Project acronym SELECTIONDRIVEN
Project Gaining insights into human evolution and disease prevention from adaptive natural selection driven by lethal epidemics
Researcher (PI) Ida MOLTKE
Host Institution (HI) KOBENHAVNS UNIVERSITET
Call Details Starting Grant (StG), LS8, ERC-2018-STG
Summary Background
Lethal epidemics like the Black Death have killed millions of people and must pose an extreme selective pressure on any genetic variant that confers disease protection. Therefore, such epidemics have been hypothesized to play a key role in the evolution of humans. To what extent this is true, is a fundamental question of wide interest. Yet, it remains unanswered, in part due to limitations of the current methods to detect signatures of selection.
Objectives
I wish to accomplish three linked goals with the proposed project. The first goal is to develop new statistical methods for detecting signatures of adaptive natural selection driven by lethal epidemics. The second goal is to use these new methods to investigate the role of epidemics in recent human evolution by applying them to genetic data from several recent epidemics. The third goal is to gain insights into mechanisms that protect against infectious disease via the identification of genetic variants that have been under selection because they confer disease protection.
Methods
To reach these goals, extensive simulations will be performed to carefully characterize the genetic signatures of adaptive selection acting on a protective genetic variant during an epidemic. Then new statistical methods that can detect these signatures will be developed. Next, the new methods will be applied to several real datasets, including one from a recent Ebola epidemic. Finally, all signatures of selection detected in these real datasets will be further investigated.
Expected outcome and importance
This project will deliver new statistical methods that will move the field of selection studies a substantial step beyond the state-of-the-art by filling an important methodological gap. It will also yield key insights into the role of epidemics in recent evolutionary history. Finally, it has the potential to provide new knowledge on the genetics of disease resistance that could help prevent future lethal epidemics.
Summary
Background
Lethal epidemics like the Black Death have killed millions of people and must pose an extreme selective pressure on any genetic variant that confers disease protection. Therefore, such epidemics have been hypothesized to play a key role in the evolution of humans. To what extent this is true, is a fundamental question of wide interest. Yet, it remains unanswered, in part due to limitations of the current methods to detect signatures of selection.
Objectives
I wish to accomplish three linked goals with the proposed project. The first goal is to develop new statistical methods for detecting signatures of adaptive natural selection driven by lethal epidemics. The second goal is to use these new methods to investigate the role of epidemics in recent human evolution by applying them to genetic data from several recent epidemics. The third goal is to gain insights into mechanisms that protect against infectious disease via the identification of genetic variants that have been under selection because they confer disease protection.
Methods
To reach these goals, extensive simulations will be performed to carefully characterize the genetic signatures of adaptive selection acting on a protective genetic variant during an epidemic. Then new statistical methods that can detect these signatures will be developed. Next, the new methods will be applied to several real datasets, including one from a recent Ebola epidemic. Finally, all signatures of selection detected in these real datasets will be further investigated.
Expected outcome and importance
This project will deliver new statistical methods that will move the field of selection studies a substantial step beyond the state-of-the-art by filling an important methodological gap. It will also yield key insights into the role of epidemics in recent evolutionary history. Finally, it has the potential to provide new knowledge on the genetics of disease resistance that could help prevent future lethal epidemics.
Max ERC Funding
1 499 600 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym SELECTIONINFORMATION
Project Limits to selection in biology and in evolutionary computation
Researcher (PI) Nicholas Hamilton Barton
Host Institution (HI) INSTITUTE OF SCIENCE AND TECHNOLOGYAUSTRIA
Call Details Advanced Grant (AdG), LS8, ERC-2009-AdG
Summary Natural selection is the central concept in biology, and selection is widely used to solve hard computational problems. This proposal aims to deepen our understanding of selection, in both evolutionary biology and evolutionary computation, and to help bring these fields together. On the one hand, population genetics can show how to optimise genetic algorithms, and can inspire new algorithms. On the other, the central problem in evolutionary computation is to optimise the "evolvability" of the algorithms - an issue that has only recently become prominent in biology. Also, computer science may give biologists insight into how selection can concentrate information from the environment into complex organisms, and how organisms can develop under the guidance of their surprisingly small genomes. This project will focus on the factors that limit natural selection: lack of recombination, interaction between genes, and spatial subdivision. Novel techniques will be applied: multilocus algebra, branching processes, an analogy with statistical mechanics, and a new model for population structure. This analysis will be applied to biological and computational problems in parallel, focusing on how recombination aids selection; how epistasis may evolve to facilitate adaptation; and how selection acts in populations subject to frequent extinction and recolonisation. A new optimisation algorithm will be developed, which is amenable to mathematical analysis. Some components are straightforward, whilst others need new ideas, drawn from the interface between population genetics and computer science. Perhaps most challenging will be to understand how selection can so effectively gather information from the environment, so as to construct complex organisms.
Summary
Natural selection is the central concept in biology, and selection is widely used to solve hard computational problems. This proposal aims to deepen our understanding of selection, in both evolutionary biology and evolutionary computation, and to help bring these fields together. On the one hand, population genetics can show how to optimise genetic algorithms, and can inspire new algorithms. On the other, the central problem in evolutionary computation is to optimise the "evolvability" of the algorithms - an issue that has only recently become prominent in biology. Also, computer science may give biologists insight into how selection can concentrate information from the environment into complex organisms, and how organisms can develop under the guidance of their surprisingly small genomes. This project will focus on the factors that limit natural selection: lack of recombination, interaction between genes, and spatial subdivision. Novel techniques will be applied: multilocus algebra, branching processes, an analogy with statistical mechanics, and a new model for population structure. This analysis will be applied to biological and computational problems in parallel, focusing on how recombination aids selection; how epistasis may evolve to facilitate adaptation; and how selection acts in populations subject to frequent extinction and recolonisation. A new optimisation algorithm will be developed, which is amenable to mathematical analysis. Some components are straightforward, whilst others need new ideas, drawn from the interface between population genetics and computer science. Perhaps most challenging will be to understand how selection can so effectively gather information from the environment, so as to construct complex organisms.
Max ERC Funding
1 975 640 €
Duration
Start date: 2010-07-01, End date: 2015-06-30
Project acronym SELFORGANICELL
Project Self-Organization of the Bacterial Cell
Researcher (PI) Martin Loose
Host Institution (HI) Institute of Science and Technology Austria
Call Details Starting Grant (StG), LS1, ERC-2015-STG
Summary One of the most remarkable features of biological systems is their ability to self-organize in space and time. Even a relatively simple cell like the bacterium Escherichia coli has a precisely regulated cellular anatomy, which emerges from dynamic interactions between proteins and the cell membrane. Self-organization allows the cell to perform extremely challenging tasks. For example, for cell division, more than ten different proteins assemble into a complex, yet highly dynamic machine, which controls the invagination of the cell while constantly remodeling itself. Although the individual components involved have been largely identified, how they act together to accomplish this challenge is not understood. It has become clear that sophisticated biochemical networks give rise to intracellular organization, but we have yet to uncover the underlying mechanistic principles.
In this research proposal, I aim to develop a detailed mechanistic understanding of the self-organizing, emergent properties of the cell. To this end, my research group will develop novel in vitro reconstitution experiments combined with high-resolution fluorescence microscopy and theoretical modeling. Following this “bottom-up” approach, we will quantitatively analyze collective protein dynamics and emergent mechanochemical properties of the bacterial cell division machinery. I aim to answer the following fundamental questions:
1) What is the biochemical network giving rise to the dynamic assembly of the divisome?
2) How do the components of the divisome interact to generate force?
3) How do peptidoglycan synthases build the cell wall?
By comparing protein dynamics in vitro with those measured in vivo, we will provide a link between molecular properties and the processes found in the living cell. This project will not only improve our understanding of the bacterial cell, but also open new research avenues for eukaryotic cell biology, synthetic biology and biophysics.
Summary
One of the most remarkable features of biological systems is their ability to self-organize in space and time. Even a relatively simple cell like the bacterium Escherichia coli has a precisely regulated cellular anatomy, which emerges from dynamic interactions between proteins and the cell membrane. Self-organization allows the cell to perform extremely challenging tasks. For example, for cell division, more than ten different proteins assemble into a complex, yet highly dynamic machine, which controls the invagination of the cell while constantly remodeling itself. Although the individual components involved have been largely identified, how they act together to accomplish this challenge is not understood. It has become clear that sophisticated biochemical networks give rise to intracellular organization, but we have yet to uncover the underlying mechanistic principles.
In this research proposal, I aim to develop a detailed mechanistic understanding of the self-organizing, emergent properties of the cell. To this end, my research group will develop novel in vitro reconstitution experiments combined with high-resolution fluorescence microscopy and theoretical modeling. Following this “bottom-up” approach, we will quantitatively analyze collective protein dynamics and emergent mechanochemical properties of the bacterial cell division machinery. I aim to answer the following fundamental questions:
1) What is the biochemical network giving rise to the dynamic assembly of the divisome?
2) How do the components of the divisome interact to generate force?
3) How do peptidoglycan synthases build the cell wall?
By comparing protein dynamics in vitro with those measured in vivo, we will provide a link between molecular properties and the processes found in the living cell. This project will not only improve our understanding of the bacterial cell, but also open new research avenues for eukaryotic cell biology, synthetic biology and biophysics.
Max ERC Funding
1 496 687 €
Duration
Start date: 2016-04-01, End date: 2021-03-31
Project acronym SexAntag
Project Prevalence and Influence of Sexual Antagonism on Genome Evolution
Researcher (PI) Beatriz Barahona Pena Vicoso
Host Institution (HI) INSTITUTE OF SCIENCE AND TECHNOLOGYAUSTRIA
Call Details Starting Grant (StG), LS8, ERC-2016-STG
Summary Males and females display profound differences in phenotype, physiology and behavior, and understanding the evolutionary forces driving this diversity is a long-standing goal in evolutionary biology. Sexually antagonistic conflict, resulting from traits and/or mutations that are beneficial to one sex but harmful to the other, can in theory lead to strong dimorphism, and has been invoked to explain many evolutionary features, including the large number of genes that acquire sex-biased expression. Quantifying it, however, has proved difficult, and only a few experimental studies have attempted to systematically identify genes under sexual conflict. Species groups with both sexual and asexual populations offer a promising approach to tackle this question, as they allow for a direct comparison of gene expression when selection occurs ½ of the time in females and ½ of the time in males (in sexual species), versus a female-only selective regime (asexual species).
Here, we will test the hypothesis that widespread sexual conflict maintains sex-biased gene expression, using the brine shrimp Artemia as a model. Specifically, we will:
1. Compare gene expression levels of closely related Artemia sexual and asexual species, to test if the absence of selection on males (in the female-only species) leads to general shifts in expression, and if these changes are consistent with predictions made under sexual antagonism.
2. Characterize the Z (sex) chromosome of the sexual and asexual species, to test if this chromosome is particularly prone to maintaining genes under sexual conflict, as predicted by theory.
3. Investigate the population genomics profile of genes under sexual conflict, to find signatures of sexual conflict that may be diagnostic for sexually antagonistic genes in other systems.
Together, these analyses will provide us with a global overview of the prevalence of sexual antagonism, and of its influence on gene expression and genome evolution.
Summary
Males and females display profound differences in phenotype, physiology and behavior, and understanding the evolutionary forces driving this diversity is a long-standing goal in evolutionary biology. Sexually antagonistic conflict, resulting from traits and/or mutations that are beneficial to one sex but harmful to the other, can in theory lead to strong dimorphism, and has been invoked to explain many evolutionary features, including the large number of genes that acquire sex-biased expression. Quantifying it, however, has proved difficult, and only a few experimental studies have attempted to systematically identify genes under sexual conflict. Species groups with both sexual and asexual populations offer a promising approach to tackle this question, as they allow for a direct comparison of gene expression when selection occurs ½ of the time in females and ½ of the time in males (in sexual species), versus a female-only selective regime (asexual species).
Here, we will test the hypothesis that widespread sexual conflict maintains sex-biased gene expression, using the brine shrimp Artemia as a model. Specifically, we will:
1. Compare gene expression levels of closely related Artemia sexual and asexual species, to test if the absence of selection on males (in the female-only species) leads to general shifts in expression, and if these changes are consistent with predictions made under sexual antagonism.
2. Characterize the Z (sex) chromosome of the sexual and asexual species, to test if this chromosome is particularly prone to maintaining genes under sexual conflict, as predicted by theory.
3. Investigate the population genomics profile of genes under sexual conflict, to find signatures of sexual conflict that may be diagnostic for sexually antagonistic genes in other systems.
Together, these analyses will provide us with a global overview of the prevalence of sexual antagonism, and of its influence on gene expression and genome evolution.
Max ERC Funding
1 444 394 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym SINCHAIS
Project In situ analysis of single channel subunit composition in neurons: physiological implication in synaptic plasticity and behavior
Researcher (PI) Ryuichi Shigemoto
Host Institution (HI) INSTITUTE OF SCIENCE AND TECHNOLOGYAUSTRIA
Call Details Advanced Grant (AdG), LS5, ERC-2015-AdG
Summary Ligand-gated and voltage-gated channels are key molecules in transforming chemical signals into electrical ones and electrical signals into chemical ones, respectively. At excitatory synaptic connections in the brain, activation of AMPA- and NMDA-type glutamate receptors elicits inward currents at the postsynaptic sites, and activation of voltage-gated calcium channels triggers vesicle release of glutamate in the presynaptic sites. Plastic changes in their number, location and property can lead to potentiation or depression of synaptic efficacy, alteration in time course, and coupling to effectors at both postsynaptic and presynaptic sites. These channels are all composed of distinct subunits and their compositions affect channel properties, trafficking to the synaptic sites, and interaction with associated molecules, creating a large diversity of synaptic functions. Although channels with different subunit compositions have been investigated using biochemical and electrophysiological detection methods, very little is known about single channel subunit composition in situ because of the lack of high resolution methods for analysis of protein complex in intact tissues. In this project, I will develop novel technologies to visualize subunit composition at the single channel level in individual synapses by electron microscopy, combining new EM tags, freeze-fracture replica labelling, and electron tomography. Synaptic plasticity will be induced by optogenetic stimulation of identified neurons or behavioural paradigms to examine the dynamic changes of subunit composition. Finally, physiological implications of such regulation of subunit composition will be investigated by genetic manipulation of mice combined with electrophysiological and behavioural analyses. This work will demonstrate unprecedented views of the subunit composition in situ and provide new insights into how regulation of subunit composition contributes to synaptic plasticity and animal behaviour.
Summary
Ligand-gated and voltage-gated channels are key molecules in transforming chemical signals into electrical ones and electrical signals into chemical ones, respectively. At excitatory synaptic connections in the brain, activation of AMPA- and NMDA-type glutamate receptors elicits inward currents at the postsynaptic sites, and activation of voltage-gated calcium channels triggers vesicle release of glutamate in the presynaptic sites. Plastic changes in their number, location and property can lead to potentiation or depression of synaptic efficacy, alteration in time course, and coupling to effectors at both postsynaptic and presynaptic sites. These channels are all composed of distinct subunits and their compositions affect channel properties, trafficking to the synaptic sites, and interaction with associated molecules, creating a large diversity of synaptic functions. Although channels with different subunit compositions have been investigated using biochemical and electrophysiological detection methods, very little is known about single channel subunit composition in situ because of the lack of high resolution methods for analysis of protein complex in intact tissues. In this project, I will develop novel technologies to visualize subunit composition at the single channel level in individual synapses by electron microscopy, combining new EM tags, freeze-fracture replica labelling, and electron tomography. Synaptic plasticity will be induced by optogenetic stimulation of identified neurons or behavioural paradigms to examine the dynamic changes of subunit composition. Finally, physiological implications of such regulation of subunit composition will be investigated by genetic manipulation of mice combined with electrophysiological and behavioural analyses. This work will demonstrate unprecedented views of the subunit composition in situ and provide new insights into how regulation of subunit composition contributes to synaptic plasticity and animal behaviour.
Max ERC Funding
2 481 437 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym SOCIALVACCINES
Project Social Vaccination in Ant Colonies: from Individual Mechanisms to Society Effects
Researcher (PI) Sylvia Maria Cremer-Sixt
Host Institution (HI) INSTITUTE OF SCIENCE AND TECHNOLOGYAUSTRIA
Call Details Starting Grant (StG), LS8, ERC-2009-StG
Summary Living in social groups leads to a high risk of disease spread. Insect societies counteracted this threat by evolving sophisticated collective anti-parasite defences, combining the immune systems of all group members and their collective social hygienic behaviours. I recently described a direct link between these social and individual defences, by showing that social contact to an infected individual results in a protective Social Vaccination in its nestmate ants: the latter survived an infection with the same pathogen significantly better than control ants that had lived with other healthy ants only. Aim of this project is to elucidate the molecular and physiological mechanisms of this Social Vaccination in individual ants, and its effect on the organisation of collective defences and disease dynamics at the colony level. Preliminary results have shown different protein upregulation in healthy, infected and vaccinated ants, and a detailed analysis of gene and protein expression patterns will define those proteins and their function in anti-parasite defence (postdoc). Observations of the social interactions between infected ants and their group members, and measures of the physiological immune responses of the individuals at different stages of the infection and after the vaccination will further reveal when and how Social Vaccination occurs, how long it endures, and how specific and plastic it is (PhD 1). At the colony level, the social role of vaccinated individuals and the fate of infected ants when loosing their vaccination potential will be determined, as well as the effects of Social Vaccination on disease spread in the colony and the regulation of collective defences between group members (PhD 2). This unique combination studying both the social and individual aspect of immune defences makes this project valuable for the fields of Social Evolution and Ecological Immunology, and further impacts on Epidemiology, Evolutionary Medicine and Biocontrol.
Summary
Living in social groups leads to a high risk of disease spread. Insect societies counteracted this threat by evolving sophisticated collective anti-parasite defences, combining the immune systems of all group members and their collective social hygienic behaviours. I recently described a direct link between these social and individual defences, by showing that social contact to an infected individual results in a protective Social Vaccination in its nestmate ants: the latter survived an infection with the same pathogen significantly better than control ants that had lived with other healthy ants only. Aim of this project is to elucidate the molecular and physiological mechanisms of this Social Vaccination in individual ants, and its effect on the organisation of collective defences and disease dynamics at the colony level. Preliminary results have shown different protein upregulation in healthy, infected and vaccinated ants, and a detailed analysis of gene and protein expression patterns will define those proteins and their function in anti-parasite defence (postdoc). Observations of the social interactions between infected ants and their group members, and measures of the physiological immune responses of the individuals at different stages of the infection and after the vaccination will further reveal when and how Social Vaccination occurs, how long it endures, and how specific and plastic it is (PhD 1). At the colony level, the social role of vaccinated individuals and the fate of infected ants when loosing their vaccination potential will be determined, as well as the effects of Social Vaccination on disease spread in the colony and the regulation of collective defences between group members (PhD 2). This unique combination studying both the social and individual aspect of immune defences makes this project valuable for the fields of Social Evolution and Ecological Immunology, and further impacts on Epidemiology, Evolutionary Medicine and Biocontrol.
Max ERC Funding
1 299 780 €
Duration
Start date: 2010-04-01, End date: 2015-03-31
Project acronym SomSOM
Project Self-organisation of microbial soil organic matter turnover
Researcher (PI) Christina KAISER
Host Institution (HI) UNIVERSITAT WIEN
Call Details Consolidator Grant (CoG), LS8, ERC-2018-COG
Summary Microbial turnover of soil organic matter (SOM) is key for the terrestrial carbon (C) cycle. Its underlying mechanisms, however, are not fully understood. The role of soil microbes for organic matter turnover has so far been studied mainly from the point of view of microbial physiology, stoichiometry or community composition. I propose to shed new light on it from the perspective of complex systems science.
Microbial decomposition of organic matter requires the concerted action of functionally different microbes interacting with each other in a spatially structured environment. From complex systems theory, it is known that interactions among individuals at the microscale can lead to an ‘emergent’ system behavior, or ‘self-organisation’, at the macroscale, which adds a new quality to the system that cannot be derived from the traits of the interacting agents. Importantly, if microbial decomposer systems are self-organised, they may behave in a different way as currently assumed, especially under changing environmental conditions.
The aim of this project is thus to investigate i) if microbial decomposition of organic matter is driven by emergent behaviour, and ii) what consequences this has for soil C and nitrogen cycling. Combining state-of-the-art methods from soil biogeochemistry, microbial ecology, and complex systems science I will
• Investigate mechanisms of spatial self-organization of microbial decomposer communities by linking microscale observations from experimental microcosms to mathematical, individual-based modelling,
• Elucidate microbial interaction networks across the soil’s microarchitecture by linking microbial community composition, process rates and chemical composition of spatially explicit soil micro-units at an unprecedented small and pertinent scale.
• Explore fundamental patterns of self-organisation by applying the framework of complex systems science to high-resolution spatial and temporal data of soil microstructure and process rates.
Summary
Microbial turnover of soil organic matter (SOM) is key for the terrestrial carbon (C) cycle. Its underlying mechanisms, however, are not fully understood. The role of soil microbes for organic matter turnover has so far been studied mainly from the point of view of microbial physiology, stoichiometry or community composition. I propose to shed new light on it from the perspective of complex systems science.
Microbial decomposition of organic matter requires the concerted action of functionally different microbes interacting with each other in a spatially structured environment. From complex systems theory, it is known that interactions among individuals at the microscale can lead to an ‘emergent’ system behavior, or ‘self-organisation’, at the macroscale, which adds a new quality to the system that cannot be derived from the traits of the interacting agents. Importantly, if microbial decomposer systems are self-organised, they may behave in a different way as currently assumed, especially under changing environmental conditions.
The aim of this project is thus to investigate i) if microbial decomposition of organic matter is driven by emergent behaviour, and ii) what consequences this has for soil C and nitrogen cycling. Combining state-of-the-art methods from soil biogeochemistry, microbial ecology, and complex systems science I will
• Investigate mechanisms of spatial self-organization of microbial decomposer communities by linking microscale observations from experimental microcosms to mathematical, individual-based modelling,
• Elucidate microbial interaction networks across the soil’s microarchitecture by linking microbial community composition, process rates and chemical composition of spatially explicit soil micro-units at an unprecedented small and pertinent scale.
• Explore fundamental patterns of self-organisation by applying the framework of complex systems science to high-resolution spatial and temporal data of soil microstructure and process rates.
Max ERC Funding
1 896 129 €
Duration
Start date: 2019-05-01, End date: 2024-04-30
Project acronym sRNA-EMB
Project Small RNA regulation of the body plan and epigenome in Arabidopsis embryos
Researcher (PI) Michael Nodine
Host Institution (HI) GREGOR MENDEL INSTITUT FUR MOLEKULARE PFLANZENBIOLOGIE GMBH
Call Details Starting Grant (StG), LS3, ERC-2014-STG
Summary Small RNAs are short non-coding RNAs that regulate gene expression in plants and animals. Although small RNAs are essential for proper differentiation and epigenome regulation, little is known regarding their embryonic functions, especially in plants. Arabidopsis thaliana is a leading system to study the regulatory roles of small RNAs because of the abundance of genetic, genomic and epigenomic resources. Moreover, Arabidopsis embryos undergo invariant division patterns and rapidly differentiate to generate the most basic plant cell-types arranged in correct positions. Early Arabidopsis embryos are therefore morphologically simple structures composed of diverse cell types making them ideal for determining the influence of small RNAs on fundamental cellular differentiation and reprogramming events. The objectives of the proposed research are designed to assess the regulatory roles of small RNAs in establishing both the basic body plan and epigenome in plant embryos. We will utilize modified next-generation sequencing technologies to identify small RNAs present in developing embryos. Because we will generate these RNA profiles from a mixture of cell-types, we will also use a fluorescent protein-based approach to quantify specific miRNA repressive activities in individual cell-types. To determine the functions of individual miRNA/target interactions during embryogenesis, we will identify miRNAs required for embryo development and use genome-wide approaches to study specific miRNA/target interactions in greater detail. Lastly, we will use a fusion of genetic and genomic methods to determine how small RNAs influence the nascent epigenome during early embryogenesis.
Summary
Small RNAs are short non-coding RNAs that regulate gene expression in plants and animals. Although small RNAs are essential for proper differentiation and epigenome regulation, little is known regarding their embryonic functions, especially in plants. Arabidopsis thaliana is a leading system to study the regulatory roles of small RNAs because of the abundance of genetic, genomic and epigenomic resources. Moreover, Arabidopsis embryos undergo invariant division patterns and rapidly differentiate to generate the most basic plant cell-types arranged in correct positions. Early Arabidopsis embryos are therefore morphologically simple structures composed of diverse cell types making them ideal for determining the influence of small RNAs on fundamental cellular differentiation and reprogramming events. The objectives of the proposed research are designed to assess the regulatory roles of small RNAs in establishing both the basic body plan and epigenome in plant embryos. We will utilize modified next-generation sequencing technologies to identify small RNAs present in developing embryos. Because we will generate these RNA profiles from a mixture of cell-types, we will also use a fluorescent protein-based approach to quantify specific miRNA repressive activities in individual cell-types. To determine the functions of individual miRNA/target interactions during embryogenesis, we will identify miRNAs required for embryo development and use genome-wide approaches to study specific miRNA/target interactions in greater detail. Lastly, we will use a fusion of genetic and genomic methods to determine how small RNAs influence the nascent epigenome during early embryogenesis.
Max ERC Funding
1 499 989 €
Duration
Start date: 2015-07-01, End date: 2020-06-30
Project acronym StAdvInn
Project Strengthening adaptive immunity via innate immunity: enhancing the CD8 T cell response by using the NKG2D ligand expressed in a herpesvirus vector
Researcher (PI) Stipan Jonjic
Host Institution (HI) SVEUCILISTE U RIJECI, MEDICINSKI FAKULTET
Call Details Advanced Grant (AdG), LS6, ERC-2012-ADG_20120314
Summary CD8+ T cells play a key role in the control of infections by intracellular pathogens. Recently, several top-notch studies provided ample evidence that NK cells are important in the regulation of CD8+ T cell response. NKG2D is an activating NK cell receptor which plays a role in the adaptive immune response by co-stimulating CD8+ T cells. Due to unique pattern of immune response, live attenuated CMVs are attractive candidates as vaccine vectors for a number of clinically relevant infections. The main idea behind this project stems from our preliminary data which suggest that a recombinant CMV vector expressing NKG2D ligand has a tremendous potential for subverting viral immunoevasion and boosting the efficiency of CD8 T cell response.
During the project we plan to systematically investigate the impact of all major innate immunity players on the CD8+ T cell response. A special focus will be given in obtaining new knowledge on the maintenance of memory CD8+ T cells during latent infection. This study will also provide novel insights on the role of NKG2D in both NK and T cell immunity. In order to test our hypothesis in vivo, we will employ state-of-the-art technology used in herpesvirus genetics coupled with high-end immune monitoring. Ultimately, we will translate our results to a human CMV vector, in order to gauge the impact of NKG2D signaling on immune response in a humanized mouse model.
We believe that the significance of the proposed study is enormous since stimulating CD8+ T cells has been widely recognized as a method of choice for vaccine development. There are relatively large number of pathogens for which the immunity acquired post-infection does not fully shelter against re-infection and disease. Therefore, we are in a desperate need for vaccines which offer superior protection compared to the one following natural infection. This study will provide groundbreaking information which will set the stage for the development of new vaccines and vaccine vectors.
Summary
CD8+ T cells play a key role in the control of infections by intracellular pathogens. Recently, several top-notch studies provided ample evidence that NK cells are important in the regulation of CD8+ T cell response. NKG2D is an activating NK cell receptor which plays a role in the adaptive immune response by co-stimulating CD8+ T cells. Due to unique pattern of immune response, live attenuated CMVs are attractive candidates as vaccine vectors for a number of clinically relevant infections. The main idea behind this project stems from our preliminary data which suggest that a recombinant CMV vector expressing NKG2D ligand has a tremendous potential for subverting viral immunoevasion and boosting the efficiency of CD8 T cell response.
During the project we plan to systematically investigate the impact of all major innate immunity players on the CD8+ T cell response. A special focus will be given in obtaining new knowledge on the maintenance of memory CD8+ T cells during latent infection. This study will also provide novel insights on the role of NKG2D in both NK and T cell immunity. In order to test our hypothesis in vivo, we will employ state-of-the-art technology used in herpesvirus genetics coupled with high-end immune monitoring. Ultimately, we will translate our results to a human CMV vector, in order to gauge the impact of NKG2D signaling on immune response in a humanized mouse model.
We believe that the significance of the proposed study is enormous since stimulating CD8+ T cells has been widely recognized as a method of choice for vaccine development. There are relatively large number of pathogens for which the immunity acquired post-infection does not fully shelter against re-infection and disease. Therefore, we are in a desperate need for vaccines which offer superior protection compared to the one following natural infection. This study will provide groundbreaking information which will set the stage for the development of new vaccines and vaccine vectors.
Max ERC Funding
1 754 897 €
Duration
Start date: 2013-05-01, End date: 2018-04-30
Project acronym STC
Project Synaptic Tagging and Capture: From Synapses to Behavior
Researcher (PI) Sayyed Mohammad Sadegh Nabavi
Host Institution (HI) AARHUS UNIVERSITET
Call Details Starting Grant (StG), LS5, ERC-2015-STG
Summary It is shown that long-term potentiation (LTP) is the cellular basis of memory formation. However, since all but small fraction of memories are forgotten, LTP has been further divided into early LTP (e-LTP), the mechanism by which short-term memories are formed, and a more stable late LTP (L-LTP), by which long-term memories are formed. Remarkably, it has been shown that an e-LTP can be stabilized if it is preceded or followed by heterosynaptic L-LTP.
According to Synaptic Tagging and Capture (STC) hypothesis, e-LTP is stabilized by capturing proteins that are made by L-LTP induction. The model proposes that this mechanism underlies the formation of late associative memory, where the stability of a memory is not only defined by the stimuli that induce the change but also by events happening before and after these stimuli. As such, the model explicitly predicts that a short-term memory can be stabilized by inducing heterosynaptic L-LTP.
In this grant, I will put this hypothesis into test. Specifically, I will test two explicit predictions of STC model: 1) A naturally formed short-term memory can be stabilized by induction of heterosynaptic L-LTP. 2) This stabilization is caused by the protein synthesis feature of L-LTP. To do this, using optogenetics, I will engineer a short-term memory in auditory fear circuit, in which an animal transiently associates a foot shock to a tone. Subsequently, I will examine if optogenetic delivery of L-LTP to the visual inputs converging on the same population of neurons in the amygdala will stabilize the short-term tone fear memory.
To be able to engineer natural memory by manipulating synaptic plasticity I will develop two systems: 1) A two-color optical activation system which permits selective manipulation of distinct neuronal populations with precise temporal and spatial resolution; 2) An inducible and activity-dependent expression system by which those neurons that are activated by a natural stimulus will be optically tagged.
Summary
It is shown that long-term potentiation (LTP) is the cellular basis of memory formation. However, since all but small fraction of memories are forgotten, LTP has been further divided into early LTP (e-LTP), the mechanism by which short-term memories are formed, and a more stable late LTP (L-LTP), by which long-term memories are formed. Remarkably, it has been shown that an e-LTP can be stabilized if it is preceded or followed by heterosynaptic L-LTP.
According to Synaptic Tagging and Capture (STC) hypothesis, e-LTP is stabilized by capturing proteins that are made by L-LTP induction. The model proposes that this mechanism underlies the formation of late associative memory, where the stability of a memory is not only defined by the stimuli that induce the change but also by events happening before and after these stimuli. As such, the model explicitly predicts that a short-term memory can be stabilized by inducing heterosynaptic L-LTP.
In this grant, I will put this hypothesis into test. Specifically, I will test two explicit predictions of STC model: 1) A naturally formed short-term memory can be stabilized by induction of heterosynaptic L-LTP. 2) This stabilization is caused by the protein synthesis feature of L-LTP. To do this, using optogenetics, I will engineer a short-term memory in auditory fear circuit, in which an animal transiently associates a foot shock to a tone. Subsequently, I will examine if optogenetic delivery of L-LTP to the visual inputs converging on the same population of neurons in the amygdala will stabilize the short-term tone fear memory.
To be able to engineer natural memory by manipulating synaptic plasticity I will develop two systems: 1) A two-color optical activation system which permits selective manipulation of distinct neuronal populations with precise temporal and spatial resolution; 2) An inducible and activity-dependent expression system by which those neurons that are activated by a natural stimulus will be optically tagged.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-04-01, End date: 2021-03-31
Project acronym StemHealth
Project Foetal Intestinal Stem Cells in Biology and Health
Researcher (PI) Kim Bak Jensen
Host Institution (HI) KOBENHAVNS UNIVERSITET
Call Details Consolidator Grant (CoG), LS7, ERC-2015-CoG
Summary There is currently no medical cure for the millions of individuals affected by inflammatory bowel disease (IBD). These patients suffer from bleeding along the gastrointestinal tract due to epithelial ulceration, which causes severe abdominal pain, diarrhoea and malnutrition. This is due to the severely compromised integrity of the intestinal epithelium. I propose that patients with IBD will benefit from an intestinal epithelial transplant.
The objectives of this research programme are two fold. Firstly, I propose to perform preclinical testing of human intestinal epithelium to pave the way for their inclusion in clinical trials for IBD patients. This will be based on a combination of state-of-the-art cell culture methods with novel transplantation methodology. By combining analysis of intestinal epithelial cells from various developmental stages, I will be able to identify the most suitable source for transplantation and define how adult stem cells are specified in the tissue. Secondly, I will utilise an in vitro culture system to identify the transcriptional networks responsible for the maturation of the foetal intestinal epithelium. Tissue maturation currently constitutes a major roadblock in regenerative medicine as cells derived from foetal and pluripotent stem cells have foetal properties. Understanding this process will therefore improve our ability to generate sustainable sources of cells for transplantation, which is pivotal for future therapies relying on regenerative medicine and in vitro modelling of disease
The proposed research programme will have significant clinical and biological impact. Clinically, it provides the framework for initiating clinical trials for patients with IBD and protocols to obtain mature adult epithelium for in vitro disease modelling. From a biological perspective, we will gain insights into how specific signalling networks maintain specific cell states and dictate tissue maturation.
Summary
There is currently no medical cure for the millions of individuals affected by inflammatory bowel disease (IBD). These patients suffer from bleeding along the gastrointestinal tract due to epithelial ulceration, which causes severe abdominal pain, diarrhoea and malnutrition. This is due to the severely compromised integrity of the intestinal epithelium. I propose that patients with IBD will benefit from an intestinal epithelial transplant.
The objectives of this research programme are two fold. Firstly, I propose to perform preclinical testing of human intestinal epithelium to pave the way for their inclusion in clinical trials for IBD patients. This will be based on a combination of state-of-the-art cell culture methods with novel transplantation methodology. By combining analysis of intestinal epithelial cells from various developmental stages, I will be able to identify the most suitable source for transplantation and define how adult stem cells are specified in the tissue. Secondly, I will utilise an in vitro culture system to identify the transcriptional networks responsible for the maturation of the foetal intestinal epithelium. Tissue maturation currently constitutes a major roadblock in regenerative medicine as cells derived from foetal and pluripotent stem cells have foetal properties. Understanding this process will therefore improve our ability to generate sustainable sources of cells for transplantation, which is pivotal for future therapies relying on regenerative medicine and in vitro modelling of disease
The proposed research programme will have significant clinical and biological impact. Clinically, it provides the framework for initiating clinical trials for patients with IBD and protocols to obtain mature adult epithelium for in vitro disease modelling. From a biological perspective, we will gain insights into how specific signalling networks maintain specific cell states and dictate tissue maturation.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-08-01, End date: 2021-07-31
Project acronym STEMMING-FROM-NERVE
Project Targeted Cell Recruitment During Organogenesis And Regeneration: Glia Makes The Tooth
Researcher (PI) Igor Adameyko
Host Institution (HI) MEDIZINISCHE UNIVERSITAET WIEN
Call Details Consolidator Grant (CoG), LS3, ERC-2014-CoG
Summary Recently we discovered an entirely new phenomenon in developmental biology – targeted recruitment of stem cells from the pervasive peripheral nerves. For example, we demonstrated that majority of melanocytes – our pigment cells, are born from peripheral glial cells. To further extend this far-reaching principle we will use tooth development as a model. Current opinion holds that sensory innervation of the tooth has a minor or no role in making a structure of the tooth during development and adulthood. On the contrary, our preliminary data strongly suggest that sensory nerve contributes pulp cells and matrix-producing cells of odontoblast lineage to the growing tooth. Our hypothesis implies that nerve-associated glial cells can be recruited from the nerve by unknown molecules presented inside of the tooth environment, and that these recruited cells are capable of producing pulp cells and odontoblasts.
WE PROPOSE TO ADDRESS THE ROLE OF A SENSORY NERVE AS A PROVIDER OF DENTAL STEM CELLS DURING THE DEVELOPMENT, ADULTHOOD AND REGENERATION.
To experimentally address our hypothesis we developed innovative and powerful approaches: we will use advanced genetics tracing with multicolor reporters, completely novel unconventional individual cell transcriptome analysis, transgenic mice with cell type-specific modifications in signaling, microsurgery and grafting, and, finally, 3D-imaging of developing tooth structures.
This project is interesting to a wide community of scientists because it addresses a novel function of peripheral nervous system which is contributing stem cells as building blocks to local tissues in development and regeneration.
The MEDICAL IMPLICATION of this project will include better understanding of tooth regeneration providing new approaches to dentin recovery and tooth restoration following trauma.
Summary
Recently we discovered an entirely new phenomenon in developmental biology – targeted recruitment of stem cells from the pervasive peripheral nerves. For example, we demonstrated that majority of melanocytes – our pigment cells, are born from peripheral glial cells. To further extend this far-reaching principle we will use tooth development as a model. Current opinion holds that sensory innervation of the tooth has a minor or no role in making a structure of the tooth during development and adulthood. On the contrary, our preliminary data strongly suggest that sensory nerve contributes pulp cells and matrix-producing cells of odontoblast lineage to the growing tooth. Our hypothesis implies that nerve-associated glial cells can be recruited from the nerve by unknown molecules presented inside of the tooth environment, and that these recruited cells are capable of producing pulp cells and odontoblasts.
WE PROPOSE TO ADDRESS THE ROLE OF A SENSORY NERVE AS A PROVIDER OF DENTAL STEM CELLS DURING THE DEVELOPMENT, ADULTHOOD AND REGENERATION.
To experimentally address our hypothesis we developed innovative and powerful approaches: we will use advanced genetics tracing with multicolor reporters, completely novel unconventional individual cell transcriptome analysis, transgenic mice with cell type-specific modifications in signaling, microsurgery and grafting, and, finally, 3D-imaging of developing tooth structures.
This project is interesting to a wide community of scientists because it addresses a novel function of peripheral nervous system which is contributing stem cells as building blocks to local tissues in development and regeneration.
The MEDICAL IMPLICATION of this project will include better understanding of tooth regeneration providing new approaches to dentin recovery and tooth restoration following trauma.
Max ERC Funding
1 964 338 €
Duration
Start date: 2015-08-01, End date: 2020-07-31
Project acronym TACKLE
Project TACK Superphylum and Lokiarchaeota Evolution: Dissecting the Ecology and Evolution of Archaea to Elucidate the Prokaryote to Eukaryote Transition
Researcher (PI) Christa Maria SCHLEPER
Host Institution (HI) UNIVERSITAT WIEN
Call Details Advanced Grant (AdG), LS8, ERC-2015-AdG
Summary The emergence of complex eukaryotic life forms on Earth from prokaryotic cells is one of the most fundamental questions in biology and also one of the least understood transitions in evolution. Phylogenomic studies recently indicated that the eukaryotic line of descent arose from within the TACK+L superphylum of Archaea. This proposal addresses the systematic analysis of two newly discovered, but largely uncharacterized lineages of Archaea from this superphylum that mark crucial evolutionary transitions. Thaumarchaeota harbor unique ‘eukaryotic’ features and represent the sole group of Archaea that has successfully radiated into virtually any moderate habitat on Earth. The Lokiarchaeota lineage found recently in deep marine sediments forms a direct sister group of Eukaryotes and exhibits an unprecedented array of genes that might have been instrumental for the ancestor of eukaryotes to develop its cellular and genomic complexity. Obviously, the molecular and biochemical investigation of both groups is timely and important, yet, it requires easy access and cultivation. We have now discovered Lokiarchaeota in local, accessible environments and were able to successfully cultivate thermophilic Thaumarchaeota. In this proposal, we will characterize the metabolic and structural traits of these two archaeal lineages and reconstruct their evolutionary ancestry in the context of the TACK+L superphylum. Beside the use of cutting edge techniques for cultivation, metagenomics and stable-isotope-based imaging, a novel method will be developed for in situ metatranscriptomic analyses of archaea. This project will give fundamental insights into the ecological success of Archaea in commonplace environments and into the biology of the closest living prokaryotic relatives of Eukaryotes. Reconstructing the ancestral gene repertoire and biological features of lineages of the TACK+L superphylum will help resolve the enigma of the emergence of eukaryotes.
Summary
The emergence of complex eukaryotic life forms on Earth from prokaryotic cells is one of the most fundamental questions in biology and also one of the least understood transitions in evolution. Phylogenomic studies recently indicated that the eukaryotic line of descent arose from within the TACK+L superphylum of Archaea. This proposal addresses the systematic analysis of two newly discovered, but largely uncharacterized lineages of Archaea from this superphylum that mark crucial evolutionary transitions. Thaumarchaeota harbor unique ‘eukaryotic’ features and represent the sole group of Archaea that has successfully radiated into virtually any moderate habitat on Earth. The Lokiarchaeota lineage found recently in deep marine sediments forms a direct sister group of Eukaryotes and exhibits an unprecedented array of genes that might have been instrumental for the ancestor of eukaryotes to develop its cellular and genomic complexity. Obviously, the molecular and biochemical investigation of both groups is timely and important, yet, it requires easy access and cultivation. We have now discovered Lokiarchaeota in local, accessible environments and were able to successfully cultivate thermophilic Thaumarchaeota. In this proposal, we will characterize the metabolic and structural traits of these two archaeal lineages and reconstruct their evolutionary ancestry in the context of the TACK+L superphylum. Beside the use of cutting edge techniques for cultivation, metagenomics and stable-isotope-based imaging, a novel method will be developed for in situ metatranscriptomic analyses of archaea. This project will give fundamental insights into the ecological success of Archaea in commonplace environments and into the biology of the closest living prokaryotic relatives of Eukaryotes. Reconstructing the ancestral gene repertoire and biological features of lineages of the TACK+L superphylum will help resolve the enigma of the emergence of eukaryotes.
Max ERC Funding
2 500 000 €
Duration
Start date: 2016-08-01, End date: 2021-07-31
Project acronym TETRAHYMENA
Project RNA directed DNA elimination in Tetrahymena
Researcher (PI) Kazufumi Mochizuki
Host Institution (HI) INSTITUT FUER MOLEKULARE BIOTECHNOLOGIE GMBH
Call Details Starting Grant (StG), LS1, ERC-2007-StG
Summary Tetrahymena eliminates micronuclear-limited sequences from the developing macronucleus during sexual reproduction (DNA elimination). It is believed that the DNA to be eliminated is identified by their homology with ~28 nt small RNAs (scnRNAs) which are made by a RNAi-related mechanism. Detailed mechanisms as well as exact biological functions of the RNA-directed DNA elimination remain obscure. The goal of this specific project is to understand: (i) How a RNAi-related mechanism directs DNA elimination. (ii) How the mating types are determined as a consequence of DNA elimination. We recently revealed that scnRNAs are processed by Dicer protein Dcl1p and form complex with Argonaute protein Twi1p. The RNA helicase Ema1p facilitates interaction of scnRNA and chromatin and this interaction induces H3K9 and K27 methylations that are catalyzed by the histone methyltransferase Ezl1p. To understand how these proteins are connected to each other, their detailed functions and novel proteins interacting with them will be analyzed. One of the possible biological functions of the DNA elimination is mating type determination. Tetrahymena possesses seven mating types and the different mating types are thought to be determined by alternative DNA elimination of a single locus. But how mating types are determined is totally unknown. We aim to identify mating type determinants using microarray screening, proteomics and mating type transformation with a cDNA library. Then, we will analyze how expression of those molecules is controlled by DNA elimination. In diverse eukaryotes, RNA silencing mechanisms mediate heterochromatin formation and regulate many chromatin functions. Through our study, we could gain deeper insights not only into DNA elimination in a curious microbe, but also into how chromatin functions are epigenetically controlled by small RNAs in most of the eukaryotes. We are confident that this work will provide a big impact on RNA silencing and chromatin studies.
Summary
Tetrahymena eliminates micronuclear-limited sequences from the developing macronucleus during sexual reproduction (DNA elimination). It is believed that the DNA to be eliminated is identified by their homology with ~28 nt small RNAs (scnRNAs) which are made by a RNAi-related mechanism. Detailed mechanisms as well as exact biological functions of the RNA-directed DNA elimination remain obscure. The goal of this specific project is to understand: (i) How a RNAi-related mechanism directs DNA elimination. (ii) How the mating types are determined as a consequence of DNA elimination. We recently revealed that scnRNAs are processed by Dicer protein Dcl1p and form complex with Argonaute protein Twi1p. The RNA helicase Ema1p facilitates interaction of scnRNA and chromatin and this interaction induces H3K9 and K27 methylations that are catalyzed by the histone methyltransferase Ezl1p. To understand how these proteins are connected to each other, their detailed functions and novel proteins interacting with them will be analyzed. One of the possible biological functions of the DNA elimination is mating type determination. Tetrahymena possesses seven mating types and the different mating types are thought to be determined by alternative DNA elimination of a single locus. But how mating types are determined is totally unknown. We aim to identify mating type determinants using microarray screening, proteomics and mating type transformation with a cDNA library. Then, we will analyze how expression of those molecules is controlled by DNA elimination. In diverse eukaryotes, RNA silencing mechanisms mediate heterochromatin formation and regulate many chromatin functions. Through our study, we could gain deeper insights not only into DNA elimination in a curious microbe, but also into how chromatin functions are epigenetically controlled by small RNAs in most of the eukaryotes. We are confident that this work will provide a big impact on RNA silencing and chromatin studies.
Max ERC Funding
900 000 €
Duration
Start date: 2008-09-01, End date: 2013-12-31
Project acronym TNT-TUMORS
Project Therapeutic approaches to enhance innate immunity against Tumors
Researcher (PI) Maria SIBILIA
Host Institution (HI) MEDIZINISCHE UNIVERSITAET WIEN
Call Details Advanced Grant (AdG), LS4, ERC-2015-AdG
Summary Recent advances using immune checkpoint inhibitors demonstrate the great potential of immunemodulation in cancer and metastasis treatment. However, the effective treatment of only a subset of patients shows that this is only the start to utilize the power of the immune system to fight cancer. An interesting approach is to harness innate immune cells, such as plasmacytoid dendritic cells (pDCs) and tumor-associated macrophages (TAM) to attack tumors and to enhance the effect of standard anti-cancer therapies. Recently, using mouse models we identified two independent mechanisms by which modulation of these two cell types restrained tumor growth. First, the direct killing of tumor cells by pDCs that occurs independent of the adaptive immune system. Second, we identified a tumor-promoting role of EGFR-expressing (EGFR+) TAMs during tumorigenesis. This enables us to look at the role of EGFR in tumorigenesis in a completely new way and we plan to exploit this novel finding to reevaluate the mechanism by which anti-EGFR drugs are effective in tumors. The mechanisms endowing pDCs with tumor-killing capacities and determining the specificity of tumor cell recognition by activated pDCs are poorly understood. Furthermore, the interaction of pDCs with macrophages has never been investigated in tumors. Here I propose to define the molecular mechanisms by which pDCs and TAMs can be instructed to adopt an anti-tumorigenic phenotype. Inducible and cell-specific genetic mouse models mimicking human cancers will allow to molecularly dissect the immunemodulatory capacity of pDCs and TAMs. State-of-the-art large scale in vitro and in vivo RNAi screens will provide a platform to identify novel molecular pathways and open the possibility for testing new strategies in cancer immunetherapy. The clinical significance of our findings will be validated in human cancer samples in close cooperation with clinicians, which ensures a fast predictive and therapeutic translation of our results.
Summary
Recent advances using immune checkpoint inhibitors demonstrate the great potential of immunemodulation in cancer and metastasis treatment. However, the effective treatment of only a subset of patients shows that this is only the start to utilize the power of the immune system to fight cancer. An interesting approach is to harness innate immune cells, such as plasmacytoid dendritic cells (pDCs) and tumor-associated macrophages (TAM) to attack tumors and to enhance the effect of standard anti-cancer therapies. Recently, using mouse models we identified two independent mechanisms by which modulation of these two cell types restrained tumor growth. First, the direct killing of tumor cells by pDCs that occurs independent of the adaptive immune system. Second, we identified a tumor-promoting role of EGFR-expressing (EGFR+) TAMs during tumorigenesis. This enables us to look at the role of EGFR in tumorigenesis in a completely new way and we plan to exploit this novel finding to reevaluate the mechanism by which anti-EGFR drugs are effective in tumors. The mechanisms endowing pDCs with tumor-killing capacities and determining the specificity of tumor cell recognition by activated pDCs are poorly understood. Furthermore, the interaction of pDCs with macrophages has never been investigated in tumors. Here I propose to define the molecular mechanisms by which pDCs and TAMs can be instructed to adopt an anti-tumorigenic phenotype. Inducible and cell-specific genetic mouse models mimicking human cancers will allow to molecularly dissect the immunemodulatory capacity of pDCs and TAMs. State-of-the-art large scale in vitro and in vivo RNAi screens will provide a platform to identify novel molecular pathways and open the possibility for testing new strategies in cancer immunetherapy. The clinical significance of our findings will be validated in human cancer samples in close cooperation with clinicians, which ensures a fast predictive and therapeutic translation of our results.
Max ERC Funding
2 499 646 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym TotipotentZygotChrom
Project Mechanisms of chromatin organization and reprogramming in totipotent mammalian zygotes
Researcher (PI) Kikue Tachibana
Host Institution (HI) INSTITUT FUER MOLEKULARE BIOTECHNOLOGIE GMBH
Call Details Consolidator Grant (CoG), LS3, ERC-2018-COG
Summary Totipotency, the developmental potential of a cell to give rise to all cell types, is naturally achieved when differentiated egg and sperm fuse to form the one-cell zygote. How chromatin is epigenetically reprogrammed to totipotency within hours after fertilisation remains a central question in biology. We aim to address this by investigating the mechanisms of reprogramming and the spatial reorganisation of chromatin in mammalian zygotes. Our interdisciplinary approach combines mechanistic cell biology with genetics and genomics to understand how chromatin reorganisation promotes totipotency and to identify key regulators of this process in zygotes. Molecular insights into reprogramming to totipotency are crucial to understand the essential zygotic stage of sexually reproducing species. A better understanding of how cells naturally reprogram chromatin to totipotency, a state upstream of pluripotency, has the potential to improve induced reprogramming technology and revolutionize regenerative medicine.
Our aim is to understand how chromatin is reprogrammed to totipotency. To reach this ambitious goal: 1) We will discover new general concepts of genome organization, as well as reprogramming-specific aspects, by capitalising on our recently developed single-nucleus Hi-C method to dissect spatial reorganisation of chromatin in zygotes. We will investigate the relationship between chromatin reorganisation and transcription. 2) We will uncover mechanisms of zygotic reprogramming by elucidating the loci and factors that support active DNA demethylation during reprogramming of the paternal genome. 3) We will illuminate the origins and contributions of the oocyte since the factors responsible for reprogramming likely reside as proteins or RNA in the unfertilized egg. Overall, these studies will provide novel insights into how chromatin is reprogrammed and spatially reorganised towards a totipotent state that facilitates zygotic genome activation.
Summary
Totipotency, the developmental potential of a cell to give rise to all cell types, is naturally achieved when differentiated egg and sperm fuse to form the one-cell zygote. How chromatin is epigenetically reprogrammed to totipotency within hours after fertilisation remains a central question in biology. We aim to address this by investigating the mechanisms of reprogramming and the spatial reorganisation of chromatin in mammalian zygotes. Our interdisciplinary approach combines mechanistic cell biology with genetics and genomics to understand how chromatin reorganisation promotes totipotency and to identify key regulators of this process in zygotes. Molecular insights into reprogramming to totipotency are crucial to understand the essential zygotic stage of sexually reproducing species. A better understanding of how cells naturally reprogram chromatin to totipotency, a state upstream of pluripotency, has the potential to improve induced reprogramming technology and revolutionize regenerative medicine.
Our aim is to understand how chromatin is reprogrammed to totipotency. To reach this ambitious goal: 1) We will discover new general concepts of genome organization, as well as reprogramming-specific aspects, by capitalising on our recently developed single-nucleus Hi-C method to dissect spatial reorganisation of chromatin in zygotes. We will investigate the relationship between chromatin reorganisation and transcription. 2) We will uncover mechanisms of zygotic reprogramming by elucidating the loci and factors that support active DNA demethylation during reprogramming of the paternal genome. 3) We will illuminate the origins and contributions of the oocyte since the factors responsible for reprogramming likely reside as proteins or RNA in the unfertilized egg. Overall, these studies will provide novel insights into how chromatin is reprogrammed and spatially reorganised towards a totipotent state that facilitates zygotic genome activation.
Max ERC Funding
2 000 000 €
Duration
Start date: 2020-02-01, End date: 2025-01-31
Project acronym TransGen RNA
Project Transgenerational regulation of glucose metabolism by noncoding RNAs
Researcher (PI) Jan-Wilhelm Kornfeld
Host Institution (HI) SYDDANSK UNIVERSITET
Call Details Starting Grant (StG), LS4, ERC-2015-STG
Summary Obesity and T2D affect large populations and cause a decline in life expectancy if untreated. The pandemic proportion of obesity and inaptitude of anti-obesity approaches reflect our limited understanding of its complex environmental and genetic etiology. Genome-wide association studies revealed that disease-associated risk variants are often situated in those 98% of the genome not encoding for proteins. This noncoding genomic space yet does not reflect ‘Junk DNA’ but gives rise to >10,000 noncoding RNAs like microRNAs and long, noncoding RNAs (lncRNAs) that implicated in control of glucose metabolism and energy homeostasis also by the applicant (Kornfeld et al. Nature 2013).
LncRNAs were paraphrased as 'Dark matter of the genome' due to their tissue-specific and dynamic expression that contrast their poorly understood role in gene regulation. In the 1st part of this proposal, we ask if lncRNAs regulate glucose metabolism and are involved in the obesity-associated dysregulation of insulin signaling in the liver, the major glucoregulatory organ in mammals. Using RNA-Seq and novel lncRNA prediction algorithms, we observed that obesity alters expression of 28 annotated and 15 hitherto unknown lncRNAs in two mouse models of obesity. To identify lncRNAs causally controlling glucose metabolism, we established a siRNA screening system that allows functional interrogation of >650 lncRNAs. These in vitro findings serve as entry for the generation of lncRNA knockout mice that are metabolically phenotyped. In the 2nd part, we hypothesize that germline ncRNAs could control the transgenerational consequences of paternal obesity as shown for lower organisms. This builds upon unpublished findings from our lab showing that obesity profoundly changes expression of germline ncRNAs. In-vitro fertilization and intergenerational breedings will trace the legacy of paternal obesity across generations and reveal ncRNAs involved in this ‘Lamarckian’ control of glucose metabolism.
Summary
Obesity and T2D affect large populations and cause a decline in life expectancy if untreated. The pandemic proportion of obesity and inaptitude of anti-obesity approaches reflect our limited understanding of its complex environmental and genetic etiology. Genome-wide association studies revealed that disease-associated risk variants are often situated in those 98% of the genome not encoding for proteins. This noncoding genomic space yet does not reflect ‘Junk DNA’ but gives rise to >10,000 noncoding RNAs like microRNAs and long, noncoding RNAs (lncRNAs) that implicated in control of glucose metabolism and energy homeostasis also by the applicant (Kornfeld et al. Nature 2013).
LncRNAs were paraphrased as 'Dark matter of the genome' due to their tissue-specific and dynamic expression that contrast their poorly understood role in gene regulation. In the 1st part of this proposal, we ask if lncRNAs regulate glucose metabolism and are involved in the obesity-associated dysregulation of insulin signaling in the liver, the major glucoregulatory organ in mammals. Using RNA-Seq and novel lncRNA prediction algorithms, we observed that obesity alters expression of 28 annotated and 15 hitherto unknown lncRNAs in two mouse models of obesity. To identify lncRNAs causally controlling glucose metabolism, we established a siRNA screening system that allows functional interrogation of >650 lncRNAs. These in vitro findings serve as entry for the generation of lncRNA knockout mice that are metabolically phenotyped. In the 2nd part, we hypothesize that germline ncRNAs could control the transgenerational consequences of paternal obesity as shown for lower organisms. This builds upon unpublished findings from our lab showing that obesity profoundly changes expression of germline ncRNAs. In-vitro fertilization and intergenerational breedings will trace the legacy of paternal obesity across generations and reveal ncRNAs involved in this ‘Lamarckian’ control of glucose metabolism.
Max ERC Funding
1 344 498 €
Duration
Start date: 2016-05-01, End date: 2021-04-30
Project acronym TROJA
Project Targeting Receptors Of Jointly Assembled Ligand-Drug Constructs
Researcher (PI) Søren Kragh Moestrup
Host Institution (HI) AARHUS UNIVERSITET
Call Details Advanced Grant (AdG), LS7, ERC-2008-AdG
Summary The TROJA proposal is an investigative bioengineering study of the exploitation of specific endocytic receptors for targeting small molecule drugs to specific cells in order to improve medical therapy. This is a new approach with scientific roots in the basic research on endocytic receptors and protein expression carried out in the laboratory of the applicant. The major line of the proposal concerns the construction of combinatory drugs for targeting the haptoglobin (Hp)-hemoglobin receptor CD163 (Kristiansen et al., Nature 409:198-201) expressed in the monocyte-macrophage system. The platform may apply to a broad spectrum of diseases such as inflammatory diseases, various infections and certain cancers which all have CD163-expressing macrophages or malignant derivatives as key cell type in the pathogenesis of the disease. Dependent of the above-mentioned diseases to be treated, the drugs are intended to have anti-inflammatory, microbiotic or cytostatic effects. Efficacy of the combinatory drug will be investigated in monocytes/macrophages, CD163-transfected cells and as well as in suitable animal models including transgenic animals. Another and minor line of the proposal concerns the construction of combinatory drugs for targeting a very recently discovered Hp-Hb receptor expressed in trypanosomes (Vanhollebeke et al., Science, in press) causing sleeping sickness. Both lines of this research proposal will take advantage of established recombinant protein expression methods and chemical coupling technology to construct jointly assembled ligand-drugs complexes. In terms of drug efficacy and toxicity, the aim is to design combinatory products that remain largely inactive in their receptor-binding form, but upon release in the cells or parasites the active small molecule components become active. The discovery of such a Trojan horse platform for cellular drug entry may have major implications for future drug development and for new applications of existent drugs.
Summary
The TROJA proposal is an investigative bioengineering study of the exploitation of specific endocytic receptors for targeting small molecule drugs to specific cells in order to improve medical therapy. This is a new approach with scientific roots in the basic research on endocytic receptors and protein expression carried out in the laboratory of the applicant. The major line of the proposal concerns the construction of combinatory drugs for targeting the haptoglobin (Hp)-hemoglobin receptor CD163 (Kristiansen et al., Nature 409:198-201) expressed in the monocyte-macrophage system. The platform may apply to a broad spectrum of diseases such as inflammatory diseases, various infections and certain cancers which all have CD163-expressing macrophages or malignant derivatives as key cell type in the pathogenesis of the disease. Dependent of the above-mentioned diseases to be treated, the drugs are intended to have anti-inflammatory, microbiotic or cytostatic effects. Efficacy of the combinatory drug will be investigated in monocytes/macrophages, CD163-transfected cells and as well as in suitable animal models including transgenic animals. Another and minor line of the proposal concerns the construction of combinatory drugs for targeting a very recently discovered Hp-Hb receptor expressed in trypanosomes (Vanhollebeke et al., Science, in press) causing sleeping sickness. Both lines of this research proposal will take advantage of established recombinant protein expression methods and chemical coupling technology to construct jointly assembled ligand-drugs complexes. In terms of drug efficacy and toxicity, the aim is to design combinatory products that remain largely inactive in their receptor-binding form, but upon release in the cells or parasites the active small molecule components become active. The discovery of such a Trojan horse platform for cellular drug entry may have major implications for future drug development and for new applications of existent drugs.
Max ERC Funding
2 400 000 €
Duration
Start date: 2009-04-01, End date: 2014-03-31
Project acronym Troy Stem Cells
Project Troy+ stomach stem cells in homeostasis, repair and pathogenesis
Researcher (PI) Bon-Kyoung Koo
Host Institution (HI) INSTITUT FUER MOLEKULARE BIOTECHNOLOGIE GMBH
Call Details Starting Grant (StG), LS4, ERC-2014-STG
Summary The adult mammalian stomach can be divided into three distinct parts: From the proximal fore-stomach over the corpus to the distal pylorus. Due to constant exposure to mechanical stress and to hostile contents of the lumen, highly specialized cell types have to be constantly reproduced in order to maintain the function of the gastrointestinal tract. Recently, the applicant identified Troy+ chief cells as a novel stem cell population in the corpus epithelium. Troy+ chief cells displayed a very low proliferation rate indicating their quiescent nature compared to other known gastro-intestinal tract stem cells. Interestingly, these stem cells can actively divide upon tissue damage, suggesting distinctive statuses under conditions of homeostasis and injury.
As Troy+ stomach stem cells exhibit interconvertible characteristics i.e. quiescent and proliferative, they represent a unique model of adult stem cells with which we can study 1) the dynamics of stem cell propagation in homeostasis and regeneration and the underlying mechanism of this switch by analysing molecular and epigenetic profiles. Subsequently, by analysing mRNA expression profiles and epigenetic changes in Troy+ stem cells between homeostasis and injury repair, we will generate a list of genes with potentially interesting functions in cell fate decisions. We will therefore investigate 2) the stomach stem cell programme in homeostasis and regeneration using in vitro and in vivo functional genetics. Lastly, we will characterise 3) human stomach stem cells in normal and pathological conditions.
Here we pursue three main aims:
- Investigating Troy+ stem cell dynamics during homeostasis and injury repair
- Unmasking the stomach stem cell programme using in vitro and in vivo functional genetics
- Characterising human stomach stem cells
Summary
The adult mammalian stomach can be divided into three distinct parts: From the proximal fore-stomach over the corpus to the distal pylorus. Due to constant exposure to mechanical stress and to hostile contents of the lumen, highly specialized cell types have to be constantly reproduced in order to maintain the function of the gastrointestinal tract. Recently, the applicant identified Troy+ chief cells as a novel stem cell population in the corpus epithelium. Troy+ chief cells displayed a very low proliferation rate indicating their quiescent nature compared to other known gastro-intestinal tract stem cells. Interestingly, these stem cells can actively divide upon tissue damage, suggesting distinctive statuses under conditions of homeostasis and injury.
As Troy+ stomach stem cells exhibit interconvertible characteristics i.e. quiescent and proliferative, they represent a unique model of adult stem cells with which we can study 1) the dynamics of stem cell propagation in homeostasis and regeneration and the underlying mechanism of this switch by analysing molecular and epigenetic profiles. Subsequently, by analysing mRNA expression profiles and epigenetic changes in Troy+ stem cells between homeostasis and injury repair, we will generate a list of genes with potentially interesting functions in cell fate decisions. We will therefore investigate 2) the stomach stem cell programme in homeostasis and regeneration using in vitro and in vivo functional genetics. Lastly, we will characterise 3) human stomach stem cells in normal and pathological conditions.
Here we pursue three main aims:
- Investigating Troy+ stem cell dynamics during homeostasis and injury repair
- Unmasking the stomach stem cell programme using in vitro and in vivo functional genetics
- Characterising human stomach stem cells
Max ERC Funding
1 570 399 €
Duration
Start date: 2015-08-01, End date: 2020-07-31
Project acronym UNICODE
Project Evolution and Impact of Heterochromatin on a Young Drosophila Y chromosome
Researcher (PI) Qi Zhou
Host Institution (HI) UNIVERSITAT WIEN
Call Details Starting Grant (StG), LS2, ERC-2015-STG
Summary The transition from euchromatin to heterochromatin is a fundamental process that particularly reshaped the epigenomic landscape of Y chromosome. Its definitive genomic underpinning and broad functional impact are still unclear, as heterochromatin (e.g., that of human Y) is usually too repetitive to study. I have previously demonstrated that, the young Y (‘neo-Y’) chromosome of Drosophila miranda has just initiated such a transition, thus is a powerful model to unveil the evolution, regulation and functional interaction of heterochromatin. I showed that this neo-Y still harbours over 1800 genes, and only 20-50% of the sequences are transposable elements (TE). Over five years, I aim to: 1) precisely resolve the structure and insertion sites of TEs as a pre-requisite for studying heterochromatin, by combining state-of-art sequencing and bioinformatic techniques. 2) I will reveal the de novo heterochromatin formation triggered by TE insertions or the heterochromatin/euchromatin boundary shifts on the neo-Y, by comparing the binding profiles of histone modification hallmarks and insulator proteins of D. miranda to its sibling species D. pseudoobscura, which lacks the neo-Y. Such epigenomic changes have likely driven the exaptation or innovation of small RNA pathways that govern the TE mobility. 3) I will then identify the responsible small RNAs and their encoding loci, which are expected to have newly emerged or differentially expressed in D. miranda relative to D. pseudoobscura. 4) Finally, I will develop CRISPR/Cas9 in D. miranda to manipulate the expression of TEs encoding such small RNAs on the neo-Y, in order to scrutinize how TE/heterochromatin evolution on the Y would impact the chromatin landscape of the entire host genome. The combined aim of this multidisciplinary project is to generate a framework for understanding the basic mechanisms of how heterochromatin evolves; and open a new avenue toward the discovery of Y chromosome function beyond male determination.
Summary
The transition from euchromatin to heterochromatin is a fundamental process that particularly reshaped the epigenomic landscape of Y chromosome. Its definitive genomic underpinning and broad functional impact are still unclear, as heterochromatin (e.g., that of human Y) is usually too repetitive to study. I have previously demonstrated that, the young Y (‘neo-Y’) chromosome of Drosophila miranda has just initiated such a transition, thus is a powerful model to unveil the evolution, regulation and functional interaction of heterochromatin. I showed that this neo-Y still harbours over 1800 genes, and only 20-50% of the sequences are transposable elements (TE). Over five years, I aim to: 1) precisely resolve the structure and insertion sites of TEs as a pre-requisite for studying heterochromatin, by combining state-of-art sequencing and bioinformatic techniques. 2) I will reveal the de novo heterochromatin formation triggered by TE insertions or the heterochromatin/euchromatin boundary shifts on the neo-Y, by comparing the binding profiles of histone modification hallmarks and insulator proteins of D. miranda to its sibling species D. pseudoobscura, which lacks the neo-Y. Such epigenomic changes have likely driven the exaptation or innovation of small RNA pathways that govern the TE mobility. 3) I will then identify the responsible small RNAs and their encoding loci, which are expected to have newly emerged or differentially expressed in D. miranda relative to D. pseudoobscura. 4) Finally, I will develop CRISPR/Cas9 in D. miranda to manipulate the expression of TEs encoding such small RNAs on the neo-Y, in order to scrutinize how TE/heterochromatin evolution on the Y would impact the chromatin landscape of the entire host genome. The combined aim of this multidisciplinary project is to generate a framework for understanding the basic mechanisms of how heterochromatin evolves; and open a new avenue toward the discovery of Y chromosome function beyond male determination.
Max ERC Funding
1 971 846 €
Duration
Start date: 2016-08-01, End date: 2021-07-31
Project acronym uPET
Project Molecular imaging and targeted therapy of the aggressive phenotype: development of uPAR theranostics for cancer patients
Researcher (PI) Andreas Kjaer
Host Institution (HI) REGION HOVEDSTADEN
Call Details Advanced Grant (AdG), LS7, ERC-2014-ADG
Summary For practice of personalized medicine in cancer, non-invasive tools for diagnosing at the molecular level are needed. Molecular imaging methods are capable of this while at the same time circumventing sampling error as the whole tumor burden is evaluated.
We recently developed and performed the first-ever clinical PET scan of uPAR, a proteolytic system known to be strongly associated with metastatic potential in most cancer forms. We believe this new concept of uPAR-PET is a major breakthrough and has the potential to become one of the most used PET tracers as it fulfils unmet needs in prostate and breast cancer. Based on this, together with additional proof-of-concept data we obtained on targeting uPAR for optical imaging and radionuclide therapy, we now plan to develop and take into patients these new technologies for improved outcome.
Specific aims are to develop and translate into human use:
1. A PET uPAR imaging ligand platform for visualization of the aggressive phenotype and risk-stratification to be used in tailoring therapy, e.g. in prostate cancer to decide whether prostatectomy is necessary.
2. uPAR-PET combined with simultaneous 13C-hyperpolarized pyruvate MRSI (Warburg effect). This will increase prognostic power, refine tumor phenotyping and thereby allow for better tailoring of therapy and early prediction of treatment response.
3. A uPAR optical imaging technology for guiding removal of cancer tissue during surgery. This will help delineate cancer tissue for removal while leaving healthy tissue behind. We expect better outcome with removal of less tissue.
4. Selective radionuclide therapies targeting uPAR positive, invasive cancer cells using β or α emitters. Dose planning will be performed using uPAR-PET imaging. This image-therapy pairing is also known as theranostics.
Our endeavour is ambitious, yet realistic considering our competencies and track-record. We expect cancer patients to benefit from our new methods within the 5 year time-frame.
Summary
For practice of personalized medicine in cancer, non-invasive tools for diagnosing at the molecular level are needed. Molecular imaging methods are capable of this while at the same time circumventing sampling error as the whole tumor burden is evaluated.
We recently developed and performed the first-ever clinical PET scan of uPAR, a proteolytic system known to be strongly associated with metastatic potential in most cancer forms. We believe this new concept of uPAR-PET is a major breakthrough and has the potential to become one of the most used PET tracers as it fulfils unmet needs in prostate and breast cancer. Based on this, together with additional proof-of-concept data we obtained on targeting uPAR for optical imaging and radionuclide therapy, we now plan to develop and take into patients these new technologies for improved outcome.
Specific aims are to develop and translate into human use:
1. A PET uPAR imaging ligand platform for visualization of the aggressive phenotype and risk-stratification to be used in tailoring therapy, e.g. in prostate cancer to decide whether prostatectomy is necessary.
2. uPAR-PET combined with simultaneous 13C-hyperpolarized pyruvate MRSI (Warburg effect). This will increase prognostic power, refine tumor phenotyping and thereby allow for better tailoring of therapy and early prediction of treatment response.
3. A uPAR optical imaging technology for guiding removal of cancer tissue during surgery. This will help delineate cancer tissue for removal while leaving healthy tissue behind. We expect better outcome with removal of less tissue.
4. Selective radionuclide therapies targeting uPAR positive, invasive cancer cells using β or α emitters. Dose planning will be performed using uPAR-PET imaging. This image-therapy pairing is also known as theranostics.
Our endeavour is ambitious, yet realistic considering our competencies and track-record. We expect cancer patients to benefit from our new methods within the 5 year time-frame.
Max ERC Funding
2 072 000 €
Duration
Start date: 2015-10-01, End date: 2020-09-30
Project acronym VIREX
Project Mumps VIRus EXploitation of the human adhesion receptor GPR125
Researcher (PI) Mette Marie Rosenkilde
Host Institution (HI) KOBENHAVNS UNIVERSITET
Call Details Consolidator Grant (CoG), LS7, ERC-2015-CoG
Summary Mumps virus is a re-emerging pathogen that causes painful inflammatory symptoms, such as parotitis (salivary gland infection) and orchitis (testis infection). It is highly neurotropic with evidence of brain infection in half of cases and clinical evidence in up to 10%. It is a small RNA virus belonging to the family of paramyxoviridae that includes e.g. viruses for measles and pneumonia, all having a huge impact on global economics and human health. Current vaccine programs have not managed to eliminate mumps and infections occur also in vaccinated individuals.
Seven transmembrane (7TM) receptors are important drug targets. Large DNA viruses (herpes- and pox-) assign large parts of their genomes to exploit 7TM receptors. No such mechanism has however yet been described for small viruses.
Based on strong preliminary data, I will in this interdisciplinary project test the groundbreaking hypothesis that the adhesion 7TM receptor GPR125 is central for the organ damage caused by mumps virus via an interaction with the mumps virus-encoded short-hydrophobic (SH)-protein. I will do so by determining:
1 - The functional consequences of GPR125-SH-interaction at a single cell, organ and whole body level within the context of mumps virus infection
2 - The structural requirements for the GPR125-mumps virus interaction using NMR and resolution of crystal structure in preparation for future drug design
The project is high risk and high gain, yet the gain clearly exceeds the risk. On account of my past expertise in pharmacology and virology, and that of several expert collaborators, the project is indeed feasible. It has tremendous perspectives as SH-proteins are present also in other viruses. The SH-GPR125 complex might thus represent a general principle for organ damage and a mode of action more generally amenable to therapeutic interference. In fact, novel approaches, mechanism-based, might be seen as more appealing to those who fear current vaccination 'modes'.
Summary
Mumps virus is a re-emerging pathogen that causes painful inflammatory symptoms, such as parotitis (salivary gland infection) and orchitis (testis infection). It is highly neurotropic with evidence of brain infection in half of cases and clinical evidence in up to 10%. It is a small RNA virus belonging to the family of paramyxoviridae that includes e.g. viruses for measles and pneumonia, all having a huge impact on global economics and human health. Current vaccine programs have not managed to eliminate mumps and infections occur also in vaccinated individuals.
Seven transmembrane (7TM) receptors are important drug targets. Large DNA viruses (herpes- and pox-) assign large parts of their genomes to exploit 7TM receptors. No such mechanism has however yet been described for small viruses.
Based on strong preliminary data, I will in this interdisciplinary project test the groundbreaking hypothesis that the adhesion 7TM receptor GPR125 is central for the organ damage caused by mumps virus via an interaction with the mumps virus-encoded short-hydrophobic (SH)-protein. I will do so by determining:
1 - The functional consequences of GPR125-SH-interaction at a single cell, organ and whole body level within the context of mumps virus infection
2 - The structural requirements for the GPR125-mumps virus interaction using NMR and resolution of crystal structure in preparation for future drug design
The project is high risk and high gain, yet the gain clearly exceeds the risk. On account of my past expertise in pharmacology and virology, and that of several expert collaborators, the project is indeed feasible. It has tremendous perspectives as SH-proteins are present also in other viruses. The SH-GPR125 complex might thus represent a general principle for organ damage and a mode of action more generally amenable to therapeutic interference. In fact, novel approaches, mechanism-based, might be seen as more appealing to those who fear current vaccination 'modes'.
Max ERC Funding
1 813 367 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym VIRUSES AND RNA
Project RNA regulation during viral infection
Researcher (PI) Troels Kasper Høyer Scheel
Host Institution (HI) KOBENHAVNS UNIVERSITET
Call Details Starting Grant (StG), LS6, ERC-2018-STG
Summary Viral infections are responsible for significant morbidity and mortality and frequency and impact of epidemics are expected to increase. Thorough understanding of basic virology is critical for informed development of prevention and control. Most systematic studies of virus-host interactions have focused on proteins, however, with recent methodological advances the intersecting fields of viral infection and RNA biology hold great promise for basic and therapeutic exploration. The goal of this application therefore is to discover and dissect RNA-based virus-host interactions and related regulatory mechanisms of gene expression.
Micro-RNAs (miRNAs) fine-tune gene expression by repressing mRNA targets. However, cellular miRNAs increase translation and replication of certain viruses. Thus, hepatitis C virus (HCV) critically depends on the liver specific miR-122, which emerged as a therapeutic target. Further, HCV sequesters enough miR-122 to indirectly regulate cellular gene expression. I hypothesize that this RNA-based mechanism contributes to virus induced liver cancer, and aim to address this using our recently developed rodent model for HCV infection (Aim 1). Better understanding of viral RNA (vRNA) interactions could significantly contribute to basic infection biology and novel therapeutics. I therefore aim to systematically identify vRNA interactions with other cellular RNAs and proteins (Aim 2). I expect to identify interactions of value for functional regulation and therapeutic targeting. I finally hypothesize that translation of certain cellular mRNAs – similarly to viruses – increase upon miRNA binding, and aim to systematically screen for such virus-like alternative regulation, with potential to change understanding of post-transcriptional regulation (Aim 3).
In conclusion, this high-risk high-gain project has potential to shape novel dogmas for virus and RNA biology and to identify novel RNA-based therapeutic targets; a promising upcoming field of discovery.
Summary
Viral infections are responsible for significant morbidity and mortality and frequency and impact of epidemics are expected to increase. Thorough understanding of basic virology is critical for informed development of prevention and control. Most systematic studies of virus-host interactions have focused on proteins, however, with recent methodological advances the intersecting fields of viral infection and RNA biology hold great promise for basic and therapeutic exploration. The goal of this application therefore is to discover and dissect RNA-based virus-host interactions and related regulatory mechanisms of gene expression.
Micro-RNAs (miRNAs) fine-tune gene expression by repressing mRNA targets. However, cellular miRNAs increase translation and replication of certain viruses. Thus, hepatitis C virus (HCV) critically depends on the liver specific miR-122, which emerged as a therapeutic target. Further, HCV sequesters enough miR-122 to indirectly regulate cellular gene expression. I hypothesize that this RNA-based mechanism contributes to virus induced liver cancer, and aim to address this using our recently developed rodent model for HCV infection (Aim 1). Better understanding of viral RNA (vRNA) interactions could significantly contribute to basic infection biology and novel therapeutics. I therefore aim to systematically identify vRNA interactions with other cellular RNAs and proteins (Aim 2). I expect to identify interactions of value for functional regulation and therapeutic targeting. I finally hypothesize that translation of certain cellular mRNAs – similarly to viruses – increase upon miRNA binding, and aim to systematically screen for such virus-like alternative regulation, with potential to change understanding of post-transcriptional regulation (Aim 3).
In conclusion, this high-risk high-gain project has potential to shape novel dogmas for virus and RNA biology and to identify novel RNA-based therapeutic targets; a promising upcoming field of discovery.
Max ERC Funding
1 500 000 €
Duration
Start date: 2019-07-01, End date: 2024-06-30
Project acronym YEAST-TRANS
Project Deciphering the transport mechanisms of small xenobiotic molecules in synthetic yeast cell factories
Researcher (PI) Irina BORODINA
Host Institution (HI) DANMARKS TEKNISKE UNIVERSITET
Call Details Starting Grant (StG), LS9, ERC-2017-STG
Summary Industrial biotechnology employs synthetic cell factories to create bulk and fine chemicals and fuels from renewable resources, laying the basis for the future bio-based economy. The major part of the wanted bio-based chemicals are not native to the host cell, such as yeast, i.e. they are xenobiotic. Some xenobiotic compounds are readily secreted by synthetic cells, some are poorly secreted and some are not secreted at all, but how does this transport occur? Or why does it not occur? These fundamental questions remain to be answered and this will have great implications on industrial biotechnology, because improved secretion would bring down the production costs and enable the emergence of novel bio-based products.
YEAST-TRANS will fill in this knowledge gap by carrying out the first systematic genome-scale transporter study to uncover the transport mechanisms of small xenobiotic molecules by synthetic yeast cells and to apply this knowledge for engineering more efficient cell factories for bio-based production of fuels and chemicals.
Summary
Industrial biotechnology employs synthetic cell factories to create bulk and fine chemicals and fuels from renewable resources, laying the basis for the future bio-based economy. The major part of the wanted bio-based chemicals are not native to the host cell, such as yeast, i.e. they are xenobiotic. Some xenobiotic compounds are readily secreted by synthetic cells, some are poorly secreted and some are not secreted at all, but how does this transport occur? Or why does it not occur? These fundamental questions remain to be answered and this will have great implications on industrial biotechnology, because improved secretion would bring down the production costs and enable the emergence of novel bio-based products.
YEAST-TRANS will fill in this knowledge gap by carrying out the first systematic genome-scale transporter study to uncover the transport mechanisms of small xenobiotic molecules by synthetic yeast cells and to apply this knowledge for engineering more efficient cell factories for bio-based production of fuels and chemicals.
Max ERC Funding
1 423 358 €
Duration
Start date: 2017-12-01, End date: 2022-11-30