Project acronym 3D_Tryps
Project The role of three-dimensional genome architecture in antigenic variation
Researcher (PI) Tim Nicolai SIEGEL
Host Institution (HI) LUDWIG-MAXIMILIANS-UNIVERSITAET MUENCHEN
Call Details Starting Grant (StG), LS6, ERC-2016-STG
Summary Antigenic variation is a widely employed strategy to evade the host immune response. It has similar functional requirements even in evolutionarily divergent pathogens. These include the mutually exclusive expression of antigens and the periodic, nonrandom switching in the expression of different antigens during the course of an infection. Despite decades of research the mechanisms of antigenic variation are not fully understood in any organism.
The recent development of high-throughput sequencing-based assays to probe the 3D genome architecture (Hi-C) has revealed the importance of the spatial organization of DNA inside the nucleus. 3D genome architecture plays a critical role in the regulation of mutually exclusive gene expression and the frequency of translocation between different genomic loci in many eukaryotes. Thus, genome architecture may also be a key regulator of antigenic variation, yet the causal links between genome architecture and the expression of antigens have not been studied systematically. In addition, the development of CRISPR-Cas9-based approaches to perform nucleotide-specific genome editing has opened unprecedented opportunities to study the influence of DNA sequence elements on the spatial organization of DNA and how this impacts antigen expression.
I have adapted both Hi-C and CRISPR-Cas9 technology to the protozoan parasite Trypanosoma brucei, one of the most important model organisms to study antigenic variation. These techniques will enable me to bridge the field of antigenic variation research with that of genome architecture. I will perform the first systematic analysis of the role of genome architecture in the mutually exclusive and hierarchical expression of antigens in any pathogen.
The experiments outlined in this proposal will provide new insight, facilitating a new view of antigenic variation and may eventually help medical intervention in T. brucei and in other pathogens relying on antigenic variation for their survival.
Summary
Antigenic variation is a widely employed strategy to evade the host immune response. It has similar functional requirements even in evolutionarily divergent pathogens. These include the mutually exclusive expression of antigens and the periodic, nonrandom switching in the expression of different antigens during the course of an infection. Despite decades of research the mechanisms of antigenic variation are not fully understood in any organism.
The recent development of high-throughput sequencing-based assays to probe the 3D genome architecture (Hi-C) has revealed the importance of the spatial organization of DNA inside the nucleus. 3D genome architecture plays a critical role in the regulation of mutually exclusive gene expression and the frequency of translocation between different genomic loci in many eukaryotes. Thus, genome architecture may also be a key regulator of antigenic variation, yet the causal links between genome architecture and the expression of antigens have not been studied systematically. In addition, the development of CRISPR-Cas9-based approaches to perform nucleotide-specific genome editing has opened unprecedented opportunities to study the influence of DNA sequence elements on the spatial organization of DNA and how this impacts antigen expression.
I have adapted both Hi-C and CRISPR-Cas9 technology to the protozoan parasite Trypanosoma brucei, one of the most important model organisms to study antigenic variation. These techniques will enable me to bridge the field of antigenic variation research with that of genome architecture. I will perform the first systematic analysis of the role of genome architecture in the mutually exclusive and hierarchical expression of antigens in any pathogen.
The experiments outlined in this proposal will provide new insight, facilitating a new view of antigenic variation and may eventually help medical intervention in T. brucei and in other pathogens relying on antigenic variation for their survival.
Max ERC Funding
1 498 175 €
Duration
Start date: 2017-04-01, End date: 2022-03-31
Project acronym AIM2 INFLAMMASOME
Project Cytosolic recognition of foreign nucleic acids: Molecular and functional characterization of AIM2, a central player in DNA-triggered inflammasome activation
Researcher (PI) Veit Hornung
Host Institution (HI) UNIVERSITAETSKLINIKUM BONN
Call Details Starting Grant (StG), LS6, ERC-2009-StG
Summary Host cytokines, chemokines and type I IFNs are critical effectors of the innate immune response to viral and bacterial pathogens. Several classes of germ-line encoded pattern recognition receptors have been identified, which sense non-self nucleic acids and trigger these responses. Recently NLRP-3, a member of the NOD-like receptor (NLR) family, has been shown to sense endogenous danger signals, environmental insults and the DNA viruses adenovirus and HSV. Activation of NLRP-3 induces the formation of a large multiprotein complex in cells termed inflammasome , which controls the activity of pro-caspase-1 and the maturation of pro-IL-1² and pro-IL18 into their active forms. NLRP-3, however, does not regulate these responses to double stranded cytosolic DNA. We identified the cytosolic protein AIM2 as the missing receptor for cytosolic DNA. AIM2 contains a HIN200 domain, which binds to DNA and a pyrin domain, which associates with the adapter molecule ASC to activate both NF-ºB and caspase-1. Knock down of AIM2 down-regulates caspase-1-mediated IL-1² responses following DNA stimulation or vaccinia virus infection. Collectively, these observations demonstrate that AIM2 forms an inflammasome with the DNA ligand and ASC to activate caspase-1. Our underlying hypothesis for this proposal is that AIM2 plays a central role in host-defence to cytosolic microbial pathogens and also in DNA-triggered autoimmunity. The goals of this research proposal are to further characterize the DNA ligand for AIM2, to explore the molecular mechanisms of AIM2 activation, to define the contribution of AIM2 to host-defence against viral and bacterial pathogens and to assess its function in nucleic acid triggered autoimmune disease. The characterization of AIM2 and its role in innate immunity could open new avenues in the advancement of immunotherapy and treatment of autoimmune disease.
Summary
Host cytokines, chemokines and type I IFNs are critical effectors of the innate immune response to viral and bacterial pathogens. Several classes of germ-line encoded pattern recognition receptors have been identified, which sense non-self nucleic acids and trigger these responses. Recently NLRP-3, a member of the NOD-like receptor (NLR) family, has been shown to sense endogenous danger signals, environmental insults and the DNA viruses adenovirus and HSV. Activation of NLRP-3 induces the formation of a large multiprotein complex in cells termed inflammasome , which controls the activity of pro-caspase-1 and the maturation of pro-IL-1² and pro-IL18 into their active forms. NLRP-3, however, does not regulate these responses to double stranded cytosolic DNA. We identified the cytosolic protein AIM2 as the missing receptor for cytosolic DNA. AIM2 contains a HIN200 domain, which binds to DNA and a pyrin domain, which associates with the adapter molecule ASC to activate both NF-ºB and caspase-1. Knock down of AIM2 down-regulates caspase-1-mediated IL-1² responses following DNA stimulation or vaccinia virus infection. Collectively, these observations demonstrate that AIM2 forms an inflammasome with the DNA ligand and ASC to activate caspase-1. Our underlying hypothesis for this proposal is that AIM2 plays a central role in host-defence to cytosolic microbial pathogens and also in DNA-triggered autoimmunity. The goals of this research proposal are to further characterize the DNA ligand for AIM2, to explore the molecular mechanisms of AIM2 activation, to define the contribution of AIM2 to host-defence against viral and bacterial pathogens and to assess its function in nucleic acid triggered autoimmune disease. The characterization of AIM2 and its role in innate immunity could open new avenues in the advancement of immunotherapy and treatment of autoimmune disease.
Max ERC Funding
1 727 920 €
Duration
Start date: 2009-12-01, End date: 2014-11-30
Project acronym ALLERGUT
Project Mucosal Tolerance and Allergic Predisposition: Does it all start in the gut?
Researcher (PI) Caspar OHNMACHT
Host Institution (HI) HELMHOLTZ ZENTRUM MUENCHEN DEUTSCHES FORSCHUNGSZENTRUM FUER GESUNDHEIT UND UMWELT GMBH
Call Details Starting Grant (StG), LS6, ERC-2016-STG
Summary Currently, more than 30% of all Europeans suffer from one or more allergic disorder but treatment is still mostly symptomatic due to a lack of understanding the underlying causality. Allergies are caused by type 2 immune responses triggered by recognition of harmless antigens. Both genetic and environmental factors have been proposed to favour allergic predisposition and both factors have a huge impact on the symbiotic microbiota and the intestinal immune system. Recently we and others showed that the transcription factor ROR(γt) seems to play a key role in mucosal tolerance in the gut and also regulates intestinal type 2 immune responses.
Based on these results I postulate two major events in the gut for the development of an allergy in the lifetime of an individual: First, a failure to establish mucosal tolerance or anergy constitutes a necessity for the outbreak of allergic symptoms and allergic disease. Second, a certain ‘core’ microbiome or pathway of the intestinal microbiota predispose certain individuals for the later development of allergic disorders. Therefore, I will address the following aims:
1) Influence of ROR(γt) on mucosal tolerance induction and allergic disorders
2) Elucidate the T cell receptor repertoire of intestinal Th2 and ROR(γt)+ Tregs and assess the role of alternative NFκB pathway for induction of mucosal tolerance
3) Identification of ‘core’ microbiome signatures or metabolic pathways that favour allergic predisposition
ALLERGUT will provide ground-breaking knowledge on molecular mechanisms of the failure of mucosal tolerance in the gut and will prove if the resident ROR(γt)+ T(reg) cells can function as a mechanistic starting point for molecular intervention strategies on the background of the hygiene hypothesis. The vision of ALLERGUT is to diagnose mucosal disbalance, prevent and treat allergic disorders even before outbreak and thereby promote Public Health initiative for better living.
Summary
Currently, more than 30% of all Europeans suffer from one or more allergic disorder but treatment is still mostly symptomatic due to a lack of understanding the underlying causality. Allergies are caused by type 2 immune responses triggered by recognition of harmless antigens. Both genetic and environmental factors have been proposed to favour allergic predisposition and both factors have a huge impact on the symbiotic microbiota and the intestinal immune system. Recently we and others showed that the transcription factor ROR(γt) seems to play a key role in mucosal tolerance in the gut and also regulates intestinal type 2 immune responses.
Based on these results I postulate two major events in the gut for the development of an allergy in the lifetime of an individual: First, a failure to establish mucosal tolerance or anergy constitutes a necessity for the outbreak of allergic symptoms and allergic disease. Second, a certain ‘core’ microbiome or pathway of the intestinal microbiota predispose certain individuals for the later development of allergic disorders. Therefore, I will address the following aims:
1) Influence of ROR(γt) on mucosal tolerance induction and allergic disorders
2) Elucidate the T cell receptor repertoire of intestinal Th2 and ROR(γt)+ Tregs and assess the role of alternative NFκB pathway for induction of mucosal tolerance
3) Identification of ‘core’ microbiome signatures or metabolic pathways that favour allergic predisposition
ALLERGUT will provide ground-breaking knowledge on molecular mechanisms of the failure of mucosal tolerance in the gut and will prove if the resident ROR(γt)+ T(reg) cells can function as a mechanistic starting point for molecular intervention strategies on the background of the hygiene hypothesis. The vision of ALLERGUT is to diagnose mucosal disbalance, prevent and treat allergic disorders even before outbreak and thereby promote Public Health initiative for better living.
Max ERC Funding
1 498 175 €
Duration
Start date: 2017-07-01, End date: 2022-06-30
Project acronym ARCHAELLUM
Project Assembly and function of the crenarchaeal flagellum
Researcher (PI) Sonja-Verena Albers
Host Institution (HI) ALBERT-LUDWIGS-UNIVERSITAET FREIBURG
Call Details Starting Grant (StG), LS6, ERC-2012-StG_20111109
Summary "Archaea constitute the third domain of life and are believed to be close to the origin of life. They comprise a diverse group of micro-organisms that combine bacterial and eukaryotic features, but also employ many novel mechanisms. They possess a unique cell envelope with a cytoplasmic membrane of ether lipids surrounded by a proteinaceous S-layer and various cell appendages such as flagella, pili and more unusual structures. Studies have shown that the archaeal flagellum is an unique structure as it functionally resembles the bacterial flagellum, but structurally it is a simple type IV pilus. Moreover, we have shown that this type IV pilus can rotate. Therefore I propose to name the archaeal flagellum, the archaellum, as it is fundamentally different from the bacterial flagellum.
In this proposal I aim to understand the assembly and mechanism of rotation of the archaellum of the thermocacidophilic crenarchaen Sulfolobus acidocaldarius by using biochemical, genetic and biophysical methods. The main milestons are:
- Biochemical and structural characterization of all archaellum subunits
- To understand the assembly pathway of the archaellum and the interactions of its different
subunits
- To understand how rotation of the filament is achieved and which subunits are important
for this movement
This work will identify a new, relatively simple motor complex that has evolved from primordial type IV pili assembly machineries and therefore uncover general principles of macromolecular assemblies at cellular surfaces and a novel mechanism to generate mechanical force that can be translated into movement."
Summary
"Archaea constitute the third domain of life and are believed to be close to the origin of life. They comprise a diverse group of micro-organisms that combine bacterial and eukaryotic features, but also employ many novel mechanisms. They possess a unique cell envelope with a cytoplasmic membrane of ether lipids surrounded by a proteinaceous S-layer and various cell appendages such as flagella, pili and more unusual structures. Studies have shown that the archaeal flagellum is an unique structure as it functionally resembles the bacterial flagellum, but structurally it is a simple type IV pilus. Moreover, we have shown that this type IV pilus can rotate. Therefore I propose to name the archaeal flagellum, the archaellum, as it is fundamentally different from the bacterial flagellum.
In this proposal I aim to understand the assembly and mechanism of rotation of the archaellum of the thermocacidophilic crenarchaen Sulfolobus acidocaldarius by using biochemical, genetic and biophysical methods. The main milestons are:
- Biochemical and structural characterization of all archaellum subunits
- To understand the assembly pathway of the archaellum and the interactions of its different
subunits
- To understand how rotation of the filament is achieved and which subunits are important
for this movement
This work will identify a new, relatively simple motor complex that has evolved from primordial type IV pili assembly machineries and therefore uncover general principles of macromolecular assemblies at cellular surfaces and a novel mechanism to generate mechanical force that can be translated into movement."
Max ERC Funding
1 464 317 €
Duration
Start date: 2013-02-01, End date: 2018-01-31
Project acronym Baby DCs
Project Age-dependent Regulation of Dendritic Cell Development and Function
Researcher (PI) Barbara Ursula SCHRAML
Host Institution (HI) LUDWIG-MAXIMILIANS-UNIVERSITAET MUENCHEN
Call Details Starting Grant (StG), LS6, ERC-2016-STG
Summary Early life immune balance is essential for survival and establishment of healthy immunity in later life. We aim to define how age-dependent regulation of dendritic cell (DC) development contributes to this crucial immune balance. DCs are versatile controllers of immunity that in neonates are qualitatively distinct from adults. Why such age-dependent differences exist is unclear but newborn DCs are considered underdeveloped and functionally immature.
Using ontogenetic tracing of conventional DC precursors, I have found a previously unappreciated developmental heterogeneity of DCs that is particularly prominent in young mice. Preliminary data indicate that distinct waves of DC poiesis contribute to the functional differences between neonatal and adult DCs. I hypothesize that the neonatal DC compartment is not immature but rather that DC poiesis is developmentally regulated to create essential age-dependent immune balance. Further, I have identified a unique situation in early life to address a fundamental biological question, namely to what extent cellular function is pre-programmed by developmental origin (nature) versus environmental factors (nurture).
In this proposal, we will first use novel models to fate map the origin of the DC compartment with age. We will then define to what extent cellular origin determines age-dependent functions of DCs in immunity. Using innovative comparative gene expression profiling and integrative epigenomic analysis the cell intrinsic mechanisms regulating the age-dependent functions of DCs will be characterized. Because environmental factors in utero and after birth critically influence immune balance, we will finally define the impact of maternal infection and metabolic disease, as well as early microbial encounter on DC poiesis. Characterizing how developmentally regulated DC poiesis shapes the unique features of early life immunity will provide novel insights into immune development that are vital to advance vaccine strategies.
Summary
Early life immune balance is essential for survival and establishment of healthy immunity in later life. We aim to define how age-dependent regulation of dendritic cell (DC) development contributes to this crucial immune balance. DCs are versatile controllers of immunity that in neonates are qualitatively distinct from adults. Why such age-dependent differences exist is unclear but newborn DCs are considered underdeveloped and functionally immature.
Using ontogenetic tracing of conventional DC precursors, I have found a previously unappreciated developmental heterogeneity of DCs that is particularly prominent in young mice. Preliminary data indicate that distinct waves of DC poiesis contribute to the functional differences between neonatal and adult DCs. I hypothesize that the neonatal DC compartment is not immature but rather that DC poiesis is developmentally regulated to create essential age-dependent immune balance. Further, I have identified a unique situation in early life to address a fundamental biological question, namely to what extent cellular function is pre-programmed by developmental origin (nature) versus environmental factors (nurture).
In this proposal, we will first use novel models to fate map the origin of the DC compartment with age. We will then define to what extent cellular origin determines age-dependent functions of DCs in immunity. Using innovative comparative gene expression profiling and integrative epigenomic analysis the cell intrinsic mechanisms regulating the age-dependent functions of DCs will be characterized. Because environmental factors in utero and after birth critically influence immune balance, we will finally define the impact of maternal infection and metabolic disease, as well as early microbial encounter on DC poiesis. Characterizing how developmentally regulated DC poiesis shapes the unique features of early life immunity will provide novel insights into immune development that are vital to advance vaccine strategies.
Max ERC Funding
1 500 000 €
Duration
Start date: 2017-06-01, End date: 2022-05-31
Project acronym Born-Immune
Project Shaping of the Human Immune System by Primal Environmental Exposures In the Newborn Child
Researcher (PI) Klas Erik Petter Brodin
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Starting Grant (StG), LS6, ERC-2015-STG
Summary Immune systems are highly variable, but the sources of this variation are poorly understood. Genetic variation only explains a minor fraction of this, and we are unable to accurately predict the risk of immune mediated disease or severe infection in any given individual. I recently found that immune cells and proteins in healthy twins vary because of non-heritable influences (infections, vaccines, microbiota etc.), with only minor influences from heritable factors (Brodin, et al, Cell 2015). When and how such environmental influences shape our immune system is now important to understand. Birth represents the most transformational change in environment during the life of any individual. I propose, that environmental influences at birth, and during the first months of life could be particularly influential by imprinting on the regulatory mechanisms forming in the developing immune system. Adaptive changes in immune cell frequencies and functional states induced by early-life exposures could determine both the immune competence of the newborn, but potentially also its long-term trajectory towards immunological health or disease. Here, I propose a study of 1000 newborn children, followed longitudinally during their first 1000 days of life. By monitoring immune profiles and recording many environmental influences, we hope to understand how early life exposures can influence human immune system development. We have established a new assay based on Mass Cytometry and necessary data analysis tools (Brodin, et al, PNAS 2014), to simultaneously monitor the frequencies, phenotypes and functional states of more than 200 blood immune cell populations from only 100 microliters of blood. By monitoring environmental influences at regular follow-up visits, by questionnaires, serum measurements of infection, and gut microbiome sequencing, we aim to provide the most comprehensive analysis to date of immune system development in newborn children.
Summary
Immune systems are highly variable, but the sources of this variation are poorly understood. Genetic variation only explains a minor fraction of this, and we are unable to accurately predict the risk of immune mediated disease or severe infection in any given individual. I recently found that immune cells and proteins in healthy twins vary because of non-heritable influences (infections, vaccines, microbiota etc.), with only minor influences from heritable factors (Brodin, et al, Cell 2015). When and how such environmental influences shape our immune system is now important to understand. Birth represents the most transformational change in environment during the life of any individual. I propose, that environmental influences at birth, and during the first months of life could be particularly influential by imprinting on the regulatory mechanisms forming in the developing immune system. Adaptive changes in immune cell frequencies and functional states induced by early-life exposures could determine both the immune competence of the newborn, but potentially also its long-term trajectory towards immunological health or disease. Here, I propose a study of 1000 newborn children, followed longitudinally during their first 1000 days of life. By monitoring immune profiles and recording many environmental influences, we hope to understand how early life exposures can influence human immune system development. We have established a new assay based on Mass Cytometry and necessary data analysis tools (Brodin, et al, PNAS 2014), to simultaneously monitor the frequencies, phenotypes and functional states of more than 200 blood immune cell populations from only 100 microliters of blood. By monitoring environmental influences at regular follow-up visits, by questionnaires, serum measurements of infection, and gut microbiome sequencing, we aim to provide the most comprehensive analysis to date of immune system development in newborn children.
Max ERC Funding
1 422 339 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym CAAXPROCESSINGHUMDIS
Project CAAX Protein Processing in Human DIsease: From Cancer to Progeria
Researcher (PI) Martin Olof Bergö
Host Institution (HI) GOETEBORGS UNIVERSITET
Call Details Starting Grant (StG), LS6, ERC-2007-StG
Summary My objective is to understand the physiologic and medical importance of the posttranslational processing of CAAX proteins (e.g., K-RAS and prelamin A) and to define the suitability of the CAAX protein processing enzymes as therapeutic targets for the treatment of cancer and progeria. CAAX proteins undergo three posttranslational processing steps at a carboxyl-terminal CAAX motif. These processing steps, which are mediated by four different enzymes (FTase, GGTase-I, RCE1, and ICMT), increase the hydrophobicity of the carboxyl terminus of the protein and thereby facilitate interactions with membrane surfaces. Somatic mutations in K-RAS deregulate cell growth and are etiologically involved in the pathogenesis of many forms of cancer. A mutation in prelamin A causes Hutchinson-Gilford progeria syndrome—a pediatric progeroid syndrome associated with misshaped cell nuclei and a host of aging-like disease phenotypes. One strategy to render the mutant K-RAS and prelamin A less harmful is to interfere with their ability to bind to membrane surfaces (e.g., the plasma membrane and the nuclear envelope). This could be accomplished by inhibiting the enzymes that modify the CAAX motif. My Specific Aims are: (1) To define the suitability of the CAAX processing enzymes as therapeutic targets in the treatment of K-RAS-induced lung cancer and leukemia; and (2) To test the hypothesis that inactivation of FTase or ICMT will ameliorate disease phenotypes of progeria. I have developed genetic strategies to produce lung cancer or leukemia in mice by activating an oncogenic K-RAS and simultaneously inactivating different CAAX processing enzymes. I will also inactivate several CAAX processing enzymes in mice with progeria—both before the emergence of phenotypes and after the development of advanced disease phenotypes. These experiments should reveal whether the absence of the different CAAX processing enzymes affects the onset, progression, or regression of cancer and progeria.
Summary
My objective is to understand the physiologic and medical importance of the posttranslational processing of CAAX proteins (e.g., K-RAS and prelamin A) and to define the suitability of the CAAX protein processing enzymes as therapeutic targets for the treatment of cancer and progeria. CAAX proteins undergo three posttranslational processing steps at a carboxyl-terminal CAAX motif. These processing steps, which are mediated by four different enzymes (FTase, GGTase-I, RCE1, and ICMT), increase the hydrophobicity of the carboxyl terminus of the protein and thereby facilitate interactions with membrane surfaces. Somatic mutations in K-RAS deregulate cell growth and are etiologically involved in the pathogenesis of many forms of cancer. A mutation in prelamin A causes Hutchinson-Gilford progeria syndrome—a pediatric progeroid syndrome associated with misshaped cell nuclei and a host of aging-like disease phenotypes. One strategy to render the mutant K-RAS and prelamin A less harmful is to interfere with their ability to bind to membrane surfaces (e.g., the plasma membrane and the nuclear envelope). This could be accomplished by inhibiting the enzymes that modify the CAAX motif. My Specific Aims are: (1) To define the suitability of the CAAX processing enzymes as therapeutic targets in the treatment of K-RAS-induced lung cancer and leukemia; and (2) To test the hypothesis that inactivation of FTase or ICMT will ameliorate disease phenotypes of progeria. I have developed genetic strategies to produce lung cancer or leukemia in mice by activating an oncogenic K-RAS and simultaneously inactivating different CAAX processing enzymes. I will also inactivate several CAAX processing enzymes in mice with progeria—both before the emergence of phenotypes and after the development of advanced disease phenotypes. These experiments should reveal whether the absence of the different CAAX processing enzymes affects the onset, progression, or regression of cancer and progeria.
Max ERC Funding
1 689 600 €
Duration
Start date: 2008-06-01, End date: 2013-05-31
Project acronym CIRCODE
Project Cell-type specific mechanisms regulating rhythms in leukocyte homing
Researcher (PI) Christoph Andreas Scheiermann
Host Institution (HI) LUDWIG-MAXIMILIANS-UNIVERSITAET MUENCHEN
Call Details Starting Grant (StG), LS6, ERC-2014-STG
Summary Leukocytes are the key components of the immune system that fight infections and provide tissue repair, yet their migration patterns throughout the body over the course of a day are completely unknown. Circadian, ~24 hour rhythms are emerging as important novel regulators of immune cell migration and function, which impacts inflammatory diseases such as myocardial infarction and sepsis. Altering leukocyte tissue infiltration and activation at the proper times provides an option for therapy that would maximize the clinical impact of drugs and vaccinations and minimize side effects.
We aim to create a four-dimensional map of leukocyte migration to organs in time and space and investigate with epigenetics techniques the molecular mechanisms that regulate cell-type specific rhythms. We will functionally define the daily oscillating molecular signature(s) of leukocytes and endothelial cells with novel proteomics approaches and thus identify a circadian traffic code that dictates the rhythmic migration of leukocyte subsets to specific organs under steady-state and inflammatory conditions with pharmacological and genetic tools. We will assess the impact of lineage-specific arrhythmicities on immune homeostasis and leukocyte trafficking using an innovative combination of novel genetic tools. Based on these data we will create a model predicting circadian leukocyte migration to tissues.
The project combines the disciplines of immunology and chronobiology by obtaining unprecedented information in time and space of circadian leukocyte trafficking and investigating how immune-cell specific oscillations are generated at the molecular level, which is of broad impact for both fields. Our extensive experience in the rhythmic control of the immune system makes us well poised to characterize the molecular components that orchestrate circadian leukocyte distribution across the body.
Summary
Leukocytes are the key components of the immune system that fight infections and provide tissue repair, yet their migration patterns throughout the body over the course of a day are completely unknown. Circadian, ~24 hour rhythms are emerging as important novel regulators of immune cell migration and function, which impacts inflammatory diseases such as myocardial infarction and sepsis. Altering leukocyte tissue infiltration and activation at the proper times provides an option for therapy that would maximize the clinical impact of drugs and vaccinations and minimize side effects.
We aim to create a four-dimensional map of leukocyte migration to organs in time and space and investigate with epigenetics techniques the molecular mechanisms that regulate cell-type specific rhythms. We will functionally define the daily oscillating molecular signature(s) of leukocytes and endothelial cells with novel proteomics approaches and thus identify a circadian traffic code that dictates the rhythmic migration of leukocyte subsets to specific organs under steady-state and inflammatory conditions with pharmacological and genetic tools. We will assess the impact of lineage-specific arrhythmicities on immune homeostasis and leukocyte trafficking using an innovative combination of novel genetic tools. Based on these data we will create a model predicting circadian leukocyte migration to tissues.
The project combines the disciplines of immunology and chronobiology by obtaining unprecedented information in time and space of circadian leukocyte trafficking and investigating how immune-cell specific oscillations are generated at the molecular level, which is of broad impact for both fields. Our extensive experience in the rhythmic control of the immune system makes us well poised to characterize the molecular components that orchestrate circadian leukocyte distribution across the body.
Max ERC Funding
1 497 688 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym CMVAGSTIMULUS
Project Molecular mechanisms of persistent antigenic stimulation in cytomegalovirus infection
Researcher (PI) Luka Cicin-Sain
Host Institution (HI) HELMHOLTZ-ZENTRUM FUR INFEKTIONSFORSCHUNG GMBH
Call Details Starting Grant (StG), LS6, ERC-2010-StG_20091118
Summary Cytomegalovirus (CMV) is a ubiquitous herpesvirus, latently persisting in the majority of the adult population worldwide. In these hosts, CMV-specific memory cells dominate the immune memory compartment. It follows that CMV-specific T-cells dominate the memory compartment of the majority of the human population worldwide.
I propose to define within this project the molecular mechanisms driving the inflation of CMV-specific T-cells. My central hypothesis is that expression levels of CMV peptides during latency, along with the avidity of T-cell receptors for peptide MHC complexes, define the amplitude of T-cell responses. A corollary hypothesis is that vigorous T-cell responses in CMV infection are defined by factors that drive CMV gene expression during latency, such as inflammatory stimuli.
This hypothesis will be verified in a model of in vivo CMV latency and immune monitoring. We will benefit from state-of-the-art inducible genetic systems, where recombinant mouse CMV will be deployed in transgenic mice. In latently infected mice, we will induce or suppress the expression of immunodominant CMV genes, and define downstream effects on T-cell response kinetics. In parallel, we will define the T-cell responses to high and low avidity peptides.
Understanding the mechanisms driving the strong T-cell response to CMV is of outstanding biological and clinical relevance. If strong T-cell responses may be redirected to target heterologous antigens of interest, CMV-based vaccine vectors might potentially allow the development of HIV or tumor vaccines. On the other hand, it is speculated that the control of latent CMV may overcommit the aging immune system and exhaust the T-cell repertoire. Given the CMV pervasiveness, discerning the mechanisms of its T-cell induction may define novel molecular targets for rejuvenation strategies. In either case, the proposed research has groundbreaking potential in the field of infection and immunity.
Summary
Cytomegalovirus (CMV) is a ubiquitous herpesvirus, latently persisting in the majority of the adult population worldwide. In these hosts, CMV-specific memory cells dominate the immune memory compartment. It follows that CMV-specific T-cells dominate the memory compartment of the majority of the human population worldwide.
I propose to define within this project the molecular mechanisms driving the inflation of CMV-specific T-cells. My central hypothesis is that expression levels of CMV peptides during latency, along with the avidity of T-cell receptors for peptide MHC complexes, define the amplitude of T-cell responses. A corollary hypothesis is that vigorous T-cell responses in CMV infection are defined by factors that drive CMV gene expression during latency, such as inflammatory stimuli.
This hypothesis will be verified in a model of in vivo CMV latency and immune monitoring. We will benefit from state-of-the-art inducible genetic systems, where recombinant mouse CMV will be deployed in transgenic mice. In latently infected mice, we will induce or suppress the expression of immunodominant CMV genes, and define downstream effects on T-cell response kinetics. In parallel, we will define the T-cell responses to high and low avidity peptides.
Understanding the mechanisms driving the strong T-cell response to CMV is of outstanding biological and clinical relevance. If strong T-cell responses may be redirected to target heterologous antigens of interest, CMV-based vaccine vectors might potentially allow the development of HIV or tumor vaccines. On the other hand, it is speculated that the control of latent CMV may overcommit the aging immune system and exhaust the T-cell repertoire. Given the CMV pervasiveness, discerning the mechanisms of its T-cell induction may define novel molecular targets for rejuvenation strategies. In either case, the proposed research has groundbreaking potential in the field of infection and immunity.
Max ERC Funding
1 498 456 €
Duration
Start date: 2011-04-01, End date: 2016-09-30
Project acronym DDRMac
Project DNA Damage Response-instructed Macrophage Differentiation in Granulomatous Diseases
Researcher (PI) Antigoni TRIANTAFYLLOPOULOU
Host Institution (HI) CHARITE - UNIVERSITAETSMEDIZIN BERLIN
Call Details Starting Grant (StG), LS6, ERC-2018-STG
Summary Macrophage differentiation programs are critical for the outcome of immunity against infection, chronic inflammatory diseases and cancer. How diverse inflammatory signals are translated to macrophage programs in the large range of human pathologies is largely unexplored. In the last years we focused on macrophage differentiation in granulomatous diseases. These affect millions worldwide, including young adults and children and tend to run a chronic course, with a high socioeconomic burden. Their common hallmark is the formation of granulomas, macrophage-driven structures of organized inflammation that replace healthy tissue. We revealed that macrophage precursors in granulomas experience a replication block and trigger the DNA Damage Response (DDR), a fundamental cellular process activated in response to genotoxic stress. This leads to the formation of multinucleated macrophages with tissue-remodelling signatures (Herrtwich, Cell 2016). Our work unravelled an intriguing link between genotoxic stress and granuloma-specific macrophage programs. The molecular pathways regulating DDR-driven macrophage differentiation and their role in chronic inflammatory pathologies remain however a black box. We hypothesize that the DDR promotes macrophage reprogramming to inflammation-maintaining modules. Such programs operate in granulomatous diseases and in chronic arthritis. Using state-of-the art genetic models, human tissues and an array of techniques crossing the fields of immunology, cell biology and cancer biology, our goal is to unravel the macrophage-specific response to genotoxic stress as an essential regulator of chronic inflammation-induced pathologies. The anticipated results will provide the scientific community with new knowledge on the role of genotoxic stress in immune dysregulation and will carry tremendous implications for the therapeutic targeting of macrophages in the context of chronic inflammatory diseases and cancer.
Summary
Macrophage differentiation programs are critical for the outcome of immunity against infection, chronic inflammatory diseases and cancer. How diverse inflammatory signals are translated to macrophage programs in the large range of human pathologies is largely unexplored. In the last years we focused on macrophage differentiation in granulomatous diseases. These affect millions worldwide, including young adults and children and tend to run a chronic course, with a high socioeconomic burden. Their common hallmark is the formation of granulomas, macrophage-driven structures of organized inflammation that replace healthy tissue. We revealed that macrophage precursors in granulomas experience a replication block and trigger the DNA Damage Response (DDR), a fundamental cellular process activated in response to genotoxic stress. This leads to the formation of multinucleated macrophages with tissue-remodelling signatures (Herrtwich, Cell 2016). Our work unravelled an intriguing link between genotoxic stress and granuloma-specific macrophage programs. The molecular pathways regulating DDR-driven macrophage differentiation and their role in chronic inflammatory pathologies remain however a black box. We hypothesize that the DDR promotes macrophage reprogramming to inflammation-maintaining modules. Such programs operate in granulomatous diseases and in chronic arthritis. Using state-of-the art genetic models, human tissues and an array of techniques crossing the fields of immunology, cell biology and cancer biology, our goal is to unravel the macrophage-specific response to genotoxic stress as an essential regulator of chronic inflammation-induced pathologies. The anticipated results will provide the scientific community with new knowledge on the role of genotoxic stress in immune dysregulation and will carry tremendous implications for the therapeutic targeting of macrophages in the context of chronic inflammatory diseases and cancer.
Max ERC Funding
1 500 000 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym Defensinactivity
Project The influence of environmental factors on antimicrobial activity of human intestinal defensins
Researcher (PI) Jan Wehkamp
Host Institution (HI) EBERHARD KARLS UNIVERSITAET TUEBINGEN
Call Details Starting Grant (StG), LS6, ERC-2012-StG_20111109
Summary Human epithelia are permanently challenged by microorganisms. In the gut, the fraction of strict anaerobic bacteria increases from proximal to distal, reaching 99% of bacterial species in the colon. Moreover, microbial metabolism causes a reduction of the environment to a low redox potential of only –200 mV to –300 mV. Defensins, characterised by three intramolecular disulfide-bridges, are key effector molecules of innate immunity that protect the host from infectious microbes. Human β-defensin 1 (hBD-1) is one of the most prominent peptides of its class but comparison with other defensins suggested only minor antibiotic killing activity. We could recently show that hBD-1 becomes a potent antimicrobial peptide against C. albicans and anaerobic, Gram-positive commensals of the human normal flora in a reducing environment (Nature 2011). The effect was attributable to the linear, reduced hBD-1 peptide.
Here we aim to investigate the antimicrobial activity of reduced hBD-1 in more detail. We will study the mechanism of its reduction by cell-culture experiments and in vitro reduction assays. The molecular details of its antibiotic action will be investigated by using bacterial mutants and further in vitro assays. Additionally we aim to characterise the antibiotic spectrum of reduced hBD-1 by using different antimicrobial assays. Also, we plan to systematically test human defensins under reducing conditions and different pH values that occur in the gut
Besides we will screen extracts of human intestinal tissue and stool samples for antimicrobial substances by using the conditions described above. Extracts will be purified by HPLC and antimicrobially active fractions will be examined by MALDI-TOF peptide mass fingerprint technique. We hope to identify novel peptides which have been overlooked due to standardized testing methods. Resembling the natural conditions as close as possible will help to better understand antibiotic mucosal host defense in the intestinal tract.
Summary
Human epithelia are permanently challenged by microorganisms. In the gut, the fraction of strict anaerobic bacteria increases from proximal to distal, reaching 99% of bacterial species in the colon. Moreover, microbial metabolism causes a reduction of the environment to a low redox potential of only –200 mV to –300 mV. Defensins, characterised by three intramolecular disulfide-bridges, are key effector molecules of innate immunity that protect the host from infectious microbes. Human β-defensin 1 (hBD-1) is one of the most prominent peptides of its class but comparison with other defensins suggested only minor antibiotic killing activity. We could recently show that hBD-1 becomes a potent antimicrobial peptide against C. albicans and anaerobic, Gram-positive commensals of the human normal flora in a reducing environment (Nature 2011). The effect was attributable to the linear, reduced hBD-1 peptide.
Here we aim to investigate the antimicrobial activity of reduced hBD-1 in more detail. We will study the mechanism of its reduction by cell-culture experiments and in vitro reduction assays. The molecular details of its antibiotic action will be investigated by using bacterial mutants and further in vitro assays. Additionally we aim to characterise the antibiotic spectrum of reduced hBD-1 by using different antimicrobial assays. Also, we plan to systematically test human defensins under reducing conditions and different pH values that occur in the gut
Besides we will screen extracts of human intestinal tissue and stool samples for antimicrobial substances by using the conditions described above. Extracts will be purified by HPLC and antimicrobially active fractions will be examined by MALDI-TOF peptide mass fingerprint technique. We hope to identify novel peptides which have been overlooked due to standardized testing methods. Resembling the natural conditions as close as possible will help to better understand antibiotic mucosal host defense in the intestinal tract.
Max ERC Funding
1 500 000 €
Duration
Start date: 2013-02-01, End date: 2018-01-31
Project acronym Diet-namic
Project From fast food to healthy diet: Addressing the dynamic molecular mechanism of sequential diet switch-induced T cell plasticity for the purpose of developing new treatments for immuno-mediated diseases
Researcher (PI) Nicola Gagliani
Host Institution (HI) UNIVERSITAETSKLINIKUM HAMBURG-EPPENDORF
Call Details Starting Grant (StG), LS6, ERC-2016-STG
Summary "The incidence of chronic immune-mediated inflammatory diseases is continually increasing. Chronic inflammation has been linked to intestinal carcinogenesis, which is the second leading cause of cancer-related deaths. The cause of this increase could be the unprecedented dietary abundance typical of “Western” countries. Different types of diets shape the genetic composition and metabolic activity of human intestinal microorganisms; microbiota. There is a continuous cross talk between the microbiota and the immune system. For these reasons, the hypothesis that a “bad” diet promotes a chronic state of intestinal inflammation by shaping the microbiota and in turn carcinogenesis could be supported. However, this hypothesis and whether this is a reversible process remain to be tested.
It has recently been shown that the composition and metabolism of the microbiota is plastic and it can be rapidly “reprogrammed” by switching to a healthier diet. This plastic behaviour has also been attributed to T helper cells. We have shown that Th17 cells, originally thought to be a stable T helper linage, can convert into a more pathogenic phenotype contributing to chronic inflammation or can acquire regulatory functions promoting the resolution of the inflammation.
This project aims to reveal whether mouse and human Th17 cells can quickly adapt to the microbiota as the microbiota does to the diet and in turn mediate the diet effects. By using a unique set of sophisticated transgenic mice we will also test whether the immune system can be corrected by a “simple” change in diet – a widely held belief not yet substantiated.
Studying the potential ""synchronized ballet"" of the diet and the immune system will reveal both the enormous dynamism and the revolutionary therapeutic opportunities intrinsic to T cell biology. This project will furthermore identify molecular targets for pharmacological treatments to reverse inflammatory diseases when a simple diet change no longer suffices."
Summary
"The incidence of chronic immune-mediated inflammatory diseases is continually increasing. Chronic inflammation has been linked to intestinal carcinogenesis, which is the second leading cause of cancer-related deaths. The cause of this increase could be the unprecedented dietary abundance typical of “Western” countries. Different types of diets shape the genetic composition and metabolic activity of human intestinal microorganisms; microbiota. There is a continuous cross talk between the microbiota and the immune system. For these reasons, the hypothesis that a “bad” diet promotes a chronic state of intestinal inflammation by shaping the microbiota and in turn carcinogenesis could be supported. However, this hypothesis and whether this is a reversible process remain to be tested.
It has recently been shown that the composition and metabolism of the microbiota is plastic and it can be rapidly “reprogrammed” by switching to a healthier diet. This plastic behaviour has also been attributed to T helper cells. We have shown that Th17 cells, originally thought to be a stable T helper linage, can convert into a more pathogenic phenotype contributing to chronic inflammation or can acquire regulatory functions promoting the resolution of the inflammation.
This project aims to reveal whether mouse and human Th17 cells can quickly adapt to the microbiota as the microbiota does to the diet and in turn mediate the diet effects. By using a unique set of sophisticated transgenic mice we will also test whether the immune system can be corrected by a “simple” change in diet – a widely held belief not yet substantiated.
Studying the potential ""synchronized ballet"" of the diet and the immune system will reveal both the enormous dynamism and the revolutionary therapeutic opportunities intrinsic to T cell biology. This project will furthermore identify molecular targets for pharmacological treatments to reverse inflammatory diseases when a simple diet change no longer suffices."
Max ERC Funding
1 499 695 €
Duration
Start date: 2016-12-01, End date: 2021-11-30
Project acronym DrySeasonPf
Project Dry season P. falciparum reservoir
Researcher (PI) Silvia VILAR PORTUGAL
Host Institution (HI) UNIVERSITATSKLINIKUM HEIDELBERG
Call Details Starting Grant (StG), LS6, ERC-2017-STG
Summary The mosquito-borne Plasmodium falciparum parasite is responsible for over 200 million malaria cases and nearly half a million deaths each year among African children. Dependent on Anopheles mosquito for transmission, the parasite faces a challenge during the dry season in the regions where rain seasonality limits vector availability for several months. While malaria cases are restricted to the wet season, clinically silent P. falciparum infections can persist through the dry season and are an important reservoir for transmission. Our preliminary data provides unequivocal evidence that P. falciparum modulates its transcription during the dry season, while the host immune response seems to be minimally affected, suggesting that the parasite has the ability to adapt to a vector-free environment for long periods of time. Understanding the mechanisms which allow the parasite to remain undetectable in absence of mosquito vector, and to restart transmission in the ensuing rainy season will reveal complex interactions between P. falciparum and its host. To that end I propose to: (i) Identify the Plasmodium signalling pathway(s) and metabolic profile associated with long-term maintenance of low parasitaemias during the dry season, (ii) Determine which PfEMP1 are expressed by parasites during the dry season and how effectively they are detected by the immune system, and (iii) Investigate the kinetics of P. falciparum gametocytogenesis, its ability to transmit during the dry season, and uncover sensing molecules and mechanisms of the disappearance and return of the mosquito vector Undoubtedly, results arising from the present multidisciplinary proposal will provide novel insights into the cell biology of dry season P. falciparum parasites, will increase our understanding of their interactions with their hosts and environment. Furthermore, it may benefit the international development agenda goals to design public health strategies to fight malaria.
Summary
The mosquito-borne Plasmodium falciparum parasite is responsible for over 200 million malaria cases and nearly half a million deaths each year among African children. Dependent on Anopheles mosquito for transmission, the parasite faces a challenge during the dry season in the regions where rain seasonality limits vector availability for several months. While malaria cases are restricted to the wet season, clinically silent P. falciparum infections can persist through the dry season and are an important reservoir for transmission. Our preliminary data provides unequivocal evidence that P. falciparum modulates its transcription during the dry season, while the host immune response seems to be minimally affected, suggesting that the parasite has the ability to adapt to a vector-free environment for long periods of time. Understanding the mechanisms which allow the parasite to remain undetectable in absence of mosquito vector, and to restart transmission in the ensuing rainy season will reveal complex interactions between P. falciparum and its host. To that end I propose to: (i) Identify the Plasmodium signalling pathway(s) and metabolic profile associated with long-term maintenance of low parasitaemias during the dry season, (ii) Determine which PfEMP1 are expressed by parasites during the dry season and how effectively they are detected by the immune system, and (iii) Investigate the kinetics of P. falciparum gametocytogenesis, its ability to transmit during the dry season, and uncover sensing molecules and mechanisms of the disappearance and return of the mosquito vector Undoubtedly, results arising from the present multidisciplinary proposal will provide novel insights into the cell biology of dry season P. falciparum parasites, will increase our understanding of their interactions with their hosts and environment. Furthermore, it may benefit the international development agenda goals to design public health strategies to fight malaria.
Max ERC Funding
1 500 000 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym ENTRI
Project Enteric-nervous-system-mediated regulation of intestinal inflammation
Researcher (PI) Christoph Klose
Host Institution (HI) CHARITE - UNIVERSITAETSMEDIZIN BERLIN
Call Details Starting Grant (StG), LS6, ERC-2018-STG
Summary Environmental and internal stimuli are constantly sensed by the body’s two large sensory units, the nervous system and the immune system. Integration of these sensory signals and translation into effector responses are essential for maintaining body homeostasis. While some of the intrinsic pathways of the immune or nervous system have been investigated, how the two sensory interfaces coordinate their responses remains elusive. We have recently investigated neuro-immune interaction at the mucosa of the intestine, which is densely innervated by the enteric nervous system (ENS). Our research has exposed a previously unrecognized pathway used by enteric neurons to shape type 2 immunity at mucosal barriers. Cholinergic enteric neurons produce the neuropeptide Neuromedin U (NMU) to elicit potent activation of type 2 innate lymphoid cells (ILC2s) via Neuromedin U receptor 1, selectively expressed by ILC2s. Interestingly, NMU stimulated protective immunity against the parasite Nippostrongylus brasiliensis but also triggered allergic lung inflammation. Therefore, the NMU-NMUR1 axis provides an excellent opportunity to study how neurons and immune cells interact to regulate immune responses and maintain body homeostasis. We propose to generate and use elegant genetic tools, which will allow us to systematically investigate the consequences of neuro-immune crosstalk at mucosal surfaces in various disease models. These tools will enable us to selectively measure and interfere with neuronal and ILC2 gene expression and function, thereby leading to an unprecedented understanding of how the components of neuro-immune crosstalk contribute to parasite immunity or allergic disease development. Furthermore, we will progress into translational aspects of NMU-regulated immune activation for human immunology. Therefore, our research has the potential to develop basic concepts of mucosal immune regulation and such discoveries could also be harnessed for therapeutic intervention.
Summary
Environmental and internal stimuli are constantly sensed by the body’s two large sensory units, the nervous system and the immune system. Integration of these sensory signals and translation into effector responses are essential for maintaining body homeostasis. While some of the intrinsic pathways of the immune or nervous system have been investigated, how the two sensory interfaces coordinate their responses remains elusive. We have recently investigated neuro-immune interaction at the mucosa of the intestine, which is densely innervated by the enteric nervous system (ENS). Our research has exposed a previously unrecognized pathway used by enteric neurons to shape type 2 immunity at mucosal barriers. Cholinergic enteric neurons produce the neuropeptide Neuromedin U (NMU) to elicit potent activation of type 2 innate lymphoid cells (ILC2s) via Neuromedin U receptor 1, selectively expressed by ILC2s. Interestingly, NMU stimulated protective immunity against the parasite Nippostrongylus brasiliensis but also triggered allergic lung inflammation. Therefore, the NMU-NMUR1 axis provides an excellent opportunity to study how neurons and immune cells interact to regulate immune responses and maintain body homeostasis. We propose to generate and use elegant genetic tools, which will allow us to systematically investigate the consequences of neuro-immune crosstalk at mucosal surfaces in various disease models. These tools will enable us to selectively measure and interfere with neuronal and ILC2 gene expression and function, thereby leading to an unprecedented understanding of how the components of neuro-immune crosstalk contribute to parasite immunity or allergic disease development. Furthermore, we will progress into translational aspects of NMU-regulated immune activation for human immunology. Therefore, our research has the potential to develop basic concepts of mucosal immune regulation and such discoveries could also be harnessed for therapeutic intervention.
Max ERC Funding
1 499 638 €
Duration
Start date: 2019-07-01, End date: 2024-06-30
Project acronym EpiTune
Project Epigenetic fine-tuning of T cells for improved adoptive cell therapy
Researcher (PI) Julia Polansky-Biskup
Host Institution (HI) CHARITE - UNIVERSITAETSMEDIZIN BERLIN
Call Details Starting Grant (StG), LS6, ERC-2018-STG
Summary "Adoptive T cell therapy is a promising approach in various clinical settings, from target-specific immune reconstitution fighting cancer and chronic infections to combating undesired immune reactivity during auto-immunity and after organ transplantation.
However, its clinical application is currently hampered by: 1) the acquisition of senescence during the required in vitro expansion phase of T cells which limits their survival and fitness after infusion into the patient, and 2) the functional plasticity of T cells, which is sensitive to the inflammatory environment they encounter after transfusion and which might result in a functional switch from the desired effect (e.g. immunosuppressive) to the opposite one (pro-inflammatory).
I want to tackle these obstacles from a new molecular angle, utilizing the profound impact of epigenetic mechanisms on the senescence process as well as on the functional imprinting of T lymphocytes. Epigenetic players such as DNA methylation essentially contribute to T cell differentiation and harbor the unique prospect to imprint a stable developmental and functional state in the genomic structure of a cell, as we could recently show in our basic immune-epigenetic studies. Therefore, I here propose to equip T lymphocytes with the required properties for their successful and safe therapeutic application, including their functional fine-tuning according to the clinical need by directed modifications of the epigenome
('Epi-tuning').
To reach these goals I want: 1) to reveal strategies for the directed manipulation of the epigenetically-driven mechanism of cellular senescence and 2) to apply state-of-the-art CRISPR/Cas9-mediated epigenetic editing approaches for the imprinting of a desired functional state of therapeutic T cell products. These innovative epigenetic ""one-shot"" manipulations during the in vitro expansion phase should advance T cell therapy towards improved efficiency, stability as well as safety."
Summary
"Adoptive T cell therapy is a promising approach in various clinical settings, from target-specific immune reconstitution fighting cancer and chronic infections to combating undesired immune reactivity during auto-immunity and after organ transplantation.
However, its clinical application is currently hampered by: 1) the acquisition of senescence during the required in vitro expansion phase of T cells which limits their survival and fitness after infusion into the patient, and 2) the functional plasticity of T cells, which is sensitive to the inflammatory environment they encounter after transfusion and which might result in a functional switch from the desired effect (e.g. immunosuppressive) to the opposite one (pro-inflammatory).
I want to tackle these obstacles from a new molecular angle, utilizing the profound impact of epigenetic mechanisms on the senescence process as well as on the functional imprinting of T lymphocytes. Epigenetic players such as DNA methylation essentially contribute to T cell differentiation and harbor the unique prospect to imprint a stable developmental and functional state in the genomic structure of a cell, as we could recently show in our basic immune-epigenetic studies. Therefore, I here propose to equip T lymphocytes with the required properties for their successful and safe therapeutic application, including their functional fine-tuning according to the clinical need by directed modifications of the epigenome
('Epi-tuning').
To reach these goals I want: 1) to reveal strategies for the directed manipulation of the epigenetically-driven mechanism of cellular senescence and 2) to apply state-of-the-art CRISPR/Cas9-mediated epigenetic editing approaches for the imprinting of a desired functional state of therapeutic T cell products. These innovative epigenetic ""one-shot"" manipulations during the in vitro expansion phase should advance T cell therapy towards improved efficiency, stability as well as safety."
Max ERC Funding
1 489 725 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym FatemapB
Project High Resolution Mapping of Fetal and Adult B Cell Fates During Ontogeny
Researcher (PI) Joan YUAN
Host Institution (HI) LUNDS UNIVERSITET
Call Details Starting Grant (StG), LS6, ERC-2016-STG
Summary FateMapB aims to understand how the unique differentiation potential of fetal hematopoietic stem and progenitor cells
(HSPCs) contribute to functionally distinct cell types of the adult immune system. While most immune cells are replenished
by HSPCs through life, others emerge during a limited window in fetal life and sustain through self-renewal in situ. The
lineage identity of fetal HSPCs, and the extent of their contribution to the adult immune repertoire remain surprisingly
unclear. I previously identified the fetal specific RNA binding protein Lin28b as a post-transcriptional molecular switch
capable of inducing fetal-like hematopoiesis in adult bone marrow HSPCs (Yuan et al. Science, 2012). This discovery has
afforded me with unique perspectives on the formation of the mammalian immune system. The concept that the mature
immune system is a mosaic of fetal and adult derived cell types is addressed herein with an emphasis on the B cell lineage.
We will use two complementary lineage-tracing technologies to stratify the immune system as a function of developmental
time, generating fundamental insight into the division of labor between fetal and adult HSPCs that ultimately provides
effective host protection.
Aim 1. Determine the qualitative and quantitative contribution of fetal HSPCs to the mature immune repertoire in situ
through Cre recombination mediated lineage-tracing.
Aim 2. Resolve the disputed lineage relationship between fetal derived B1a cells and adult derived B2 cells by single cell
lineage-tracing using cellular barcoding in vivo.
Aim 3. Characterize the mechanism and effector functions of Lin28b induced B1a cell development for assessing the
clinical utility of inducible fetal-like lymphopoiesis.
The implications of FateMapB extend beyond normal development to immune regeneration and age-related features of
leukemogenesis. Finally, our combinatorial lineage-tracing approach enables dissection of cell fates with previously
unattainable resolution.
Summary
FateMapB aims to understand how the unique differentiation potential of fetal hematopoietic stem and progenitor cells
(HSPCs) contribute to functionally distinct cell types of the adult immune system. While most immune cells are replenished
by HSPCs through life, others emerge during a limited window in fetal life and sustain through self-renewal in situ. The
lineage identity of fetal HSPCs, and the extent of their contribution to the adult immune repertoire remain surprisingly
unclear. I previously identified the fetal specific RNA binding protein Lin28b as a post-transcriptional molecular switch
capable of inducing fetal-like hematopoiesis in adult bone marrow HSPCs (Yuan et al. Science, 2012). This discovery has
afforded me with unique perspectives on the formation of the mammalian immune system. The concept that the mature
immune system is a mosaic of fetal and adult derived cell types is addressed herein with an emphasis on the B cell lineage.
We will use two complementary lineage-tracing technologies to stratify the immune system as a function of developmental
time, generating fundamental insight into the division of labor between fetal and adult HSPCs that ultimately provides
effective host protection.
Aim 1. Determine the qualitative and quantitative contribution of fetal HSPCs to the mature immune repertoire in situ
through Cre recombination mediated lineage-tracing.
Aim 2. Resolve the disputed lineage relationship between fetal derived B1a cells and adult derived B2 cells by single cell
lineage-tracing using cellular barcoding in vivo.
Aim 3. Characterize the mechanism and effector functions of Lin28b induced B1a cell development for assessing the
clinical utility of inducible fetal-like lymphopoiesis.
The implications of FateMapB extend beyond normal development to immune regeneration and age-related features of
leukemogenesis. Finally, our combinatorial lineage-tracing approach enables dissection of cell fates with previously
unattainable resolution.
Max ERC Funding
1 499 905 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym FLAMMASEC
Project "Inflammasome-induced IL-1 Secretion: Route, Mechanism, and Cell Fate"
Researcher (PI) Olaf Groß
Host Institution (HI) UNIVERSITAETSKLINIKUM FREIBURG
Call Details Starting Grant (StG), LS6, ERC-2013-StG
Summary "Inflammasomes are intracellular danger-sensing protein complexes that are important for host protection. They initiate inflammation by controlling the activity of the proinflammatory cytokine interleukin-1β (IL-1β). Unlike most other cytokines, IL-1β is produced and retained in the cytoplasm in an inactive pro-form. Inflammasome-dependent maturation of proIL-1β is mediated by the common component of all inflammasomes, the protease caspase-1. Caspase-1 also controls the secretion of IL-1β, but the mechanism and route of secretion are unknown. We have recently demonstrated that the ability of caspase-1 to control IL-1β secretion is not dependent on its protease activity, but rather on a scaffold or adapter function of caspase-1. Furthermore, we and others could show that caspase-1 can control the secretion of non-substrates like IL-1α. These insights provide us with new and potentially revealing means to investigate the downstream effector functions of caspase-1, including the route and mechanism of IL-1 secretion. We will develop new tools to study the process of IL-1 secretion by microscopy and the novel mode-of-action of caspase-1 through the generation of transgenic models.
Despite the important role of IL-1 in host defence against infection, dysregulated inflammasome activation and IL-1 production has a causal role in a number of acquired and hereditary auto-inflammatory conditions. These include particle-induced sterile inflammation (as is seen in gout and asbestosis), hereditary periodic fever syndromes, and metabolic diseases like diabetes and atherosclerosis. Currently, recombinant proteins that block the IL-1 receptor or deplete secreted IL-1 are used to treat IL-1-dependent diseases. These are costly treatments, and are also therapeutically cumbersome since they are not orally available. We hope that a better understanding of caspase-1-mediated secretion of IL-1 will unveil mechanisms that may serve as targets for future therapies for these diseases."
Summary
"Inflammasomes are intracellular danger-sensing protein complexes that are important for host protection. They initiate inflammation by controlling the activity of the proinflammatory cytokine interleukin-1β (IL-1β). Unlike most other cytokines, IL-1β is produced and retained in the cytoplasm in an inactive pro-form. Inflammasome-dependent maturation of proIL-1β is mediated by the common component of all inflammasomes, the protease caspase-1. Caspase-1 also controls the secretion of IL-1β, but the mechanism and route of secretion are unknown. We have recently demonstrated that the ability of caspase-1 to control IL-1β secretion is not dependent on its protease activity, but rather on a scaffold or adapter function of caspase-1. Furthermore, we and others could show that caspase-1 can control the secretion of non-substrates like IL-1α. These insights provide us with new and potentially revealing means to investigate the downstream effector functions of caspase-1, including the route and mechanism of IL-1 secretion. We will develop new tools to study the process of IL-1 secretion by microscopy and the novel mode-of-action of caspase-1 through the generation of transgenic models.
Despite the important role of IL-1 in host defence against infection, dysregulated inflammasome activation and IL-1 production has a causal role in a number of acquired and hereditary auto-inflammatory conditions. These include particle-induced sterile inflammation (as is seen in gout and asbestosis), hereditary periodic fever syndromes, and metabolic diseases like diabetes and atherosclerosis. Currently, recombinant proteins that block the IL-1 receptor or deplete secreted IL-1 are used to treat IL-1-dependent diseases. These are costly treatments, and are also therapeutically cumbersome since they are not orally available. We hope that a better understanding of caspase-1-mediated secretion of IL-1 will unveil mechanisms that may serve as targets for future therapies for these diseases."
Max ERC Funding
1 495 533 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym FUTURE T3SS
Project Bacterial effector secretion: Function and Architecture of the Type 3 Secretion System
Researcher (PI) Michael Kolbe
Host Institution (HI) HELMHOLTZ-ZENTRUM FUR INFEKTIONSFORSCHUNG GMBH
Call Details Starting Grant (StG), LS6, ERC-2012-StG_20111109
Summary Bacterial pathogens secrete effector proteins to manipulate host cells during infection. In Gram-negative bacteria the conserved type 3 secretion system (T3SS) delivers effector proteins to the host cell in a spatiotemporal manner. Although major T3SS constituting components were identified the function of this macromolecular complex remains elusive which is why the transport mechanism is not understood yet.
Here, I present my research proposal of the functional and structural analysis of the T3SS with respect to the effector transport dynamics. The proposal is divided in three sections addressing the T3SS architecture, the cytosolic mechanism preceding effector molecule release and the molecular mechanisms of secretion regulation.
Research focus of the first section is on the 3-dimensional structural analysis of isolated T3SS. The proposed studies should help detecting structural features related to the effector transport. A combination of electron microscopy (EM) and mass spectrometry is proposed to analyze the structure and surface properties of the transport channel together with bound effector molecule. I am also planning localization of bound lipids as well as to detect lipid induced structural changes in the T3SS by EM. Characterization of cytosolic processes, preceding the translocation is the focus of the second section. Effector molecule targeting, insertion into the T3SS channel and the chaperone function will be studied using a combination of biochemical and biophysical techniques.Finally, I propose experiments to analyze the T3SS regulation. Here, the research focus is on host signal reception and downstream posttranslational modifications inside bacteria as well as on conformational dynamics of the T3SS needle tip complex.
The overall goal of the proposed work is to understand the molecular mechanisms of the T3SS. I believe these studies will impact both the understanding of bacterial pathogenesis as well as the transmembrane transport of proteins.
Summary
Bacterial pathogens secrete effector proteins to manipulate host cells during infection. In Gram-negative bacteria the conserved type 3 secretion system (T3SS) delivers effector proteins to the host cell in a spatiotemporal manner. Although major T3SS constituting components were identified the function of this macromolecular complex remains elusive which is why the transport mechanism is not understood yet.
Here, I present my research proposal of the functional and structural analysis of the T3SS with respect to the effector transport dynamics. The proposal is divided in three sections addressing the T3SS architecture, the cytosolic mechanism preceding effector molecule release and the molecular mechanisms of secretion regulation.
Research focus of the first section is on the 3-dimensional structural analysis of isolated T3SS. The proposed studies should help detecting structural features related to the effector transport. A combination of electron microscopy (EM) and mass spectrometry is proposed to analyze the structure and surface properties of the transport channel together with bound effector molecule. I am also planning localization of bound lipids as well as to detect lipid induced structural changes in the T3SS by EM. Characterization of cytosolic processes, preceding the translocation is the focus of the second section. Effector molecule targeting, insertion into the T3SS channel and the chaperone function will be studied using a combination of biochemical and biophysical techniques.Finally, I propose experiments to analyze the T3SS regulation. Here, the research focus is on host signal reception and downstream posttranslational modifications inside bacteria as well as on conformational dynamics of the T3SS needle tip complex.
The overall goal of the proposed work is to understand the molecular mechanisms of the T3SS. I believe these studies will impact both the understanding of bacterial pathogenesis as well as the transmembrane transport of proteins.
Max ERC Funding
1 488 240 €
Duration
Start date: 2013-01-01, End date: 2017-12-31
Project acronym GAMES
Project Gut Microbiota in Nervous System Autoimmunity: Molecular Mechanisms of Disease Initiation and Regulation
Researcher (PI) Gurumoorthy Krishnamoorthy
Host Institution (HI) MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN EV
Call Details Starting Grant (StG), LS6, ERC-2014-STG
Summary Multiple Sclerosis (MS), an autoimmune demyelinating disease affecting the central nervous system (CNS), causes tremendous disability in young adults and inflicts huge economic burden on the society. The incidence of MS is steadily increasing in many countries arguing for environmental factors driven changes in disease induction. How and which environmental factors contribute to disease initiation and progression is unknown. Using a spontaneous mouse model of MS, we have shown that the gut microbiota is essential in triggering CNS autoimmunity. In contrast to the mice housed in conventional housing conditions, germ free (GF) mice, devoid of gut bacteria, were protected from spontaneous experimental autoimmune encephalomyelitis (sEAE). Re-colonization of GF mice with a complex regular gut flora derived from specific pathogen free (SPF) mice resulted in sEAE within 2-3 months. The re-colonization also triggered pro-inflammatory T and B cell responses. However, colonization of GF mice with a reduced gut flora failed to induce sEAE during our observation period suggesting a “specific” rather than a “broader” microbial trigger. In this proposal, I want to study the role of gut microbiota in CNS autoimmunity with the following aims:
Aim 1: CNS autoimmunity triggering/protecting gut microbes and host immune responses
I want to study how and which gut bacterial species are modulating CNS autoimmunity to better understand the origin of autoimmune responses and their relation to host immune responses.
Aim 2: Molecular mechanisms of sensing of gut microbiota and microbial metabolites during CNS autoimmunity
I want to identify the molecular pathways that are involved in sensing the gut microbiota and its metabolites which are relevant to CNS autoimmunity.
Aim 3: Therapeutic application of gut microbiota for CNS autoimmunity
I want to identify therapeutic strategies targeting gut microbiota to limit the development of inflammatory processes during CNS autoimmunity.
Summary
Multiple Sclerosis (MS), an autoimmune demyelinating disease affecting the central nervous system (CNS), causes tremendous disability in young adults and inflicts huge economic burden on the society. The incidence of MS is steadily increasing in many countries arguing for environmental factors driven changes in disease induction. How and which environmental factors contribute to disease initiation and progression is unknown. Using a spontaneous mouse model of MS, we have shown that the gut microbiota is essential in triggering CNS autoimmunity. In contrast to the mice housed in conventional housing conditions, germ free (GF) mice, devoid of gut bacteria, were protected from spontaneous experimental autoimmune encephalomyelitis (sEAE). Re-colonization of GF mice with a complex regular gut flora derived from specific pathogen free (SPF) mice resulted in sEAE within 2-3 months. The re-colonization also triggered pro-inflammatory T and B cell responses. However, colonization of GF mice with a reduced gut flora failed to induce sEAE during our observation period suggesting a “specific” rather than a “broader” microbial trigger. In this proposal, I want to study the role of gut microbiota in CNS autoimmunity with the following aims:
Aim 1: CNS autoimmunity triggering/protecting gut microbes and host immune responses
I want to study how and which gut bacterial species are modulating CNS autoimmunity to better understand the origin of autoimmune responses and their relation to host immune responses.
Aim 2: Molecular mechanisms of sensing of gut microbiota and microbial metabolites during CNS autoimmunity
I want to identify the molecular pathways that are involved in sensing the gut microbiota and its metabolites which are relevant to CNS autoimmunity.
Aim 3: Therapeutic application of gut microbiota for CNS autoimmunity
I want to identify therapeutic strategies targeting gut microbiota to limit the development of inflammatory processes during CNS autoimmunity.
Max ERC Funding
1 499 946 €
Duration
Start date: 2015-06-01, End date: 2020-05-31
Project acronym HIV1ABTHERAPY
Project Antibody-Mediated Therapy of HIV-1 Infection
Researcher (PI) Florian Klein
Host Institution (HI) KLINIKUM DER UNIVERSITAET ZU KOELN
Call Details Starting Grant (StG), LS6, ERC-2014-STG
Summary Antibodies are destined to neutralize pathogens and can prevent and fight infectious diseases. Over the last years, advances in single B cell cloning resulted in the isolation of highly potent and broad HIV-1 neutralizing antibodies (bNAbs) that have been shown to prevent SHIV infection in non-human primates (NHPs). Recently, we have demonstrated that a combination of bNAbs can suppress HIV-1 replication in humanized mice, reducing viremia to undetectable levels. Moreover, bNAb therapy of SHIV-infected NHPs induced a rapid decline in viremia, followed by a prolonged control due to the long half-life of the antibodies. While these results strongly encourage the clinical evaluation of bNAbs in HIV-1 therapy, it is of critical importance to understand how the therapeutic potential of antibodies can be harnessed in the most effective way. Therefore, we aim to: I.) Identify exceptionally potent HIV-1 neutralizing antibodies that will be a crucial component of immunotherapy. By establishing novel methods for single-cell sorting and high-throughput sequencing we want to identify bNAbs targeting novel epitopes. II.) Prevent HIV-1 escape applying rationally designed treatment strategies targeting conserved functional sites for HIV-1 entry. III.) Evaluate immune markers and function in relation to bNAb administration in humans. Being at the forefront of one of the first clinical trials studying an HIV-1-directed bNAb, we will have the unique opportunity to investigate the interplay of antibody therapy and the host immune system. This proposal aims to strongly advance the field of HIV-1 antibody therapy and therefore enable the introduction of a new therapeutic modality for HIV-1, and will gain insights for antibody-mediated therapy in other infectious diseases.
Summary
Antibodies are destined to neutralize pathogens and can prevent and fight infectious diseases. Over the last years, advances in single B cell cloning resulted in the isolation of highly potent and broad HIV-1 neutralizing antibodies (bNAbs) that have been shown to prevent SHIV infection in non-human primates (NHPs). Recently, we have demonstrated that a combination of bNAbs can suppress HIV-1 replication in humanized mice, reducing viremia to undetectable levels. Moreover, bNAb therapy of SHIV-infected NHPs induced a rapid decline in viremia, followed by a prolonged control due to the long half-life of the antibodies. While these results strongly encourage the clinical evaluation of bNAbs in HIV-1 therapy, it is of critical importance to understand how the therapeutic potential of antibodies can be harnessed in the most effective way. Therefore, we aim to: I.) Identify exceptionally potent HIV-1 neutralizing antibodies that will be a crucial component of immunotherapy. By establishing novel methods for single-cell sorting and high-throughput sequencing we want to identify bNAbs targeting novel epitopes. II.) Prevent HIV-1 escape applying rationally designed treatment strategies targeting conserved functional sites for HIV-1 entry. III.) Evaluate immune markers and function in relation to bNAb administration in humans. Being at the forefront of one of the first clinical trials studying an HIV-1-directed bNAb, we will have the unique opportunity to investigate the interplay of antibody therapy and the host immune system. This proposal aims to strongly advance the field of HIV-1 antibody therapy and therefore enable the introduction of a new therapeutic modality for HIV-1, and will gain insights for antibody-mediated therapy in other infectious diseases.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym IBDlipids
Project Lipid antigens in intestinal inflammation and tumor development
Researcher (PI) Sebastian Zeißig
Host Institution (HI) TECHNISCHE UNIVERSITAET DRESDEN
Call Details Starting Grant (StG), LS6, ERC-2013-StG
Summary Lipids play crucial roles in metabolism, immunity and cancer. In addition to their function as inflammatory mediators, lipids serve as antigens presented by CD1d and activate a subset of T cells called natural killer T (NKT) cells. While NKT cells are critical for human immunity, their uncontrolled activation contributes to inflammatory bowel disease (IBD), a group of diseases characterized by chronic intestinal inflammation and an increased risk of colorectal cancer (CRC). Specifically, NKT cells are the major source of pathogenic TH2 cytokines in the inflammatory bowel disease ulcerative colitis (UC), are sufficient to cause intestinal inflammation in mice, and are required for colitis and colitis-associated cancer in a mouse model of UC. These observations suggest that targeting of lipid antigen presentation may be of therapeutic value in IBD, where current therapies are of limited efficacy and aim at control rather than cure of disease.
Here, I propose to identify the lipid antigens responsible for NKT cell-mediated intestinal inflammation and colitis-associated cancer in human IBD and mouse models of intestinal inflammation and to develop therapeutic strategies for interference with pathogenic lipid antigen presentation. Specifically, I propose to characterize the intestinal inflammation- and cancer-associated CD1d lipidome based on novel in vitro and in vivo models of cleavable CD1d and a recently established lipidomics approach. Furthermore, I propose to develop strategies for inhibition of the generation, loading and presentation of inflammation- and cancer-associated lipid antigens. These studies combine biochemical, immunological and high-throughput technologies in an interdisciplinary manner to provide the knowledge required for the generation of novel, efficacious therapies for the treatment of IBD. These studies will have major implications for IBD and other inflammatory, infectious, and neoplastic diseases at mucosal barriers.
Summary
Lipids play crucial roles in metabolism, immunity and cancer. In addition to their function as inflammatory mediators, lipids serve as antigens presented by CD1d and activate a subset of T cells called natural killer T (NKT) cells. While NKT cells are critical for human immunity, their uncontrolled activation contributes to inflammatory bowel disease (IBD), a group of diseases characterized by chronic intestinal inflammation and an increased risk of colorectal cancer (CRC). Specifically, NKT cells are the major source of pathogenic TH2 cytokines in the inflammatory bowel disease ulcerative colitis (UC), are sufficient to cause intestinal inflammation in mice, and are required for colitis and colitis-associated cancer in a mouse model of UC. These observations suggest that targeting of lipid antigen presentation may be of therapeutic value in IBD, where current therapies are of limited efficacy and aim at control rather than cure of disease.
Here, I propose to identify the lipid antigens responsible for NKT cell-mediated intestinal inflammation and colitis-associated cancer in human IBD and mouse models of intestinal inflammation and to develop therapeutic strategies for interference with pathogenic lipid antigen presentation. Specifically, I propose to characterize the intestinal inflammation- and cancer-associated CD1d lipidome based on novel in vitro and in vivo models of cleavable CD1d and a recently established lipidomics approach. Furthermore, I propose to develop strategies for inhibition of the generation, loading and presentation of inflammation- and cancer-associated lipid antigens. These studies combine biochemical, immunological and high-throughput technologies in an interdisciplinary manner to provide the knowledge required for the generation of novel, efficacious therapies for the treatment of IBD. These studies will have major implications for IBD and other inflammatory, infectious, and neoplastic diseases at mucosal barriers.
Max ERC Funding
1 500 000 €
Duration
Start date: 2014-01-01, End date: 2018-12-31
Project acronym IL-22 AND IL-22BP
Project Identifying the immune and microbial network controlling the IL-22 – IL-22bp axis to open the doors for targeted therapies
Researcher (PI) Samuel Huber
Host Institution (HI) UNIVERSITAETSKLINIKUM HAMBURG-EPPENDORF
Call Details Starting Grant (StG), LS6, ERC-2013-StG
Summary Chronic mucosal inflammation and tissue damage predisposes patients to the development of colorectal cancer. One hypothesis is that the same factors important for wound healing, if left unchecked, also promote tumorigenesis. Tight control by a sensor of tissue damage should induce these factors to promote tissue repair, while limiting their activity to prevent development of cancer.
IL-22, a prototypical tissue repair factor, plays an important role in a wide variety of intestinal disease including infection, wound healing, colitis, and cancer. Indeed, IL-22 has protective and detrimental effects dependent on the milieu and disease suggesting that proper regulation is required. IL-22 expression is directly regulated, additionally a soluble IL-22 receptor (IL-22 binding protein; IL-22bp), can bind and neutralize IL-22. We reported recently that sensing of intestinal tissue damage and components of the microbiota via the NLRP3 or NLRP6 inflammasomes led to a down regulation of IL-22bp, thereby increasing bioavailability of IL-22. IL-22, which is induced during intestinal tissue damage, exerted protective properties during the peak of damage, but promoted tumor development if not controlled by IL-22bp during the recovery phase.
Accordingly a spatial and temporal regulation of IL-22 is crucial. Hence, global administration or blockade of IL-22 is unlikely to be therapeutically beneficial. We are using several newly generated conditional knock-out (cCasp1-/-, cIL-18R-/-, cIL-18-/-, cIL-22R1-/-), knock-in (IL-22 BFP), and gnotobiotic mice, aiming to analyze the cellular and microbial network regulating the IL-22 – IL-22bp axis at a resolution previously unfeasible. Our results will provide novel insights into the network between microflora, epithelium, and immune system regulating tissue regeneration and tumor development, and can lead to therapies for potentially a wide variety of intestinal diseases, such as infection, colon cancer, IBD, or wound healing.
Summary
Chronic mucosal inflammation and tissue damage predisposes patients to the development of colorectal cancer. One hypothesis is that the same factors important for wound healing, if left unchecked, also promote tumorigenesis. Tight control by a sensor of tissue damage should induce these factors to promote tissue repair, while limiting their activity to prevent development of cancer.
IL-22, a prototypical tissue repair factor, plays an important role in a wide variety of intestinal disease including infection, wound healing, colitis, and cancer. Indeed, IL-22 has protective and detrimental effects dependent on the milieu and disease suggesting that proper regulation is required. IL-22 expression is directly regulated, additionally a soluble IL-22 receptor (IL-22 binding protein; IL-22bp), can bind and neutralize IL-22. We reported recently that sensing of intestinal tissue damage and components of the microbiota via the NLRP3 or NLRP6 inflammasomes led to a down regulation of IL-22bp, thereby increasing bioavailability of IL-22. IL-22, which is induced during intestinal tissue damage, exerted protective properties during the peak of damage, but promoted tumor development if not controlled by IL-22bp during the recovery phase.
Accordingly a spatial and temporal regulation of IL-22 is crucial. Hence, global administration or blockade of IL-22 is unlikely to be therapeutically beneficial. We are using several newly generated conditional knock-out (cCasp1-/-, cIL-18R-/-, cIL-18-/-, cIL-22R1-/-), knock-in (IL-22 BFP), and gnotobiotic mice, aiming to analyze the cellular and microbial network regulating the IL-22 – IL-22bp axis at a resolution previously unfeasible. Our results will provide novel insights into the network between microflora, epithelium, and immune system regulating tissue regeneration and tumor development, and can lead to therapies for potentially a wide variety of intestinal diseases, such as infection, colon cancer, IBD, or wound healing.
Max ERC Funding
1 498 392 €
Duration
Start date: 2014-01-01, End date: 2018-12-31
Project acronym ImmProDynamics
Project Dissecting the interplay between the dynamics of immune responses and pathogen proliferation in vivo
Researcher (PI) Andreas J. Müller
Host Institution (HI) OTTO-VON-GUERICKE-UNIVERSITAET MAGDEBURG
Call Details Starting Grant (StG), LS6, ERC-2016-STG
Summary Pathogen proliferation has profound implications for its persistence, treatment strategies, and the induction and execution of protective immune responses. In vivo, pathogen proliferation rates are heterogenic, confronting the immune system with a variety of microbial physiological states. It is unknown if, and by what molecular mechanism, the immune response can distinguish these different states on a cellular level. Also, understanding the link between pathogen proliferation and immune cell dynamics could provide critical information on how infections can be controlled, and how to counteract pathogen persistence and antibiotic resistance. However, this question has never been addressed due to difficulties in studying the dynamics of immune cells and at the same time probing pathogen proliferation.
In this project, we will make use of a novel in vivo reporter system that I have developed, in order to determine the role of the pathogen's proliferation for its interaction with the immune system. Specifically, we will (1) determine the tissue niche in which the pathogen proliferates, (2) investigate the differential dynamics of phagocyte-pathogen- and of T cell-APC-interactions related to pathogen proliferation rate, (3) manipulate the relationship between pathogen proliferation and immune cell dynamics by using proliferation-deficient mutants and optogenetic pathogen inactivation, (4) identify signaling pathways that are differentially induced in cells infected by high versus low proliferating pathogens, and test their involvement in differential immune cell dynamics related to pathogen proliferation.
ImmProDynamics will for the first time provide insights into how cells of the immune system react to distinct pathogen proliferative states in vivo. This will greatly expand our knowledge of host-pathogen interactions, which will be critical for the design of efficient vaccines and antimicrobial therapy.
Summary
Pathogen proliferation has profound implications for its persistence, treatment strategies, and the induction and execution of protective immune responses. In vivo, pathogen proliferation rates are heterogenic, confronting the immune system with a variety of microbial physiological states. It is unknown if, and by what molecular mechanism, the immune response can distinguish these different states on a cellular level. Also, understanding the link between pathogen proliferation and immune cell dynamics could provide critical information on how infections can be controlled, and how to counteract pathogen persistence and antibiotic resistance. However, this question has never been addressed due to difficulties in studying the dynamics of immune cells and at the same time probing pathogen proliferation.
In this project, we will make use of a novel in vivo reporter system that I have developed, in order to determine the role of the pathogen's proliferation for its interaction with the immune system. Specifically, we will (1) determine the tissue niche in which the pathogen proliferates, (2) investigate the differential dynamics of phagocyte-pathogen- and of T cell-APC-interactions related to pathogen proliferation rate, (3) manipulate the relationship between pathogen proliferation and immune cell dynamics by using proliferation-deficient mutants and optogenetic pathogen inactivation, (4) identify signaling pathways that are differentially induced in cells infected by high versus low proliferating pathogens, and test their involvement in differential immune cell dynamics related to pathogen proliferation.
ImmProDynamics will for the first time provide insights into how cells of the immune system react to distinct pathogen proliferative states in vivo. This will greatly expand our knowledge of host-pathogen interactions, which will be critical for the design of efficient vaccines and antimicrobial therapy.
Max ERC Funding
1 499 525 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym IMMUNE CELL SWARMS
Project Innate Immune Cell Swarms: Integrating and Adapting Single Cell and Population Dynamics in Inflamed and Infected Tissues
Researcher (PI) Tim LÄMMERMANN
Host Institution (HI) MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN EV
Call Details Starting Grant (StG), LS6, ERC-2016-STG
Summary Neutrophils are essential effector cells of the innate immune response. Intravital microscopy studies have recently changed our perspective on neutrophil tissue dynamics. They revealed swarm-like migration patterns in several models of inflammation and infection: Neutrophil populations show strikingly coordinated behavior with phases of highly directed chemotaxis and clustering at local sites of tissue damage. My previous work established that neutrophils self-amplify this swarming response by auto-signaling, which provided the first molecular basis for the collective nature of neutrophil swarms (Lämmermann et al., Nature 2013). However, we are still at the beginning of unraveling the molecular pathways behind this newly discovered phenomenon.
Most importantly, we completely lack insight into the signals and mechanisms that stop neutrophil swarms in the resolution phase of an immune response. Since excess neutrophil accumulations cause deleterious tissue destruction in many inflammatory diseases, novel insights into the mechanisms, which prevent extensive swarm aggregation, might be of considerable therapeutic value. In accord with this, our proposal follows three aims: (i) dissecting the cellular and molecular mechanisms that control the resolution phase of neutrophil swarming, (ii) establishing a conceptual framework of how swarming immune cells adapt their dynamics to changing inflammatory milieus, and (iii) developing an integrated view on how neutrophil swarms are controlled by secondary waves of myeloid cell swarms. To achieve our goals, we will combine targeted mouse genetics with live cell imaging of immune cell dynamics in living tissues and the use of innovative mimics of physiological environments.
Our future findings on innate immune cell swarms promise to (i) advance our knowledge on leukocyte navigation in complex inflammatory tissues and (ii) provide new avenues for the therapeutic modulation of tissue regeneration after inflammation and infection.
Summary
Neutrophils are essential effector cells of the innate immune response. Intravital microscopy studies have recently changed our perspective on neutrophil tissue dynamics. They revealed swarm-like migration patterns in several models of inflammation and infection: Neutrophil populations show strikingly coordinated behavior with phases of highly directed chemotaxis and clustering at local sites of tissue damage. My previous work established that neutrophils self-amplify this swarming response by auto-signaling, which provided the first molecular basis for the collective nature of neutrophil swarms (Lämmermann et al., Nature 2013). However, we are still at the beginning of unraveling the molecular pathways behind this newly discovered phenomenon.
Most importantly, we completely lack insight into the signals and mechanisms that stop neutrophil swarms in the resolution phase of an immune response. Since excess neutrophil accumulations cause deleterious tissue destruction in many inflammatory diseases, novel insights into the mechanisms, which prevent extensive swarm aggregation, might be of considerable therapeutic value. In accord with this, our proposal follows three aims: (i) dissecting the cellular and molecular mechanisms that control the resolution phase of neutrophil swarming, (ii) establishing a conceptual framework of how swarming immune cells adapt their dynamics to changing inflammatory milieus, and (iii) developing an integrated view on how neutrophil swarms are controlled by secondary waves of myeloid cell swarms. To achieve our goals, we will combine targeted mouse genetics with live cell imaging of immune cell dynamics in living tissues and the use of innovative mimics of physiological environments.
Our future findings on innate immune cell swarms promise to (i) advance our knowledge on leukocyte navigation in complex inflammatory tissues and (ii) provide new avenues for the therapeutic modulation of tissue regeneration after inflammation and infection.
Max ERC Funding
1 500 000 €
Duration
Start date: 2017-02-01, End date: 2022-01-31
Project acronym IMMUNOSWITCH
Project Switch recombination: a model system for DNA editing and repair in human lymphocytes with relevance for primary immunodeficiency and cancer formation
Researcher (PI) Qiang Pan Hammarström
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Starting Grant (StG), LS6, ERC-2009-StG
Summary The aim of this project is to try to understand the complex molecular mechanisms involved in DNA editing, repair and recombination during immunoglobulin class switch recombination (CSR) and somatic hypermutation (SHM). We have developed a series of PCR-based assays to study in vivo generated CSR junctions and the pattern of mutations introduced in the immunoglobulin variable region genes in human B cells, allowing us to characterize CSR and SHM in patients with immunodeficiency due to defect(s) in DNA repair/recombination. Novel in vitro CSR assays, based on GFP expression, allowing quantitative measurement of substrate recombination, are also being developed. In addition, we have initiated an evolutionary analysis of the function and structure of activation-induced deaminase, an essential molecule involved both in CSR and SHM, aiming to identify CSR specific-cofactor(s). Combining these approaches, we will be able to define the DNA repair pathways involved in CSR and SHM. The suggested project requires access to patients with various defects in the DNA repair pathways. Many of these diseases are exceedingly rare. However, through worldwide collaboration, we have obtained samples from a majority of the diagnosed patients. We are also refining the existing screening methods and developing novel methods, that will allow identification of additional patients both with recognized and new diseases caused by mutations in DNA repair pathways. Finally, we hope to be able to address the question whether illegitimate CSR events are associated with predisposition to lymphomagenesis in patients with immunodeficiency/DNA repair defect(s), by analyzing the CSR induced chromosomal breaks and translocations in these patients. A large-scale sequencing project is also planned to characterize the CSRnome in B-cell lymphoma samples.
Summary
The aim of this project is to try to understand the complex molecular mechanisms involved in DNA editing, repair and recombination during immunoglobulin class switch recombination (CSR) and somatic hypermutation (SHM). We have developed a series of PCR-based assays to study in vivo generated CSR junctions and the pattern of mutations introduced in the immunoglobulin variable region genes in human B cells, allowing us to characterize CSR and SHM in patients with immunodeficiency due to defect(s) in DNA repair/recombination. Novel in vitro CSR assays, based on GFP expression, allowing quantitative measurement of substrate recombination, are also being developed. In addition, we have initiated an evolutionary analysis of the function and structure of activation-induced deaminase, an essential molecule involved both in CSR and SHM, aiming to identify CSR specific-cofactor(s). Combining these approaches, we will be able to define the DNA repair pathways involved in CSR and SHM. The suggested project requires access to patients with various defects in the DNA repair pathways. Many of these diseases are exceedingly rare. However, through worldwide collaboration, we have obtained samples from a majority of the diagnosed patients. We are also refining the existing screening methods and developing novel methods, that will allow identification of additional patients both with recognized and new diseases caused by mutations in DNA repair pathways. Finally, we hope to be able to address the question whether illegitimate CSR events are associated with predisposition to lymphomagenesis in patients with immunodeficiency/DNA repair defect(s), by analyzing the CSR induced chromosomal breaks and translocations in these patients. A large-scale sequencing project is also planned to characterize the CSRnome in B-cell lymphoma samples.
Max ERC Funding
1 888 166 €
Duration
Start date: 2009-12-01, End date: 2014-11-30
Project acronym INTEGHER
Project Integration of herpesvirus into telomeres: From the mechanism of genome integration and mobilization to therapeutic intervention
Researcher (PI) Benedikt Kaufer
Host Institution (HI) FREIE UNIVERSITAET BERLIN
Call Details Starting Grant (StG), LS6, ERC-2015-STG
Summary Herpesviruses cause serious diseases in humans and animals. After initial lytic infection, herpesviruses establish a quiescent (latent) infection, which allows their persistence in the host for life. We and others recently identified a novel mechanism that allows maintenance of the genome of certain herpesviruses during latency, by integrating their complete genetic material into host telomeres. One of these viruses is human herpesvirus 6 (HHV-6) which is associated with seizures, encephalitis, and graft rejection in transplant patients. Sporadic reactivation of the integrated virus ensures continued evolution of the virus as it spreads to a new cadre of susceptible individuals. There are critical gaps in our knowledge regarding the fate of herpesvirus genomes during integration and reactivation as well as of viral and cellular factors involved in these processes.
INTEGHER will make use of novel technologies to close these gaps and to devise new therapeutic approaches. Specifically, we will 1) determine the fate of the HHV-6 genome during latency by developing a novel reporter system that allows live-cell imaging of the virus genome in living cells and elucidate epigenetic changes of the HHV-6 genome during integration and reactivation; 2) identify viral and cellular factors that drive virus genome integration and reactivation, using recombinant viruses, drugs and CRISPR/Cas9 genome engineering 3) employ genome-editing tools to eliminate the virus genome integrated in host chromosomes in vitro and in an in vivo model. The proposal utilizes state-of-the-art technologies and pioneers new approaches, particularly with regard to visualization and excision of virus genomes in latently infected cells that are also present in (bone marrow) transplants. Altogether, these studies will define the mechanism of herpesvirus integration and reactivation and will provide new tools for therapeutic excision of virus genomes from living cells.
Summary
Herpesviruses cause serious diseases in humans and animals. After initial lytic infection, herpesviruses establish a quiescent (latent) infection, which allows their persistence in the host for life. We and others recently identified a novel mechanism that allows maintenance of the genome of certain herpesviruses during latency, by integrating their complete genetic material into host telomeres. One of these viruses is human herpesvirus 6 (HHV-6) which is associated with seizures, encephalitis, and graft rejection in transplant patients. Sporadic reactivation of the integrated virus ensures continued evolution of the virus as it spreads to a new cadre of susceptible individuals. There are critical gaps in our knowledge regarding the fate of herpesvirus genomes during integration and reactivation as well as of viral and cellular factors involved in these processes.
INTEGHER will make use of novel technologies to close these gaps and to devise new therapeutic approaches. Specifically, we will 1) determine the fate of the HHV-6 genome during latency by developing a novel reporter system that allows live-cell imaging of the virus genome in living cells and elucidate epigenetic changes of the HHV-6 genome during integration and reactivation; 2) identify viral and cellular factors that drive virus genome integration and reactivation, using recombinant viruses, drugs and CRISPR/Cas9 genome engineering 3) employ genome-editing tools to eliminate the virus genome integrated in host chromosomes in vitro and in an in vivo model. The proposal utilizes state-of-the-art technologies and pioneers new approaches, particularly with regard to visualization and excision of virus genomes in latently infected cells that are also present in (bone marrow) transplants. Altogether, these studies will define the mechanism of herpesvirus integration and reactivation and will provide new tools for therapeutic excision of virus genomes from living cells.
Max ERC Funding
1 810 747 €
Duration
Start date: 2016-04-01, End date: 2021-03-31
Project acronym IVIP
Project Impact of virus infection on the host proteome
Researcher (PI) Andreas Pichlmair
Host Institution (HI) MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN EV
Call Details Starting Grant (StG), LS6, ERC-2012-StG_20111109
Summary Pathogens exploit cellular resources for their own benefit and their propagation. Co-evolution of viruses and their hosts led to the establishment of very specific interactions between both partners. The balance between both, the viral attack and the cellular defense mechanisms, dictates the outcome of an infection, either leading to a cure, to disease or life-long co-existence. Efforts from many laboratories have focused on virus-host interactions, mostly by testing isolated protein-protein interactions or by employing transcriptome analysis. Although this undoubtedly has been highly valuable to our knowledge on virus-host interactions, it is also clear that most virus-host interactions do not occur on transcriptome level and neither involve single protein-protein interactions. Here I propose to systematically test the impact of virus infection on the host proteome (iViP) by using the newest generation of mass spectrometry-based discovery tools combined with infection biology. iViP divides in three interconnected parts that individually and collectively warrant success: Part I evaluates changes in the abundance of the proteins after virus infection and correlates this to mRNA levels, determined by RNA sequencing. Part II identifies proteome-wide post-translational modifications that would be indicative for an involvement in infection biology. Part III dissects the role of virus stimulated/altered proteins in the antiviral protein-protein interaction network. Lastly, identified proteins will be validated in a two –step procedure involving a large-scale validation strategy and further focusing on few selected interactors. A comprehensive coherent data set describing and functionally explaining the cellular changes after encounter of a variety of viral pathogens on a proteome level would complement or even succeed currently available data sets and become invaluable to basic and translational research of the future.
Summary
Pathogens exploit cellular resources for their own benefit and their propagation. Co-evolution of viruses and their hosts led to the establishment of very specific interactions between both partners. The balance between both, the viral attack and the cellular defense mechanisms, dictates the outcome of an infection, either leading to a cure, to disease or life-long co-existence. Efforts from many laboratories have focused on virus-host interactions, mostly by testing isolated protein-protein interactions or by employing transcriptome analysis. Although this undoubtedly has been highly valuable to our knowledge on virus-host interactions, it is also clear that most virus-host interactions do not occur on transcriptome level and neither involve single protein-protein interactions. Here I propose to systematically test the impact of virus infection on the host proteome (iViP) by using the newest generation of mass spectrometry-based discovery tools combined with infection biology. iViP divides in three interconnected parts that individually and collectively warrant success: Part I evaluates changes in the abundance of the proteins after virus infection and correlates this to mRNA levels, determined by RNA sequencing. Part II identifies proteome-wide post-translational modifications that would be indicative for an involvement in infection biology. Part III dissects the role of virus stimulated/altered proteins in the antiviral protein-protein interaction network. Lastly, identified proteins will be validated in a two –step procedure involving a large-scale validation strategy and further focusing on few selected interactors. A comprehensive coherent data set describing and functionally explaining the cellular changes after encounter of a variety of viral pathogens on a proteome level would complement or even succeed currently available data sets and become invaluable to basic and translational research of the future.
Max ERC Funding
1 498 896 €
Duration
Start date: 2012-11-01, End date: 2017-10-31
Project acronym LUEDDE-MED3-AACHEN
Project The Function of inflammatory signalling pathways in acute and chronic liver disease and liver cancer
Researcher (PI) Tom Luedde
Host Institution (HI) UNIVERSITAETSKLINIKUM AACHEN
Call Details Starting Grant (StG), LS6, ERC-2007-StG
Summary The aim of this proposal is to examine the role of inflammatory signalling pathways in murine models of liver and biliary disease by application of conditional gene targeting using cre/loxP technology. Previous studies have provided evidence that the NF-kB pathway and its activating kinase complex – consisting of three subunits: IKK1, IKK2 and NEMO – are crucial regulators of liver physiology and pathology, but their differential, cell specific functions in the liver are currently only poorly understood. The first part of this proposal will focus on the evaluation of molecular mechanisms underlying the development of hepatocellular carcinoma in a setting of chronic hepatitis. By using a novel mouse model of spontaneous liver cancer based on conditional deletion of NEMO in hepatocytes, the functions of cytokines, specific intracellular signalling pathways, the innate and adaptive immune system and the role of hepatic stem cells in hepatitis and carcinogenesis will be examined. In the second part of this proposal, we will extend these studies by evaluating the function of NEMO/NF-kB in other hepatic cell compartments, specifically the function of NEMO in hepatic stellate cells and liver fibrosis, the endothelial function of NEMO/NF-kB in an in vivo model of hepatic ischemia-reperfusion injury and the role of the NF-kB pathway in biliary epithelial cells and inflammatory biliary diseases. Finally, in the third part of this proposal we will analyse the unknown intrahepatic role of non-canonical, IKK1-dependent signalling pathways and the function of TAK1 – a molecule at the interface between inflammatory and developmental pathways – in liver injury, fatty-liver-disease and insulin-resistance. Knowledge gained by these studies and the further understanding of the cell specific hepatic function of NF-kB and related pathways might build the basis for the development of novel pharmacological approaches for the future treatment of liver diseases and cancer in humans.
Summary
The aim of this proposal is to examine the role of inflammatory signalling pathways in murine models of liver and biliary disease by application of conditional gene targeting using cre/loxP technology. Previous studies have provided evidence that the NF-kB pathway and its activating kinase complex – consisting of three subunits: IKK1, IKK2 and NEMO – are crucial regulators of liver physiology and pathology, but their differential, cell specific functions in the liver are currently only poorly understood. The first part of this proposal will focus on the evaluation of molecular mechanisms underlying the development of hepatocellular carcinoma in a setting of chronic hepatitis. By using a novel mouse model of spontaneous liver cancer based on conditional deletion of NEMO in hepatocytes, the functions of cytokines, specific intracellular signalling pathways, the innate and adaptive immune system and the role of hepatic stem cells in hepatitis and carcinogenesis will be examined. In the second part of this proposal, we will extend these studies by evaluating the function of NEMO/NF-kB in other hepatic cell compartments, specifically the function of NEMO in hepatic stellate cells and liver fibrosis, the endothelial function of NEMO/NF-kB in an in vivo model of hepatic ischemia-reperfusion injury and the role of the NF-kB pathway in biliary epithelial cells and inflammatory biliary diseases. Finally, in the third part of this proposal we will analyse the unknown intrahepatic role of non-canonical, IKK1-dependent signalling pathways and the function of TAK1 – a molecule at the interface between inflammatory and developmental pathways – in liver injury, fatty-liver-disease and insulin-resistance. Knowledge gained by these studies and the further understanding of the cell specific hepatic function of NF-kB and related pathways might build the basis for the development of novel pharmacological approaches for the future treatment of liver diseases and cancer in humans.
Max ERC Funding
1 600 356 €
Duration
Start date: 2008-09-01, End date: 2014-08-31
Project acronym NUTRIMMUNE
Project NutrImmune: Nutrient-controlled molecular pathways instructing development and function of mucosa-associated innate lymphocytes
Researcher (PI) Andreas Diefenbach
Host Institution (HI) CHARITE - UNIVERSITAETSMEDIZIN BERLIN
Call Details Starting Grant (StG), LS6, ERC-2012-StG_20111109
Summary The last decade has witnessed an explosion of research into the molecular networks ensuring maintenance of a mutualistic relationship between microbes and host cell networks at mucosal surfaces. Failure of such homeostatic or adaptive programs lead to susceptibility to intestinal infections or to chronic inflammation causing debilitating human diseases such as inflammatory bowel diseases or inflammation-induced intestinal cancer. In contrast to the role of the microbiota and its composition, the role of nutrients for development and function of the intestinal immune system has been a matter of speculation owing to the fact that molecular sensors of dietary molecules were widely unknown. Given the broad impact of nutrients on metabolic diseases and human health, research into the question of how the power of nutrients can be harnessed for improving human health and for the prevention of disease is much warranted. We have recently found that the aryl hydrocarbon receptor (AhR) is required for the development and function of an innate lymphocyte subset (RORγt+ ILC) that protects against intestinal infections and inflammatory bowel disease (Kiss, Science 2011). AhR serves as a ligand-inducible transcription factor sensing plant-derived phytochemicals and directly controls expression of genes required for the maintenance of RORγt+ ILC. The data established the first molecular link between diets and the development of immune system components. Here, we will test our central hypotheses that (1) diets adapt the function of the intestinal immune system by controlling the pool size of innate lymphocytes, and that (2) RORγt+ ILC directly control epithelial homeostasis and adaptation by regulating niche programs that control intestinal stem cell population dynamics. These aims link nutrient-controlled function of innate lymphocytes to the processes regulating organ homeostasis and may reveal new potential therapeutic strategies for intestinal diseases and cancer.
Summary
The last decade has witnessed an explosion of research into the molecular networks ensuring maintenance of a mutualistic relationship between microbes and host cell networks at mucosal surfaces. Failure of such homeostatic or adaptive programs lead to susceptibility to intestinal infections or to chronic inflammation causing debilitating human diseases such as inflammatory bowel diseases or inflammation-induced intestinal cancer. In contrast to the role of the microbiota and its composition, the role of nutrients for development and function of the intestinal immune system has been a matter of speculation owing to the fact that molecular sensors of dietary molecules were widely unknown. Given the broad impact of nutrients on metabolic diseases and human health, research into the question of how the power of nutrients can be harnessed for improving human health and for the prevention of disease is much warranted. We have recently found that the aryl hydrocarbon receptor (AhR) is required for the development and function of an innate lymphocyte subset (RORγt+ ILC) that protects against intestinal infections and inflammatory bowel disease (Kiss, Science 2011). AhR serves as a ligand-inducible transcription factor sensing plant-derived phytochemicals and directly controls expression of genes required for the maintenance of RORγt+ ILC. The data established the first molecular link between diets and the development of immune system components. Here, we will test our central hypotheses that (1) diets adapt the function of the intestinal immune system by controlling the pool size of innate lymphocytes, and that (2) RORγt+ ILC directly control epithelial homeostasis and adaptation by regulating niche programs that control intestinal stem cell population dynamics. These aims link nutrient-controlled function of innate lymphocytes to the processes regulating organ homeostasis and may reveal new potential therapeutic strategies for intestinal diseases and cancer.
Max ERC Funding
1 499 760 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym PARAMOTSIG
Project Receptor signalling mediating malaria parasite motility
Researcher (PI) Friedrich Frischknecht
Host Institution (HI) UNIVERSITATSKLINIKUM HEIDELBERG
Call Details Starting Grant (StG), LS6, ERC-2011-StG_20101109
Summary Plasmodium sporozoites are the motile forms of the malaria parasite injected into the host by a mosquito. Sporozoite motility is essential for tissue penetration as well as host cell invasion and thus pathogenesis suggesting that blocking motility could potentially add a new way in controlling malaria. It is dependent on a parasite specific myosin, a highly divergent actin and plasma membrane proteins, adhesins that link the substrate to the actomyosin motor. We want to understand the molecular and biophysical basis that underlies the motility of sporozoites to eventually be able to block it. Consequently, we developed methods that allow a systematic probing of key variables important in motility in order to reveal the basic mechanisms of sporozoite locomotion and to screen for small molecules that inhibit motility. Using these assays, we made a number of groundbreaking observations on the cellular and molecular level that gave new insights into the mechanisms of sporozoite adhesion and motility. For example, the dynamic, actin-dependent turnover of adhesion sites was found to be a key factor in sporozoite motility. It is our ultimate goal to understand sporozoite motility to a degree that we can provide a comprehensive dynamic model of sporozoite movement. With the current proposal we aim at unravelling the initial molecular events leading to sporozoite motility focussing on three different adhesins that are known or suspected to be involved in motility. We hypothesize that outside-in signalling leading to actin rearrangements originates from the formation of homo- or heterodimers between these adhesins. Additionally we suggest that inside-out signalling contributes to modulation of adhesion strengths mediated by these adhesins. To test these hypotheses we will generate recombinant parasites that lack two adhesins or express fluorescently tagged adhesin fusions, chimeric or mutant adhesins and investigate these with our recently developed toolbox of novel assays.
Summary
Plasmodium sporozoites are the motile forms of the malaria parasite injected into the host by a mosquito. Sporozoite motility is essential for tissue penetration as well as host cell invasion and thus pathogenesis suggesting that blocking motility could potentially add a new way in controlling malaria. It is dependent on a parasite specific myosin, a highly divergent actin and plasma membrane proteins, adhesins that link the substrate to the actomyosin motor. We want to understand the molecular and biophysical basis that underlies the motility of sporozoites to eventually be able to block it. Consequently, we developed methods that allow a systematic probing of key variables important in motility in order to reveal the basic mechanisms of sporozoite locomotion and to screen for small molecules that inhibit motility. Using these assays, we made a number of groundbreaking observations on the cellular and molecular level that gave new insights into the mechanisms of sporozoite adhesion and motility. For example, the dynamic, actin-dependent turnover of adhesion sites was found to be a key factor in sporozoite motility. It is our ultimate goal to understand sporozoite motility to a degree that we can provide a comprehensive dynamic model of sporozoite movement. With the current proposal we aim at unravelling the initial molecular events leading to sporozoite motility focussing on three different adhesins that are known or suspected to be involved in motility. We hypothesize that outside-in signalling leading to actin rearrangements originates from the formation of homo- or heterodimers between these adhesins. Additionally we suggest that inside-out signalling contributes to modulation of adhesion strengths mediated by these adhesins. To test these hypotheses we will generate recombinant parasites that lack two adhesins or express fluorescently tagged adhesin fusions, chimeric or mutant adhesins and investigate these with our recently developed toolbox of novel assays.
Max ERC Funding
1 453 800 €
Duration
Start date: 2012-04-01, End date: 2017-03-31
Project acronym PAS
Project Persistence of allergic sensitization
Researcher (PI) David Voehringer
Host Institution (HI) UNIVERSITATSKLINIKUM ERLANGEN
Call Details Starting Grant (StG), LS6, ERC-2009-StG
Summary Allergic disorders are dramatically increasing in the western world over the past several years. Allergies are inappropriate immune responses directed against harmless environmental antigens. Upon primary allergen encounter B cells are induced to differentiate into IgE producing plasma cells. Mast cells and basophils are then sensitized by binding of allergen-specific IgE molecules to the high-affinity IgE receptor on the cell surface. A second allergen exposure causes cross-linking of IgE molecules on mast cells and basophils which results in degranulation of pro-inflammatory mediators. Allergen-specific IgE can be detected many months after the sensitization phase despite the fact that IgE has a very short half-life. This suggests that small amounts of allergen-specific IgE are constantly secreted by long-lived IgE producing plasma cells. The development, turnover and fate of IgE producing cells in vivo are largely unknown. Therefore, we propose to study these important issues by using genetically modified mouse strains and cutting edge technology. We will use IgE-FLAG-GFP reporter mice to trace IgE-producing cells in vivo by fluorescence microscopy and 2-photon live imaging. The reporter mice will allow us to isolate IgE-producing cells so that we can determine their gene expression profile. Furthermore, we will determine the turnover and lifespan of IgE producing cells in vivo by BrdU incorporation. Finally, we will generate an IgE-Cre knock-in mouse to specifically delete conditional alleles in IgE-producing cells. Taken together, these important experiments will help us to better understand the biology of IgE-producing cells and may result in development of novel therapeutic strategies.
Summary
Allergic disorders are dramatically increasing in the western world over the past several years. Allergies are inappropriate immune responses directed against harmless environmental antigens. Upon primary allergen encounter B cells are induced to differentiate into IgE producing plasma cells. Mast cells and basophils are then sensitized by binding of allergen-specific IgE molecules to the high-affinity IgE receptor on the cell surface. A second allergen exposure causes cross-linking of IgE molecules on mast cells and basophils which results in degranulation of pro-inflammatory mediators. Allergen-specific IgE can be detected many months after the sensitization phase despite the fact that IgE has a very short half-life. This suggests that small amounts of allergen-specific IgE are constantly secreted by long-lived IgE producing plasma cells. The development, turnover and fate of IgE producing cells in vivo are largely unknown. Therefore, we propose to study these important issues by using genetically modified mouse strains and cutting edge technology. We will use IgE-FLAG-GFP reporter mice to trace IgE-producing cells in vivo by fluorescence microscopy and 2-photon live imaging. The reporter mice will allow us to isolate IgE-producing cells so that we can determine their gene expression profile. Furthermore, we will determine the turnover and lifespan of IgE producing cells in vivo by BrdU incorporation. Finally, we will generate an IgE-Cre knock-in mouse to specifically delete conditional alleles in IgE-producing cells. Taken together, these important experiments will help us to better understand the biology of IgE-producing cells and may result in development of novel therapeutic strategies.
Max ERC Funding
1 727 981 €
Duration
Start date: 2009-12-01, End date: 2014-11-30
Project acronym Plat-IL-1
Project Pathophysiology of platelet-derived Interleukin 1
Researcher (PI) BERNARDO SIMOES FRANKLIN
Host Institution (HI) UNIVERSITAETSKLINIKUM BONN
Call Details Starting Grant (StG), LS6, ERC-2016-STG
Summary The Interleukin (IL)-1 family of pro-inflammatory cytokines are among the most potent pyrogens, and their excessive production can cause several auto-inflammatory syndromes. Additionally, overabundance of IL-1 cytokines can trigger, or contribute to a range of inflammatory and metabolic disorders. The expression of the key members of the IL-1 family, such as IL-1β and IL-18, is regulated at both the transcriptional and post-transcriptional levels. IL-1β and IL-18, are produced as inactive precursors, which require activation of caspase-1 by the inflammasomes for their maturation and release by from cells, occasionally at the cost of caspase-1 mediated-cell death. We have recently discovered that inflammasomes are released into the extracellular space where they remain active after the demise of activated cells, and that extracellular inflammasomes can amplify inflammation by sustaining extracellular production of IL-1β. However, the sources of extracellular pro-IL-1β are not known. Recent advances in platelet proteomics have revealed that these non-nucleated cells are able to produce their own cytokines, including soluble IL-1β and membrane-bound IL-1α, and are able to significantly magnify IL-1 production by immune cells. As platelets outnumber leukocytes by several folds, they could potentially be the major source of extracellular inflammasomes in the body, or be a major producer of IL-1 precursors that are cleaved by extracellular inflammasomes released from dying immune cells. In this proposal, we will investigate the mechanism(s) by which platelets produce IL-1, and the specific contribution of platelet-derived IL-1 to sterile inflammation, or host resistance to bacterial and viral infection. We believe that a deeper understanding of platelet-IL-1 and their interaction with immune cells during sterile inflammation, or infection might help to uncover new targets for immune-therapies.
Summary
The Interleukin (IL)-1 family of pro-inflammatory cytokines are among the most potent pyrogens, and their excessive production can cause several auto-inflammatory syndromes. Additionally, overabundance of IL-1 cytokines can trigger, or contribute to a range of inflammatory and metabolic disorders. The expression of the key members of the IL-1 family, such as IL-1β and IL-18, is regulated at both the transcriptional and post-transcriptional levels. IL-1β and IL-18, are produced as inactive precursors, which require activation of caspase-1 by the inflammasomes for their maturation and release by from cells, occasionally at the cost of caspase-1 mediated-cell death. We have recently discovered that inflammasomes are released into the extracellular space where they remain active after the demise of activated cells, and that extracellular inflammasomes can amplify inflammation by sustaining extracellular production of IL-1β. However, the sources of extracellular pro-IL-1β are not known. Recent advances in platelet proteomics have revealed that these non-nucleated cells are able to produce their own cytokines, including soluble IL-1β and membrane-bound IL-1α, and are able to significantly magnify IL-1 production by immune cells. As platelets outnumber leukocytes by several folds, they could potentially be the major source of extracellular inflammasomes in the body, or be a major producer of IL-1 precursors that are cleaved by extracellular inflammasomes released from dying immune cells. In this proposal, we will investigate the mechanism(s) by which platelets produce IL-1, and the specific contribution of platelet-derived IL-1 to sterile inflammation, or host resistance to bacterial and viral infection. We believe that a deeper understanding of platelet-IL-1 and their interaction with immune cells during sterile inflammation, or infection might help to uncover new targets for immune-therapies.
Max ERC Funding
1 488 854 €
Duration
Start date: 2017-03-01, End date: 2022-02-28
Project acronym PROTECTC
Project Identify novel pathways to enhance the induction of protective CD8+ T cell responses
Researcher (PI) Dietmar Zehn
Host Institution (HI) TECHNISCHE UNIVERSITAET MUENCHEN
Call Details Starting Grant (StG), LS6, ERC-2013-StG
Summary There is an urgent need for progress in developing prophylactic and therapeutic vaccination strategies that induce polyfunctional, strongly protective cytotoxic CD8 T cell responses. These could shield us from pathogens against which the presently available, neutralizing antibody-inducing vaccine approaches confer limited or no protection and they could be used to eliminate tumors or chronic infections. In contrast to this need, we currently fail to induce effector and memory CD8 T cells in numbers high enough to effectively impact an infection or the growth of tumors. As protective CD8 T cell responses are readily generated during several viral infections, we need to improve our insight into how pathogen protection is naturally achieved, identify why immune protection sometimes fails, and use this knowledge to develop novel vaccine strategies. We will use a well balanced approach of hypothesis stimulated and unbiased multisystem observations to exploit novel mechanisms and to find ways to augment the CD8 T cell response to a vaccine. We have established model systems that are uniquely suited to extract and test molecules that determine T cell differentiation and expansion magnitude. Along with that we aim to enhance our insight of immune responses in vaccinated individuals to prevent creating situation in which vaccines fail to confer protection or may cause adverse effects. We recently made very unexpected observations that challenge our current concept of T cell differentiation in chronic infections, which proposes that T cells terminally differentiate and become senescent. We therefore aim to redefine our understanding of T cell responses in such infections. This will also be pursued to unravel novel strategies to reactivate T cells in persisting infections. Overall, the project will strongly further our insight into CD8 T cell responses during infections and will support the development of more effective vaccine strategies to induce antigen-specific CD8 T cells
Summary
There is an urgent need for progress in developing prophylactic and therapeutic vaccination strategies that induce polyfunctional, strongly protective cytotoxic CD8 T cell responses. These could shield us from pathogens against which the presently available, neutralizing antibody-inducing vaccine approaches confer limited or no protection and they could be used to eliminate tumors or chronic infections. In contrast to this need, we currently fail to induce effector and memory CD8 T cells in numbers high enough to effectively impact an infection or the growth of tumors. As protective CD8 T cell responses are readily generated during several viral infections, we need to improve our insight into how pathogen protection is naturally achieved, identify why immune protection sometimes fails, and use this knowledge to develop novel vaccine strategies. We will use a well balanced approach of hypothesis stimulated and unbiased multisystem observations to exploit novel mechanisms and to find ways to augment the CD8 T cell response to a vaccine. We have established model systems that are uniquely suited to extract and test molecules that determine T cell differentiation and expansion magnitude. Along with that we aim to enhance our insight of immune responses in vaccinated individuals to prevent creating situation in which vaccines fail to confer protection or may cause adverse effects. We recently made very unexpected observations that challenge our current concept of T cell differentiation in chronic infections, which proposes that T cells terminally differentiate and become senescent. We therefore aim to redefine our understanding of T cell responses in such infections. This will also be pursued to unravel novel strategies to reactivate T cells in persisting infections. Overall, the project will strongly further our insight into CD8 T cell responses during infections and will support the development of more effective vaccine strategies to induce antigen-specific CD8 T cells
Max ERC Funding
1 499 850 €
Duration
Start date: 2013-10-01, End date: 2018-09-30
Project acronym PyraSig
Project Pyrazine Signalling in Commensal and Pathogenic Bacteria
Researcher (PI) Kai PAPENFORT
Host Institution (HI) LUDWIG-MAXIMILIANS-UNIVERSITAET MUENCHEN
Call Details Starting Grant (StG), LS6, ERC-2017-STG
Summary Quorum sensing (QS) is a bacterial cell–cell communication process involving the production, release, and detection of extracellular signal molecules called autoinducers. QS is key to all microbiology as it enables otherwise solitary bacteria to coordinate complex cooperative tasks such as biofilm formation and pathogenesis. Consequently, targeting QS is a promising new concept for antimicrobial therapy. However, for this concept to become reality, we must first identify QS systems in pathogenic bacteria, discover the relevant autoinducers and study the underlying regulatory principles.
I recently identified a new QS pathway in Vibrio cholerae, the causative agent of cholera disease. The autoinducer of the system is DPO (3,5-dimethylpyrazin-2-ol), a new molecule to biology and the first pyrazine involved in QS. DPO production is widespread among microbes including pathogenic and commensal bacteria. V. cholerae synthesizes DPO from host mucins and our preliminary data show that DPO controls collective phenotypes, such as biofilm formation and toxin production in this major human pathogen. I therefore hypothesize that DPO connects virulence, QS and communication with the host microbiota in V. cholerae and related bacteria.
The overarching goal of this project is to understand the roles of DPO in host-microbe interaction and collective behaviours. To this end, we will pursue three key research goals. First, we will study the molecular parameters underlying DPO-signalling and probe the global effects of DPO on gene expression. Second, we will focus on the role of DPO in virulence of V. cholerae and other pathogens. Third, we will probe the effect of DPO on microbial behaviours, such as swarming and biofilm formation. This combined work will provide a comprehensive model for DPO-signalling in bacteria, which will not only advance the fundamental understanding of QS-based communication strategies, but might also provide the framework for QS-inspired anti-infectives.
Summary
Quorum sensing (QS) is a bacterial cell–cell communication process involving the production, release, and detection of extracellular signal molecules called autoinducers. QS is key to all microbiology as it enables otherwise solitary bacteria to coordinate complex cooperative tasks such as biofilm formation and pathogenesis. Consequently, targeting QS is a promising new concept for antimicrobial therapy. However, for this concept to become reality, we must first identify QS systems in pathogenic bacteria, discover the relevant autoinducers and study the underlying regulatory principles.
I recently identified a new QS pathway in Vibrio cholerae, the causative agent of cholera disease. The autoinducer of the system is DPO (3,5-dimethylpyrazin-2-ol), a new molecule to biology and the first pyrazine involved in QS. DPO production is widespread among microbes including pathogenic and commensal bacteria. V. cholerae synthesizes DPO from host mucins and our preliminary data show that DPO controls collective phenotypes, such as biofilm formation and toxin production in this major human pathogen. I therefore hypothesize that DPO connects virulence, QS and communication with the host microbiota in V. cholerae and related bacteria.
The overarching goal of this project is to understand the roles of DPO in host-microbe interaction and collective behaviours. To this end, we will pursue three key research goals. First, we will study the molecular parameters underlying DPO-signalling and probe the global effects of DPO on gene expression. Second, we will focus on the role of DPO in virulence of V. cholerae and other pathogens. Third, we will probe the effect of DPO on microbial behaviours, such as swarming and biofilm formation. This combined work will provide a comprehensive model for DPO-signalling in bacteria, which will not only advance the fundamental understanding of QS-based communication strategies, but might also provide the framework for QS-inspired anti-infectives.
Max ERC Funding
1 499 250 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym RC3H1/2-SPECIFICITY
Project Specificity of Rc3h1/2 proteins in post-transcriptional control of immunity and autoimmune disease
Researcher (PI) Vigo Heissmeyer
Host Institution (HI) HELMHOLTZ ZENTRUM MUENCHEN DEUTSCHES FORSCHUNGSZENTRUM FUER GESUNDHEIT UND UMWELT GMBH
Call Details Starting Grant (StG), LS6, ERC-2011-StG_20101109
Summary We are constantly protected by our adaptive immune system. Its functioning requires precise control of gene expression in lymphocytes, since deregulation can cause autoimmune diseases (affecting ~5% of our population) as well as allergic reactions (~9-16%, with increasing incidence). Post-transcriptional control of gene expression is crucial in many immune decisions, however the determinants of specificity in this type of regulation are less well defined. The recently described Rc3h1 or Roquin protein prevents the development of autoimmune disease in mice. Rc3h1 destabilizes the mRNA of the inducible costimulator (ICOS), a co-receptor on T cells. ICOS is critical in the germinal center reaction in which T cell help selects B cells making high affinity antibodies. However, the molecular interactions of this posttranscriptional regulation and the pathways that specify such repressor/target relations are unsolved, and they are the focus of my work in this proposal.
Rc3h1 is an essential factor of peripheral T cell tolerance, whereas the role of its paralog Rc3h2 has not been addressed. We have recently shown that Rc3h1 is an RNA binding-protein that recognizes the 3’UTR of ICOS mRNA. Our preliminary data suggest that Rc3h2 is co-expressed in T cells and binds ICOS mRNA indistinguishably in vitro, however it does not repress ICOS. Major challenges are therefore to define how the Rc3h1/2 proteins recognize cis-elements in the RNA, which cofactors they require for repression and how these proteins exert diverse functions. My project proposes to pursue three aims: (1) to describe Rc3h1/2 target recognition in the T cell transcriptome, (2) to globally identify the essential genes in this pathway of post-transcriptional repression and (3) to analyze redundant and unique roles of Rc3h1 and Rc3h2 proteins in the mouse embryo, the hematopoietic system and in models of immunity and spontaneous development of autoimmune disease.
Summary
We are constantly protected by our adaptive immune system. Its functioning requires precise control of gene expression in lymphocytes, since deregulation can cause autoimmune diseases (affecting ~5% of our population) as well as allergic reactions (~9-16%, with increasing incidence). Post-transcriptional control of gene expression is crucial in many immune decisions, however the determinants of specificity in this type of regulation are less well defined. The recently described Rc3h1 or Roquin protein prevents the development of autoimmune disease in mice. Rc3h1 destabilizes the mRNA of the inducible costimulator (ICOS), a co-receptor on T cells. ICOS is critical in the germinal center reaction in which T cell help selects B cells making high affinity antibodies. However, the molecular interactions of this posttranscriptional regulation and the pathways that specify such repressor/target relations are unsolved, and they are the focus of my work in this proposal.
Rc3h1 is an essential factor of peripheral T cell tolerance, whereas the role of its paralog Rc3h2 has not been addressed. We have recently shown that Rc3h1 is an RNA binding-protein that recognizes the 3’UTR of ICOS mRNA. Our preliminary data suggest that Rc3h2 is co-expressed in T cells and binds ICOS mRNA indistinguishably in vitro, however it does not repress ICOS. Major challenges are therefore to define how the Rc3h1/2 proteins recognize cis-elements in the RNA, which cofactors they require for repression and how these proteins exert diverse functions. My project proposes to pursue three aims: (1) to describe Rc3h1/2 target recognition in the T cell transcriptome, (2) to globally identify the essential genes in this pathway of post-transcriptional repression and (3) to analyze redundant and unique roles of Rc3h1 and Rc3h2 proteins in the mouse embryo, the hematopoietic system and in models of immunity and spontaneous development of autoimmune disease.
Max ERC Funding
1 392 400 €
Duration
Start date: 2011-11-01, End date: 2016-10-31
Project acronym RNANTIBIOTICS
Project RNA-mediated virulence gene regulation: Identification of novel antibacterial compounds
Researcher (PI) Jan Jörgen Johansson
Host Institution (HI) UMEA UNIVERSITET
Call Details Starting Grant (StG), LS6, ERC-2010-StG_20091118
Summary All kingdoms possess a large fraction of RNA-based regulation. We identified several small non-coding regulatory RNAs (ncRNAs) in the human bacterial pathogen Listeria monocytogenes that controlled virulence by a direct RNA:RNA interaction. My group have also identified several 5´-untranslated RNAs (5´-UTRs) known to control expression of their downstream mRNA by a switch mechanism triggered by certain metabolites, specific compartments of the host or by different temperatures.
In the suggested project, we will analyze the mechanism by how various RNA-species function on a molecular level by biochemical and genetic approaches. By constructing mutations (deletion and base-substitutions), the role of the regulatory RNAs and their targets during pathogenesis will be pin-pointed using different virulence model organisms. For 5´-UTRs binding specific metabolites, we will add non-metabolic analogs to examine if such molecules can block the function of the 5´-UTRs and hence infection. The core structure of one identified ncRNA will be used as a scaffold to develop an RNA interference system in bacteria.
At least one RNA-helicase has been shown to be essential for bacterial motility and growth at 4°C. It is being purified to test its in vitro properties at mRNA targets and at different temperatures. Its in vivo role will be analyzed by genetic techniques.
Bacterial resistance against different antibiotics is an increasing problem worldwide. We have identified one pyridine molecule specifically targeting listerial virulence gene expression and its mechanism of action will be revealed by genetic and biochemical techniques. A diffusible, although yet unknown molecule, with bacteriostatic activity was observed and its nature and mechanism will be revealed mainly by biochemical experiments.
Our work will give important knowledge of how the bacterium uses RNA to sense its surroundings, but will also identifiy new types of antibacterial agents.
Summary
All kingdoms possess a large fraction of RNA-based regulation. We identified several small non-coding regulatory RNAs (ncRNAs) in the human bacterial pathogen Listeria monocytogenes that controlled virulence by a direct RNA:RNA interaction. My group have also identified several 5´-untranslated RNAs (5´-UTRs) known to control expression of their downstream mRNA by a switch mechanism triggered by certain metabolites, specific compartments of the host or by different temperatures.
In the suggested project, we will analyze the mechanism by how various RNA-species function on a molecular level by biochemical and genetic approaches. By constructing mutations (deletion and base-substitutions), the role of the regulatory RNAs and their targets during pathogenesis will be pin-pointed using different virulence model organisms. For 5´-UTRs binding specific metabolites, we will add non-metabolic analogs to examine if such molecules can block the function of the 5´-UTRs and hence infection. The core structure of one identified ncRNA will be used as a scaffold to develop an RNA interference system in bacteria.
At least one RNA-helicase has been shown to be essential for bacterial motility and growth at 4°C. It is being purified to test its in vitro properties at mRNA targets and at different temperatures. Its in vivo role will be analyzed by genetic techniques.
Bacterial resistance against different antibiotics is an increasing problem worldwide. We have identified one pyridine molecule specifically targeting listerial virulence gene expression and its mechanism of action will be revealed by genetic and biochemical techniques. A diffusible, although yet unknown molecule, with bacteriostatic activity was observed and its nature and mechanism will be revealed mainly by biochemical experiments.
Our work will give important knowledge of how the bacterium uses RNA to sense its surroundings, but will also identifiy new types of antibacterial agents.
Max ERC Funding
999 996 €
Duration
Start date: 2010-10-01, End date: 2015-09-30
Project acronym SEQ-CYTOTOX
Project Next generation sequencing for detection of human primary immunodeficiency syndromes characterized by impaired cytotoxic lymphocyte function
Researcher (PI) Yenan Bryceson
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Starting Grant (StG), LS6, ERC-2012-StG_20111109
Summary Human primary immunodeficiencies affecting cytotoxic lymphocyte function are associated with high morbidity and mortality. They often present as hyperinflammatory syndromes triggered by viral infections, hematological malignancies, or systemic autoimmunity. Although recent advances have provided molecular diagnoses for some of the most severe cases, only a handful of genes have been identified and disease in a majority of patients cannot be explained by current insights. My work has elucidated fundamental mechanisms of lymphocyte cytotoxicity. Furthermore, my group is leading international efforts in developing sensitive assays of human lymphocyte cytotoxicity. Some of our assays have been implemented for clinical diagnostics world-wide. Recently, we have also identified new, widespread mutations that explain many cases of fatal immunodeficiencies early in life. We now seek support for taking such studies to yet a higher level. The outlined interdisciplinary approach presented here aims at evaluating cytotoxic lymphocyte development and function in large numbers of patients, in addition to healthy individuals in a complementary approach, using state-of-the-art methodology. We will employ next-generation sequencing to identify novel genes associated with disease. The significance of putative mutations for lymphocyte cytotoxicity will be assessed using cutting-edge experimental platforms. Together, such genotype-immunotype studies promise to reveal novel genes associated with disease and further provide understanding of human genetic diversity. Thus, these studies can directly benefit patients through improved diagnosis, aiding targeted treatments, and offer new insights into human cytotoxic lymphocyte development and function with relevance to health. Insights will also pave the way for prospective, population-based molecular epidemiology studies required to more accurately assess the panorama of diseases associated with congenital defects in lymphocyte cytotoxicity.
Summary
Human primary immunodeficiencies affecting cytotoxic lymphocyte function are associated with high morbidity and mortality. They often present as hyperinflammatory syndromes triggered by viral infections, hematological malignancies, or systemic autoimmunity. Although recent advances have provided molecular diagnoses for some of the most severe cases, only a handful of genes have been identified and disease in a majority of patients cannot be explained by current insights. My work has elucidated fundamental mechanisms of lymphocyte cytotoxicity. Furthermore, my group is leading international efforts in developing sensitive assays of human lymphocyte cytotoxicity. Some of our assays have been implemented for clinical diagnostics world-wide. Recently, we have also identified new, widespread mutations that explain many cases of fatal immunodeficiencies early in life. We now seek support for taking such studies to yet a higher level. The outlined interdisciplinary approach presented here aims at evaluating cytotoxic lymphocyte development and function in large numbers of patients, in addition to healthy individuals in a complementary approach, using state-of-the-art methodology. We will employ next-generation sequencing to identify novel genes associated with disease. The significance of putative mutations for lymphocyte cytotoxicity will be assessed using cutting-edge experimental platforms. Together, such genotype-immunotype studies promise to reveal novel genes associated with disease and further provide understanding of human genetic diversity. Thus, these studies can directly benefit patients through improved diagnosis, aiding targeted treatments, and offer new insights into human cytotoxic lymphocyte development and function with relevance to health. Insights will also pave the way for prospective, population-based molecular epidemiology studies required to more accurately assess the panorama of diseases associated with congenital defects in lymphocyte cytotoxicity.
Max ERC Funding
1 499 996 €
Duration
Start date: 2013-05-01, End date: 2018-04-30
Project acronym SOS
Project Sorting of Self
Researcher (PI) Gerhard Krönke
Host Institution (HI) UNIVERSITATSKLINIKUM ERLANGEN
Call Details Starting Grant (StG), LS6, ERC-2014-STG
Summary During inflammation and infection, we are simultaneously confronted with both self and non-self in form of dying cells and microbes, respectively. Mechanisms that facilitate the non-immunogenic clearance of self-antigens derived from apoptotic and necrotic cells and that, in parallel, allow the initiation of an immune response against invading pathogens are incompletely understood.
Recent data from our laboratory show that the immune system actively sorts apoptotic cells (ACs) and bacteria into distinct subspecies of macrophages and dendritic cells thereby enabling a segregated processing of self and non-self as well as a differential immune response against these two entities. Incorrect sorting and aberrant uptake of AC-derived self-antigens by pro-inflammatory and antigen-presenting dendritic cells, however, results in the break of self-tolerance and autoimmunity.
Due to technical limitations, the identification and fate-mapping of specific phagocyte subsets that mediate the simultaneous clearance of dying cells and pathogens in vivo has remained largely elusive. We thus plan to develop novel tools that are based on cutting-edge technologies to comprehensively elucidate the sorting of dying cells and pathogens under inflammatory conditions in vivo. We plan to generate TAT-Cre transgenic mice and bacteria that will be used in conjunction with R26-eYFP reporter animals to permanently track phagocytes after ingestion of endogenously accumulated dying cells and pathogens, respectively. These approaches will enable us to characterize the involved phagocytes, study molecular mechanisms underlying the differential processing of self and non-self and follow the phagocyte’s migratory behaviour and its subsequent differentiation. The obtained data will not only provide insights into the pathogenesis of autoimmune and infectious diseases, but will also foster the development of novel therapeutic strategies for the treatment of such disorders.
Summary
During inflammation and infection, we are simultaneously confronted with both self and non-self in form of dying cells and microbes, respectively. Mechanisms that facilitate the non-immunogenic clearance of self-antigens derived from apoptotic and necrotic cells and that, in parallel, allow the initiation of an immune response against invading pathogens are incompletely understood.
Recent data from our laboratory show that the immune system actively sorts apoptotic cells (ACs) and bacteria into distinct subspecies of macrophages and dendritic cells thereby enabling a segregated processing of self and non-self as well as a differential immune response against these two entities. Incorrect sorting and aberrant uptake of AC-derived self-antigens by pro-inflammatory and antigen-presenting dendritic cells, however, results in the break of self-tolerance and autoimmunity.
Due to technical limitations, the identification and fate-mapping of specific phagocyte subsets that mediate the simultaneous clearance of dying cells and pathogens in vivo has remained largely elusive. We thus plan to develop novel tools that are based on cutting-edge technologies to comprehensively elucidate the sorting of dying cells and pathogens under inflammatory conditions in vivo. We plan to generate TAT-Cre transgenic mice and bacteria that will be used in conjunction with R26-eYFP reporter animals to permanently track phagocytes after ingestion of endogenously accumulated dying cells and pathogens, respectively. These approaches will enable us to characterize the involved phagocytes, study molecular mechanisms underlying the differential processing of self and non-self and follow the phagocyte’s migratory behaviour and its subsequent differentiation. The obtained data will not only provide insights into the pathogenesis of autoimmune and infectious diseases, but will also foster the development of novel therapeutic strategies for the treatment of such disorders.
Max ERC Funding
1 479 781 €
Duration
Start date: 2015-05-01, End date: 2020-04-30
Project acronym TissueLymphoContexts
Project Tissue-resident Lymphocytes: Development and Function in “real-life” Contexts
Researcher (PI) Georg GASTEIGER
Host Institution (HI) JULIUS-MAXIMILIANS-UNIVERSITAT WURZBURG
Call Details Starting Grant (StG), LS6, ERC-2017-STG
Summary Most anatomical compartments, including mucosal barrier surfaces, solid organs and vascular spaces host different types of tissue-resident lymphocytes providing local networks for immune surveillance and front-line defense to microbial invasion. In addition to immediate effector functions, tissue-resident lymphocytes orchestrate and regulate inflammatory responses, and contribute to tissue homeostasis, repair and barrier function. Understanding the generation and function of tissue-resident lymphocytes is therefore expected to reveal targets for improving vaccination and immunotherapy. Barrier tissues of free-living mice and men, but not those of laboratory mice kept under specific pathogen free conditions, are continuously exposed to an array of antigenically complex pathogens, microbial symbionts and environmental factors, which dramatically alter the abundance, composition and basal activation state of local pools of tissue-resident cells. Therefore, we propose to study the development, functions and cellular interactions of tissue-resident lymphocytes in experimental models that mirror “real-life” contexts. First, we will investigate how polyclonal pools of tissue-resident memory CD8+ T cells (TRMs) are established during infection with antigenically complex pathogens. Second, we will restore physiologic exposure to specific pathogens and induce alterations of local microbiota in SPF mice, in order to investigate the induction, function and local interactions of tissue-resident lymphocytes in physiologic tissue environments. In addition, we will explore targeted microbial exposure as a „vaccination“ strategy to induce „non-canonical“ tissue-resident cells in the lung in order to improve protection against infections with multi-resistant bacteria representing major clinical problems in hospitalized patients. In summary, we will investigate fundamental mechanisms of tissue immunity and vaccination in contexts relevant for human physiology and disease.
Summary
Most anatomical compartments, including mucosal barrier surfaces, solid organs and vascular spaces host different types of tissue-resident lymphocytes providing local networks for immune surveillance and front-line defense to microbial invasion. In addition to immediate effector functions, tissue-resident lymphocytes orchestrate and regulate inflammatory responses, and contribute to tissue homeostasis, repair and barrier function. Understanding the generation and function of tissue-resident lymphocytes is therefore expected to reveal targets for improving vaccination and immunotherapy. Barrier tissues of free-living mice and men, but not those of laboratory mice kept under specific pathogen free conditions, are continuously exposed to an array of antigenically complex pathogens, microbial symbionts and environmental factors, which dramatically alter the abundance, composition and basal activation state of local pools of tissue-resident cells. Therefore, we propose to study the development, functions and cellular interactions of tissue-resident lymphocytes in experimental models that mirror “real-life” contexts. First, we will investigate how polyclonal pools of tissue-resident memory CD8+ T cells (TRMs) are established during infection with antigenically complex pathogens. Second, we will restore physiologic exposure to specific pathogens and induce alterations of local microbiota in SPF mice, in order to investigate the induction, function and local interactions of tissue-resident lymphocytes in physiologic tissue environments. In addition, we will explore targeted microbial exposure as a „vaccination“ strategy to induce „non-canonical“ tissue-resident cells in the lung in order to improve protection against infections with multi-resistant bacteria representing major clinical problems in hospitalized patients. In summary, we will investigate fundamental mechanisms of tissue immunity and vaccination in contexts relevant for human physiology and disease.
Max ERC Funding
1 498 750 €
Duration
Start date: 2018-03-01, End date: 2023-02-28
Project acronym VIRAFRONT
Project Viral frontiers – species barriers of hepatitis C virus replication
Researcher (PI) Thomas Pietschmann
Host Institution (HI) "TWINCORE, ZENTRUM FUR EXPERIMENTELLE UND KLINISCHE INFEKTIONSFORSCHUNG GMBH"
Call Details Starting Grant (StG), LS6, ERC-2011-StG_20101109
Summary "RNA viruses are responsible for important acute, chronic as well as emerging human infections. While some of these viruses – like for instance the hepatitis C virus (HCV) – are highly species-specific, others (e.g. Dengue Virus) infect different host species. These latter viruses cause zoonotic infections, i.e. infections of animal hosts that are transmitted to humans by unintentional contact. Why some RNA viruses are highly species-specific, whereas others have a broad species-tropism is poorly understood. Moreover, host-derived control mechanisms that prevent viruses to transmit across species barriers and viral evasion and adaptation strategies are only beginning to emerge. While climatic changes, augmented population density and mobility increase the risk for human exposure to novel RNA viruses, therapeutics preventing disease in humans or precluding transmission are lacking. In some cases, small animal models for drug discovery, vaccine research and pathogenesis studies are not available. The objective of the proposal is to define all factors that prevent HCV propagation in mouse liver cells. The inability of HCV to propagate in non-human cells may be due to lack or genetic incompatibility of essential viral replication co-factors and/or due to antiviral restriction mechanisms effectively suppressing viral replication in non-human host cells. Taking this into account, both rationale and unbiased genetic screening systems will be employed to derive a comprehensive profile of all factors relevant to the species barrier for HCV infection of mouse cells. Moreover, viral adaptation and evasion strategies to overcome these barriers will be explored. These pioneer studies will prepare the ground for development of urgently needed HCV small animal models and provide valuable insights into the mechanisms of pathogen control at species barriers. This should promote future development of strategies which prevent zoonotic transmission of emerging RNA viruses to humans."
Summary
"RNA viruses are responsible for important acute, chronic as well as emerging human infections. While some of these viruses – like for instance the hepatitis C virus (HCV) – are highly species-specific, others (e.g. Dengue Virus) infect different host species. These latter viruses cause zoonotic infections, i.e. infections of animal hosts that are transmitted to humans by unintentional contact. Why some RNA viruses are highly species-specific, whereas others have a broad species-tropism is poorly understood. Moreover, host-derived control mechanisms that prevent viruses to transmit across species barriers and viral evasion and adaptation strategies are only beginning to emerge. While climatic changes, augmented population density and mobility increase the risk for human exposure to novel RNA viruses, therapeutics preventing disease in humans or precluding transmission are lacking. In some cases, small animal models for drug discovery, vaccine research and pathogenesis studies are not available. The objective of the proposal is to define all factors that prevent HCV propagation in mouse liver cells. The inability of HCV to propagate in non-human cells may be due to lack or genetic incompatibility of essential viral replication co-factors and/or due to antiviral restriction mechanisms effectively suppressing viral replication in non-human host cells. Taking this into account, both rationale and unbiased genetic screening systems will be employed to derive a comprehensive profile of all factors relevant to the species barrier for HCV infection of mouse cells. Moreover, viral adaptation and evasion strategies to overcome these barriers will be explored. These pioneer studies will prepare the ground for development of urgently needed HCV small animal models and provide valuable insights into the mechanisms of pathogen control at species barriers. This should promote future development of strategies which prevent zoonotic transmission of emerging RNA viruses to humans."
Max ERC Funding
1 444 516 €
Duration
Start date: 2012-02-01, End date: 2017-01-31