be contrasted, via specific testable predictions, with the OLP scheme. In the event that CLP is validated, HOWPER will introduce a radical paradigm shift in the study of perception, since almost all current experiments are guided, implicitly or explicitly, by the OLP scheme. If OLP is confirmed, HOWPER will provide the first formal affirmation for its superiority over CLP.
Our approach in this novel paradigm is based on a triangle of interactive efforts comprising theory, analytical experiments, and synthetic experiments. The theoretical effort (WP1) will be based on the core theoretical framework already developed in our lab. The analytical experiments (WP2) will involve human perceivers. The synthetic experiments (WP3) will be performed on synthesized artificial perceivers. The fourth WP will exploit our novel rat-machine hybrid model for testing the neural applicability of the insights gained in the other WPs, whereas the fifth WP will translate our insights into novel visual-to-tactile sensory substitution algorithms.
HOWPER is expected to either revolutionize or significantly advance the field of human perception, to greatly improve visual to tactile sensory substitution approaches and to contribute novel biomimetic algorithms for autonomous robotic agents.
Max ERC Funding
2 493 441 €
Duration
Start date: 2018-06-01, End date: 2023-11-30
Project acronym HUCNC
Project Conserved Non-Coding Sequences; function, variability and phenotypic consequences
Researcher (PI) Stylianos Antonarakis
Host Institution (HI) UNIVERSITE DE GENEVE
Country Switzerland
Call Details Advanced Grant (AdG), LS2, ERC-2009-AdG
Summary Comparative genomics revealed that ~5% of the human genome is conserved among mammals. This fraction is likely functional, and could harbor pathogenic mutations. We have shown (Nature 2002, Science 2003) that more than half of the constrained fraction of the genome consists of Conserved Non-Coding sequences (CNCs). Model organisms provided evidence for enhancer activity for a fraction of CNCs; in addition another fraction is part of large non-coding RNAs (lincRNA). However, the function of the majority of CNCs is unknown. Importantly, a few pathogenic mutations in CNCs have been associated with genetic disorders. We propose to i) perform functional analysis of CNCs, and ii) identify the spectrum of pathogenic CNC mutations in recognizable human phenotypes. The aims are: 1. Functional genomic connectivity of CNCs 1a. Use 4C in CNCs in various cell types and determine their physical genomic interactions. 1b. Perform targeted disruption of CNCs in cells and assess the functional outcomes. 2. Pathogenic variation of CNCs 2a. Assess the common variation in CNCs: i) common deletion/insertions in 350 samples by aCGH of all human CNCs; ii) common SNP/small indels using DNA selection and High Throughput Sequencing (HTS) of CNCs in 100 samples. 2b. Identify likely pathogenic mutations in developmental syndromes. Search for i) large deletions and duplications of CNCs using aCGH in 1500 samples with malformation syndromes, 1000 from spontaneous abortions, and 500 with X-linked mental retardation; and ii) point mutations in these samples by targeted HTS. The distinction between pathogenic and non-pathogenic variants is difficult, and we propose approaches to meet the challenge. 3. Genetic control (cis and trans eQTLs) of expression variation of CNC lincRNAs, using 200 samples.
Summary
Comparative genomics revealed that ~5% of the human genome is conserved among mammals. This fraction is likely functional, and could harbor pathogenic mutations. We have shown (Nature 2002, Science 2003) that more than half of the constrained fraction of the genome consists of Conserved Non-Coding sequences (CNCs). Model organisms provided evidence for enhancer activity for a fraction of CNCs; in addition another fraction is part of large non-coding RNAs (lincRNA). However, the function of the majority of CNCs is unknown. Importantly, a few pathogenic mutations in CNCs have been associated with genetic disorders. We propose to i) perform functional analysis of CNCs, and ii) identify the spectrum of pathogenic CNC mutations in recognizable human phenotypes. The aims are: 1. Functional genomic connectivity of CNCs 1a. Use 4C in CNCs in various cell types and determine their physical genomic interactions. 1b. Perform targeted disruption of CNCs in cells and assess the functional outcomes. 2. Pathogenic variation of CNCs 2a. Assess the common variation in CNCs: i) common deletion/insertions in 350 samples by aCGH of all human CNCs; ii) common SNP/small indels using DNA selection and High Throughput Sequencing (HTS) of CNCs in 100 samples. 2b. Identify likely pathogenic mutations in developmental syndromes. Search for i) large deletions and duplications of CNCs using aCGH in 1500 samples with malformation syndromes, 1000 from spontaneous abortions, and 500 with X-linked mental retardation; and ii) point mutations in these samples by targeted HTS. The distinction between pathogenic and non-pathogenic variants is difficult, and we propose approaches to meet the challenge. 3. Genetic control (cis and trans eQTLs) of expression variation of CNC lincRNAs, using 200 samples.
Max ERC Funding
2 353 920 €
Duration
Start date: 2010-07-01, End date: 2015-06-30
Project acronym HURET
Project The human retina at single cell resolution: functional architecture, disease mechanism and therapy development
Researcher (PI) Botond Roska
Host Institution (HI) INSTITUT FUR MOLEKULARE UND KLINISCHE OPHTHALMOLOGIE BASEL
Country Switzerland
Call Details Advanced Grant (AdG), LS5, ERC-2019-ADG
Summary Vision is of key importance for humans and losing vision has a major effect on day-to-day life. Vision starts in the retina, where an image captured by photoreceptors is processed by retinal circuits built from more than hundred cell types. Information flows from the retina via the thalamus to a number of cortical areas. Despite the large number of cortical neurons involved in vision, most blinding diseases originate in the retina and are cell-type specific. Although the vertebrate retina has a conserved cellular architecture, only a few animal models of visual diseases reproduce the pathology found in humans. Therefore, there is a major need for understanding the healthy and the disease-affected human retina. Recently my laboratory developed a set of new technologies that enable us to study the human retina, to understand its functional architecture and disease mechanism in its cell types, and so to develop therapies. Using these technologies, we first aim to describe the functional diversity as well as the function of ganglion cell types and their circuits in the human retina. Second, we aim to reveal mechanisms of cell-type vulnerability in human and mouse retinas. Third, we aim to provide proof of principle for cell type-targeted near infrared vision restoration in the human retina. Taken together, this study will provide insights into the structure, function, and mechanisms of disease of the cell types in the human visual system and will investigate a new approach to restore vision in patients with blinding diseases.
Summary
Vision is of key importance for humans and losing vision has a major effect on day-to-day life. Vision starts in the retina, where an image captured by photoreceptors is processed by retinal circuits built from more than hundred cell types. Information flows from the retina via the thalamus to a number of cortical areas. Despite the large number of cortical neurons involved in vision, most blinding diseases originate in the retina and are cell-type specific. Although the vertebrate retina has a conserved cellular architecture, only a few animal models of visual diseases reproduce the pathology found in humans. Therefore, there is a major need for understanding the healthy and the disease-affected human retina. Recently my laboratory developed a set of new technologies that enable us to study the human retina, to understand its functional architecture and disease mechanism in its cell types, and so to develop therapies. Using these technologies, we first aim to describe the functional diversity as well as the function of ganglion cell types and their circuits in the human retina. Second, we aim to reveal mechanisms of cell-type vulnerability in human and mouse retinas. Third, we aim to provide proof of principle for cell type-targeted near infrared vision restoration in the human retina. Taken together, this study will provide insights into the structure, function, and mechanisms of disease of the cell types in the human visual system and will investigate a new approach to restore vision in patients with blinding diseases.
Max ERC Funding
2 500 000 €
Duration
Start date: 2020-11-01, End date: 2025-10-31
Project acronym ICEBERG
Project Discovery of Type 2 Diabetes Targets
Researcher (PI) Juleen Rae Zierath
Host Institution (HI) KAROLINSKA INSTITUTET
Country Sweden
Call Details Advanced Grant (AdG), LS4, ERC-2008-AdG
Summary This proposal is focused on the identification and biological validation of the metabolic pathways and key regulatory genes that control insulin sensitivity in Type 2 diabetes mellitus (T2DM). We are focusing on skeletal muscle because it is quantitatively the most important tissue involved in maintaining glucose homeostasis under insulin-stimulated conditions and it is a major site of insulin resistance in T2DM. Our central hypothesis is that alterations in insulin signal transduction to glucose transport contribute to the profound impairment in whole body glucose homeostasis and T2DM pathogenesis. Identification of the defects in T2DM can lead to the development of new therapeutic strategies to prevent and cure this disease. The proposal is divided into two main objectives: We will apply: 1) target identification platforms including microarray, proteomics and bioinformatics to identify dysregulated genes in normal glucose tolerant versus T2DM subjects or genetically modified model systems and 2) functional genomics to assign a physiological role of the identified targets in Aim 1 using cellular and whole-body approaches. We will focus on the mitogen-activated protein kinase family, the energy-sensing enzyme AMP-activated protein kinase, and the lipid intermediate metabolizing enzyme diacylglycerol kinase delta. Our previous work indicates that these candidates play a role in the regulation of glucose metabolism, triglyceride storage, and energy homeostasis.
Summary
This proposal is focused on the identification and biological validation of the metabolic pathways and key regulatory genes that control insulin sensitivity in Type 2 diabetes mellitus (T2DM). We are focusing on skeletal muscle because it is quantitatively the most important tissue involved in maintaining glucose homeostasis under insulin-stimulated conditions and it is a major site of insulin resistance in T2DM. Our central hypothesis is that alterations in insulin signal transduction to glucose transport contribute to the profound impairment in whole body glucose homeostasis and T2DM pathogenesis. Identification of the defects in T2DM can lead to the development of new therapeutic strategies to prevent and cure this disease. The proposal is divided into two main objectives: We will apply: 1) target identification platforms including microarray, proteomics and bioinformatics to identify dysregulated genes in normal glucose tolerant versus T2DM subjects or genetically modified model systems and 2) functional genomics to assign a physiological role of the identified targets in Aim 1 using cellular and whole-body approaches. We will focus on the mitogen-activated protein kinase family, the energy-sensing enzyme AMP-activated protein kinase, and the lipid intermediate metabolizing enzyme diacylglycerol kinase delta. Our previous work indicates that these candidates play a role in the regulation of glucose metabolism, triglyceride storage, and energy homeostasis.
Max ERC Funding
2 500 000 €
Duration
Start date: 2009-01-01, End date: 2013-12-31
Project acronym IMMUNE/MEMORY AGING
Project Can immune system rejuvenation restore age-related memory loss?
Researcher (PI) Michal Eisenbach-Schwartz
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Country Israel
Call Details Advanced Grant (AdG), LS5, ERC-2008-AdG
Summary With increased life expectancy, there has been a critical growth in the portion of the population that suffers from age-related cognitive decline and dementia. Attempts are therefore being made to find ways to slow brain-aging processes; successful therapies would have a significant impact on the quality of life of individuals, and decrease healthcare expenditures. Aging of the immune system has never been suggested as a factor in memory loss. My group formulated the concept of protective autoimmunity , suggesting a linkage between immunity and self-maintenance in the context of the brain in health and disease. Recently, we showed that T lymphocytes recognizing brain-self antigens have a pivotal role in maintaining hippocampal plasticity, as manifested by reduced neurogenesis and impaired cognitive abilities in T-cell deficient mice. Taken together, our novel observations that T cell immunity contributes to hippocampal plasticity, and the fact that T cell immunity decreases with progressive aging create the basis for the present proposal. We will focus on the following questions: (a) Which aspects of cognition are supported by the immune system- learning, memory or both; (b) whether aging of the immune system is sufficient to induce aging of the brain; (c) whether activation of the immune system is sufficient to reverse age-related cognitive decline; (d) the mechanism underlying the effect of peripheral immunity on brain cognition; and (e) potential therapeutic implications of our findings. Our preliminary results demonstrate that the immune system contributes to spatial memory, and that imposing an immune deficiency is sufficient to cause a reversible memory deficit. These findings give strong reason for optimism that memory loss in the elderly is preventable and perhaps reversible by immune-based therapies; we hope that, in the not too distant future, our studies will enable development of a vaccine to prevent CNS aging and cognitive loss in elderly.
Summary
With increased life expectancy, there has been a critical growth in the portion of the population that suffers from age-related cognitive decline and dementia. Attempts are therefore being made to find ways to slow brain-aging processes; successful therapies would have a significant impact on the quality of life of individuals, and decrease healthcare expenditures. Aging of the immune system has never been suggested as a factor in memory loss. My group formulated the concept of protective autoimmunity , suggesting a linkage between immunity and self-maintenance in the context of the brain in health and disease. Recently, we showed that T lymphocytes recognizing brain-self antigens have a pivotal role in maintaining hippocampal plasticity, as manifested by reduced neurogenesis and impaired cognitive abilities in T-cell deficient mice. Taken together, our novel observations that T cell immunity contributes to hippocampal plasticity, and the fact that T cell immunity decreases with progressive aging create the basis for the present proposal. We will focus on the following questions: (a) Which aspects of cognition are supported by the immune system- learning, memory or both; (b) whether aging of the immune system is sufficient to induce aging of the brain; (c) whether activation of the immune system is sufficient to reverse age-related cognitive decline; (d) the mechanism underlying the effect of peripheral immunity on brain cognition; and (e) potential therapeutic implications of our findings. Our preliminary results demonstrate that the immune system contributes to spatial memory, and that imposing an immune deficiency is sufficient to cause a reversible memory deficit. These findings give strong reason for optimism that memory loss in the elderly is preventable and perhaps reversible by immune-based therapies; we hope that, in the not too distant future, our studies will enable development of a vaccine to prevent CNS aging and cognitive loss in elderly.
Max ERC Funding
1 650 000 €
Duration
Start date: 2009-01-01, End date: 2012-12-31
Project acronym ImmuneCheckpointsAD
Project Immune checkpoint blockade for fighting Alzheimer’s disease
Researcher (PI) Michal EISENBACH-SCHWARTZ
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Country Israel
Call Details Advanced Grant (AdG), LS5, ERC-2016-ADG
Summary Understanding, and ultimately treating Alzheimer’s disease (AD) is a major need in Western countries. Currently, there is no available treatment to modify the disease. Several pioneering discoveries made by my team, attributing a key role to systemic immunity in brain maintenance and repair, and identifying unique interface between the brain’s borders through which the immune system assists the brain, led us to our recent discovery that transient reduction of systemic immune suppression could modify disease pathology, and reverse cognitive loss in mouse models of AD (Nature Communications, 2015; Nature Medicine, 2016; Science, 2014). This discovery emphasizes that AD is not restricted to the brain, but is associated with systemic immune dysfunction. Thus, the goal of addressing numerous risk factors that go awry in the AD brain, many of which are -as yet- unknown, could be accomplished by immunotherapy, using immune checkpoint blockade directed at the Programmed-death (PD)-1 pathway, to empower the immune system. In this proposal, we will adopt our new experimental paradigm to discover mechanisms through which the immune system supports the brain, and to identify key/novel molecular and cellular processes at various stages of the disease that are responsible for cognitive decline long before neurons are lost, and whose reversal or modification is needed to mitigate AD pathology, and prevent cognitive loss. Achieving our goals requires the multidisciplinary approaches and expertise at our disposal, including state-of-the art immunological, cellular, molecular, and genomic tools. The results will pave the way for developing a novel next-generation immunotherapy, by targeting additional selective immune checkpoint pathways, or identifying a specific immune-based therapeutic target, for prevention and treatment of AD. We expect that our results will help attain the ultimate goal of converting an escalating untreatable disease into a chronic treatable one.
Summary
Understanding, and ultimately treating Alzheimer’s disease (AD) is a major need in Western countries. Currently, there is no available treatment to modify the disease. Several pioneering discoveries made by my team, attributing a key role to systemic immunity in brain maintenance and repair, and identifying unique interface between the brain’s borders through which the immune system assists the brain, led us to our recent discovery that transient reduction of systemic immune suppression could modify disease pathology, and reverse cognitive loss in mouse models of AD (Nature Communications, 2015; Nature Medicine, 2016; Science, 2014). This discovery emphasizes that AD is not restricted to the brain, but is associated with systemic immune dysfunction. Thus, the goal of addressing numerous risk factors that go awry in the AD brain, many of which are -as yet- unknown, could be accomplished by immunotherapy, using immune checkpoint blockade directed at the Programmed-death (PD)-1 pathway, to empower the immune system. In this proposal, we will adopt our new experimental paradigm to discover mechanisms through which the immune system supports the brain, and to identify key/novel molecular and cellular processes at various stages of the disease that are responsible for cognitive decline long before neurons are lost, and whose reversal or modification is needed to mitigate AD pathology, and prevent cognitive loss. Achieving our goals requires the multidisciplinary approaches and expertise at our disposal, including state-of-the art immunological, cellular, molecular, and genomic tools. The results will pave the way for developing a novel next-generation immunotherapy, by targeting additional selective immune checkpoint pathways, or identifying a specific immune-based therapeutic target, for prevention and treatment of AD. We expect that our results will help attain the ultimate goal of converting an escalating untreatable disease into a chronic treatable one.
Max ERC Funding
2 287 500 €
Duration
Start date: 2017-06-01, End date: 2022-11-30
Project acronym IMMUNEDIVERSITY
Project Defining of human adaptive immune gene diversity and its impact on disease
Researcher (PI) Gunilla KARLSSON HEDESTAM
Host Institution (HI) KAROLINSKA INSTITUTET
Country Sweden
Call Details Advanced Grant (AdG), LS6, ERC-2017-ADG
Summary Why do vaccines protect some people but not all? Why can some people develop potent neutralizing antibodies to infections, while others cannot? Why do some people develop immune-associated diseases such as allergy, rheumatoid arthritis, multiple sclerosis and diabetes over their lifetimes, when the body has evolved to tolerate self? The answer lies in the adaptive immune system. B and T lymphocytes of the adaptive system express highly polymorphic receptors that allow for the recognition of large numbers of antigens. Our research is now uncovering an enormous heterogeneity in the germline genes that encode our B and T cell receptors. This fact has not been well appreciated because of the high complexity of the genomic regions that encode these receptors, with the presence of large insertions and deletions, a high degree of repetitiveness and gene copy number variations, which cannot be adequately met with conventional whole genome sequencing approaches.
We have developed a state-of-the-art approach we call IgDiscover, which comprises novel molecular library preparation approaches, next generation immune repertoire sequencing, computational analysis and a software that allows rapid construction of personalized antibody gene databases encompassing the hundreds of germline gene segments that rearrange to make up each individual’s naïve B cell repertoire. Thus, IgDiscover offers new possibilities to define human genetic diversity in these loci. This proposal also describes our development of ImmuneDiscover, a high throughput approach enabling personalized immune-profiling of very large numbers of individuals (>1000), encompassing not only Ig genes but also T cell receptor genes and the genes encoding the human major histocompatibility complexes. Here, we will use IgDiscover and ImmuneDiscover to elucidate global diversity in adaptive immune genes and we will investigate potential associations between antibody germline genes and the development of rheumatoid arthritis.
Summary
Why do vaccines protect some people but not all? Why can some people develop potent neutralizing antibodies to infections, while others cannot? Why do some people develop immune-associated diseases such as allergy, rheumatoid arthritis, multiple sclerosis and diabetes over their lifetimes, when the body has evolved to tolerate self? The answer lies in the adaptive immune system. B and T lymphocytes of the adaptive system express highly polymorphic receptors that allow for the recognition of large numbers of antigens. Our research is now uncovering an enormous heterogeneity in the germline genes that encode our B and T cell receptors. This fact has not been well appreciated because of the high complexity of the genomic regions that encode these receptors, with the presence of large insertions and deletions, a high degree of repetitiveness and gene copy number variations, which cannot be adequately met with conventional whole genome sequencing approaches.
We have developed a state-of-the-art approach we call IgDiscover, which comprises novel molecular library preparation approaches, next generation immune repertoire sequencing, computational analysis and a software that allows rapid construction of personalized antibody gene databases encompassing the hundreds of germline gene segments that rearrange to make up each individual’s naïve B cell repertoire. Thus, IgDiscover offers new possibilities to define human genetic diversity in these loci. This proposal also describes our development of ImmuneDiscover, a high throughput approach enabling personalized immune-profiling of very large numbers of individuals (>1000), encompassing not only Ig genes but also T cell receptor genes and the genes encoding the human major histocompatibility complexes. Here, we will use IgDiscover and ImmuneDiscover to elucidate global diversity in adaptive immune genes and we will investigate potential associations between antibody germline genes and the development of rheumatoid arthritis.
Max ERC Funding
2 500 000 €
Duration
Start date: 2018-12-01, End date: 2023-11-30
Project acronym IMMUNEXPLORE
Project New approaches to analyze and exploit the human B and T cell response against viruses
Researcher (PI) Antonio Lanzavecchia
Host Institution (HI) FONDAZIONE PER L ISTITUTO DI RICERCA IN BIOMEDICINA
Country Switzerland
Call Details Advanced Grant (AdG), LS6, ERC-2009-AdG
Summary Immunological memory confers long term protection against pathogens and is the basis of successful vaccination.
Following antigenic stimulation long lived plasma cells and memory B cells are maintained for a lifetime, conferring immediate protection and enhanced responsiveness to the eliciting antigen. However, in the case of variable pathogens such as influenza virus, B cell memory is only partially effective, depending on the extent of similarity between the preceding and the new viruses. The B cell response is dominated by serotype-specific antibodies and heterosubtypic antibodies capable of neutralizing several serotypes appear to be extremely rare.
Understanding the basis of broadly neutralizing antibody responses is a critical aspect for the development of more effective vaccines. In this project we will explore the specificity and dynamics of human antibody responses to influenza virus by using newly developed technological platforms to culture human B cells and plasma cells and to analyze the repertoire of human naïve and memory T cells. High throughput functional screenings, structural analysis and testing in animal models will provide a thorough characterization of the human immune response. The B cell and T cell analysis aims at understanding fundamental aspects of the immune response such as: the selection and diversification of memory B cells; the individual variability of the antibody response, the mechanisms of T-B cooperation and the consequences of the original antigenic sin and of aging on the immune response. This analysis will be complemented by a translational approach whereby broadly neutralizing human monoclonal antibodies will be developed and used: i) for passive vaccination against highly variable viruses; ii) for vaccine design through the identification and production of recombinant antigens to be used as effective vaccines; and iii) for active vaccination in order to facilitate T cell priming and jump start the immune responses.
Summary
Immunological memory confers long term protection against pathogens and is the basis of successful vaccination.
Following antigenic stimulation long lived plasma cells and memory B cells are maintained for a lifetime, conferring immediate protection and enhanced responsiveness to the eliciting antigen. However, in the case of variable pathogens such as influenza virus, B cell memory is only partially effective, depending on the extent of similarity between the preceding and the new viruses. The B cell response is dominated by serotype-specific antibodies and heterosubtypic antibodies capable of neutralizing several serotypes appear to be extremely rare.
Understanding the basis of broadly neutralizing antibody responses is a critical aspect for the development of more effective vaccines. In this project we will explore the specificity and dynamics of human antibody responses to influenza virus by using newly developed technological platforms to culture human B cells and plasma cells and to analyze the repertoire of human naïve and memory T cells. High throughput functional screenings, structural analysis and testing in animal models will provide a thorough characterization of the human immune response. The B cell and T cell analysis aims at understanding fundamental aspects of the immune response such as: the selection and diversification of memory B cells; the individual variability of the antibody response, the mechanisms of T-B cooperation and the consequences of the original antigenic sin and of aging on the immune response. This analysis will be complemented by a translational approach whereby broadly neutralizing human monoclonal antibodies will be developed and used: i) for passive vaccination against highly variable viruses; ii) for vaccine design through the identification and production of recombinant antigens to be used as effective vaccines; and iii) for active vaccination in order to facilitate T cell priming and jump start the immune responses.
Max ERC Funding
1 979 200 €
Duration
Start date: 2010-09-01, End date: 2015-08-31
Project acronym INPHORS
Project Intracellular phosphate reception and signaling: A novel homeostatic system with roles for an orphan organelle?
Researcher (PI) Andreas MAYER
Host Institution (HI) UNIVERSITE DE LAUSANNE
Country Switzerland
Call Details Advanced Grant (AdG), LS3, ERC-2017-ADG
Summary Cells face a phosphate challenge. Growth requires a minimal concentration of this limiting resource because intracellular phosphate (Pi) is a compound of nucleic acids and modifies most cellular proteins. At the same time, cytosolic Pi may not rise much, because elevated cytosolic Pi can stall metabolism. It reduces the free energy that nucleotide triphosphate hydrolysis can provide to drive energetically unfavorable reactions.
I will undertake a pioneering study to elucidate how cells strike this critical balance. We will identify a novel pathway for intracellular phosphate reception and signaling (INPHORS) and explore the role of acidocalcisomes in it. These studies may identify a key function of these very poorly understood organelles, provide one reason for their evolutionary conservation and elucidate a novel homeostatic system of critical importance for cellular metabolism.
We recently provided first hints that a dedicated pathway for sensing and signaling intracellular Pi might exist, which regulates multiple systems for import, export and acidocalcisomal storage of Pi, such that cytosolic Pi homeostasis is guaranteed 1. Yeast cells will serve as an powerful model system for exploring this pathway and its physiological relevance. Yeast Pi transport and storage proteins are known. Furthermore, we can establish cell-free in vitro systems that reconstitute Pi-regulated transport and storage processes, providing an excellent basis for identifying signaling complexes and studying their dynamics.
We will (A) generate novel tools to uncouple, individually manipulate and measure key parameters for the INPHORS pathway; (B) identify its components, study their interactions and regulation; (C) elucidate how acidocalcisomes are targeted by INPHORS and how they contribute to Pi homeostasis; (D) study the crosstalk between INPHORS and Pi-regulated transcriptional responses; (E) test the relevance of INPHORS for Pi homeostasis in mammalian cells.
Summary
Cells face a phosphate challenge. Growth requires a minimal concentration of this limiting resource because intracellular phosphate (Pi) is a compound of nucleic acids and modifies most cellular proteins. At the same time, cytosolic Pi may not rise much, because elevated cytosolic Pi can stall metabolism. It reduces the free energy that nucleotide triphosphate hydrolysis can provide to drive energetically unfavorable reactions.
I will undertake a pioneering study to elucidate how cells strike this critical balance. We will identify a novel pathway for intracellular phosphate reception and signaling (INPHORS) and explore the role of acidocalcisomes in it. These studies may identify a key function of these very poorly understood organelles, provide one reason for their evolutionary conservation and elucidate a novel homeostatic system of critical importance for cellular metabolism.
We recently provided first hints that a dedicated pathway for sensing and signaling intracellular Pi might exist, which regulates multiple systems for import, export and acidocalcisomal storage of Pi, such that cytosolic Pi homeostasis is guaranteed 1. Yeast cells will serve as an powerful model system for exploring this pathway and its physiological relevance. Yeast Pi transport and storage proteins are known. Furthermore, we can establish cell-free in vitro systems that reconstitute Pi-regulated transport and storage processes, providing an excellent basis for identifying signaling complexes and studying their dynamics.
We will (A) generate novel tools to uncouple, individually manipulate and measure key parameters for the INPHORS pathway; (B) identify its components, study their interactions and regulation; (C) elucidate how acidocalcisomes are targeted by INPHORS and how they contribute to Pi homeostasis; (D) study the crosstalk between INPHORS and Pi-regulated transcriptional responses; (E) test the relevance of INPHORS for Pi homeostasis in mammalian cells.
Max ERC Funding
2 499 998 €
Duration
Start date: 2018-09-01, End date: 2023-08-31