Project acronym COGSYSTEMS
Project Understanding actions and intentions of others
Researcher (PI) Giacomo Rizzolatti
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PARMA
Call Details Advanced Grant (AdG), LS5, ERC-2009-AdG
Summary How do we understand the actions and intentions of others? Hereby we intend to address this issue by using a multidisciplinary approach. Our project is subdivided into four parts. In the first part we investigate the neural organization of monkey area F5, an area deeply involved in motor act understanding. By using a new set of electrodes we will describe the columnar organization of the area F5, establish the temporal relationships between the activity of F5 mirror and motor neurons, and correlate the activity of mirror neurons coding the observed motor acts in peripersonal and extrapersonal space with the activity of motor neurons in the same cortical column. In the second part we will assess the neural mechanism underlying the understanding of the intention of complex actions , i.e. actions formed by a sequence of two (or more) individual actions. The focus will be on the neurons located in ventrolateral prefrontal cortex, an area involved in the organization of high-order motor behavior. The rational of the experiment is that, while the organization of single actions and the understanding of intention behind them is function of parietal neurons, that of complex actions relies on the activity of the prefrontal lobe. In the third and fourth parts of the project we will delimit the cortical areas involved in understanding the goal (the what) and the intention (the why) of the observed actions in individuals with typical development (TD) and in children with autism and will establish the time relation between these two processes. Our hypothesis is that the chained organization of intentional motor acts is impaired in children with autism and this impairment prevents them from organizing normally their actions and from understanding others intentions.
Summary
How do we understand the actions and intentions of others? Hereby we intend to address this issue by using a multidisciplinary approach. Our project is subdivided into four parts. In the first part we investigate the neural organization of monkey area F5, an area deeply involved in motor act understanding. By using a new set of electrodes we will describe the columnar organization of the area F5, establish the temporal relationships between the activity of F5 mirror and motor neurons, and correlate the activity of mirror neurons coding the observed motor acts in peripersonal and extrapersonal space with the activity of motor neurons in the same cortical column. In the second part we will assess the neural mechanism underlying the understanding of the intention of complex actions , i.e. actions formed by a sequence of two (or more) individual actions. The focus will be on the neurons located in ventrolateral prefrontal cortex, an area involved in the organization of high-order motor behavior. The rational of the experiment is that, while the organization of single actions and the understanding of intention behind them is function of parietal neurons, that of complex actions relies on the activity of the prefrontal lobe. In the third and fourth parts of the project we will delimit the cortical areas involved in understanding the goal (the what) and the intention (the why) of the observed actions in individuals with typical development (TD) and in children with autism and will establish the time relation between these two processes. Our hypothesis is that the chained organization of intentional motor acts is impaired in children with autism and this impairment prevents them from organizing normally their actions and from understanding others intentions.
Max ERC Funding
1 992 000 €
Duration
Start date: 2010-05-01, End date: 2015-04-30
Project acronym CONCEPT
Project Construction of Perception from Touch Signals
Researcher (PI) Mathew Diamond
Host Institution (HI) SCUOLA INTERNAZIONALE SUPERIORE DI STUDI AVANZATI DI TRIESTE
Call Details Advanced Grant (AdG), LS5, ERC-2011-ADG_20110310
Summary Our sensory systems gather stimuli as elemental physical features yet we perceive a world made up of familiar objects, not wavelengths or vibrations. Perception occurs when the neuronal representation of physical parameters is transformed into the neuronal representation of meaningful objects. How does this recoding occur? An ideal platform for the inquiry is the rat whisker sensory system: it produces fast and accurate judgments of complex stimuli, yet can be broken down into accessible neuronal mechanisms. CONCEPT will examine the process that begins with whisker motion and ends with perception of the contacted object. Understanding the general principles for the construction of perception will help explain why we experience the world as we do.
The main hypothesis is that graded neuronal representations at early processing stages are “fractured” to generate discrete object representations at late processing stages. Of particular interest is the emergence of object representations as the meaning of new stimuli is acquired.
We will collect multi-site single-unit and local field potential signals simultaneously with precise behavioral indices, and will interpret data through advanced computational methods. We will begin by quantifying whisker motion as rats discriminate texture, thus defining the raw material on which the brain operates. Next, we will characterize the transformation of texture along an intracortical stream from sensory areas (where we expect that neurons encode whisker kinematics) to frontal and rhinal areas (where we expect that neurons encode objects extracted from the graded physical continuum) and hippocampus (where we expect that neurons encode objects in conjunction with context). We will test candidate processing schemes by manipulating perception on single trials using optogenetic methods.
Summary
Our sensory systems gather stimuli as elemental physical features yet we perceive a world made up of familiar objects, not wavelengths or vibrations. Perception occurs when the neuronal representation of physical parameters is transformed into the neuronal representation of meaningful objects. How does this recoding occur? An ideal platform for the inquiry is the rat whisker sensory system: it produces fast and accurate judgments of complex stimuli, yet can be broken down into accessible neuronal mechanisms. CONCEPT will examine the process that begins with whisker motion and ends with perception of the contacted object. Understanding the general principles for the construction of perception will help explain why we experience the world as we do.
The main hypothesis is that graded neuronal representations at early processing stages are “fractured” to generate discrete object representations at late processing stages. Of particular interest is the emergence of object representations as the meaning of new stimuli is acquired.
We will collect multi-site single-unit and local field potential signals simultaneously with precise behavioral indices, and will interpret data through advanced computational methods. We will begin by quantifying whisker motion as rats discriminate texture, thus defining the raw material on which the brain operates. Next, we will characterize the transformation of texture along an intracortical stream from sensory areas (where we expect that neurons encode whisker kinematics) to frontal and rhinal areas (where we expect that neurons encode objects extracted from the graded physical continuum) and hippocampus (where we expect that neurons encode objects in conjunction with context). We will test candidate processing schemes by manipulating perception on single trials using optogenetic methods.
Max ERC Funding
2 500 000 €
Duration
Start date: 2012-06-01, End date: 2018-05-31
Project acronym DIDO
Project Innovative drugs targeting IDO molecular dynamics in autoimmunity and neoplasia
Researcher (PI) Ursula Grohmann
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PERUGIA
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary "Catabolism of amino acids is an ancient survival strategy that also controls immune responses in mammals. Indoleamine 2,3-dioxygenase (IDO), a tryptophan catabolizing enzyme, is recognized as an authentic regulator of immunity in several physiopathologic conditions, including autoimmune diseases, in which it is often defective, and neoplasia, in which it promotes immune unresponsiveness. The PI’s group recently revealed that IDO does not merely degrade tryptophan and produce immunoregulatory kynurenines but also acts as a signal-transducing molecule independently of its enzyme activity. IDO’s signaling function relies on the presence of phosphorylable motifs in a region (small IDO domain) distant from the catalytic site (large IDO domain). Preliminary data indicate that IDO, depending on microenvironmental conditions, can move among distinct cellular compartments. Thus IDO may be considered a ‘moonligthing’ protein, i.e., an ancestral metabolic molecule that, during evolution, has acquired the DYNAMIC feature of moving intracellularly and switching among distinct functions by changing its conformational state. By means of computational studies, Macchiarulo’s group (team member) has identified distinct conformations of IDO, some of which are associated with optimal catalytic activity of the enzyme whereas others may favor tyrosine phosphorylation of IDO’s small domain. A switch between distinct conformations can be induced by the use of ligands that bind either the catalytic site or an accessory pocket outside the IDO catalytic site. The first aim of DIDO is to decipher the relationships between IDO conformations and multiple functions of the enzyme. A second aim is to identify small molecules with drug-like properties capable of modulating distinct IDO’s molecular conformations in order to either potentiate (a new therapeutic approach in autoimmune diseases) or inhibit (more efficient anti-tumor therapeutic strategy) immunoregulatory signaling ability of IDO."
Summary
"Catabolism of amino acids is an ancient survival strategy that also controls immune responses in mammals. Indoleamine 2,3-dioxygenase (IDO), a tryptophan catabolizing enzyme, is recognized as an authentic regulator of immunity in several physiopathologic conditions, including autoimmune diseases, in which it is often defective, and neoplasia, in which it promotes immune unresponsiveness. The PI’s group recently revealed that IDO does not merely degrade tryptophan and produce immunoregulatory kynurenines but also acts as a signal-transducing molecule independently of its enzyme activity. IDO’s signaling function relies on the presence of phosphorylable motifs in a region (small IDO domain) distant from the catalytic site (large IDO domain). Preliminary data indicate that IDO, depending on microenvironmental conditions, can move among distinct cellular compartments. Thus IDO may be considered a ‘moonligthing’ protein, i.e., an ancestral metabolic molecule that, during evolution, has acquired the DYNAMIC feature of moving intracellularly and switching among distinct functions by changing its conformational state. By means of computational studies, Macchiarulo’s group (team member) has identified distinct conformations of IDO, some of which are associated with optimal catalytic activity of the enzyme whereas others may favor tyrosine phosphorylation of IDO’s small domain. A switch between distinct conformations can be induced by the use of ligands that bind either the catalytic site or an accessory pocket outside the IDO catalytic site. The first aim of DIDO is to decipher the relationships between IDO conformations and multiple functions of the enzyme. A second aim is to identify small molecules with drug-like properties capable of modulating distinct IDO’s molecular conformations in order to either potentiate (a new therapeutic approach in autoimmune diseases) or inhibit (more efficient anti-tumor therapeutic strategy) immunoregulatory signaling ability of IDO."
Max ERC Funding
2 442 078 €
Duration
Start date: 2014-02-01, End date: 2019-01-31
Project acronym EU-rhythmy
Project Molecular strategies to treat inherited arrhythmias
Researcher (PI) Silvia Giuliana Priori
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PAVIA
Call Details Advanced Grant (AdG), LS7, ERC-2014-ADG
Summary Sudden cardiac death (SCD) is a leading cause of death in western countries: coronary artery disease is the major cause of SCD in older subjects while inherited arrhythmogenic diseases are the leading cause of SCD in younger individuals. After 25 years dedicated to research of the molecular bases of heritable arrhythmias, the PI of this proposal now intends to pioneer gene therapy for prevention of SCD: a virtually unexplored field. The development of molecular therapies for rhythm disturbances is a high risk effort however, if successful, it will be highly rewarding. The PI has envisioned an ambitious and comprehensive project to target two severe inherited arrhythmogenic diseases: dominant catecholaminergic polymorphic ventricular tachycardia (CPVT) and Long QT syndrome type 8 (LQT8). The availability of a clinically relevant model is critical to ensure clinical translation of results: the team will exploit an existing CPVT model and will engineer a knock-in pig to model LQT8. The PI and her team will investigate innovative strategies of gene-delivery, gene-silencing and gene-editing to the heart comparing efficacy of different constructs and promoters. The team will also carefully engineer novel gene-therapy approaches to avoid the development of regional inhomogeneity in protein expression that may facilitate proarrhythmic events. Such a comprehensive approach will provide a most valuable core of knowledge on the comparative efficacy of a broad range of molecular strategies on the electrical milieu of the heart. It is expected that these results will not only benefit CPVT and LQT8 but rather they will foster development of gene therapy for other inherited and acquired arrhythmias.
Summary
Sudden cardiac death (SCD) is a leading cause of death in western countries: coronary artery disease is the major cause of SCD in older subjects while inherited arrhythmogenic diseases are the leading cause of SCD in younger individuals. After 25 years dedicated to research of the molecular bases of heritable arrhythmias, the PI of this proposal now intends to pioneer gene therapy for prevention of SCD: a virtually unexplored field. The development of molecular therapies for rhythm disturbances is a high risk effort however, if successful, it will be highly rewarding. The PI has envisioned an ambitious and comprehensive project to target two severe inherited arrhythmogenic diseases: dominant catecholaminergic polymorphic ventricular tachycardia (CPVT) and Long QT syndrome type 8 (LQT8). The availability of a clinically relevant model is critical to ensure clinical translation of results: the team will exploit an existing CPVT model and will engineer a knock-in pig to model LQT8. The PI and her team will investigate innovative strategies of gene-delivery, gene-silencing and gene-editing to the heart comparing efficacy of different constructs and promoters. The team will also carefully engineer novel gene-therapy approaches to avoid the development of regional inhomogeneity in protein expression that may facilitate proarrhythmic events. Such a comprehensive approach will provide a most valuable core of knowledge on the comparative efficacy of a broad range of molecular strategies on the electrical milieu of the heart. It is expected that these results will not only benefit CPVT and LQT8 but rather they will foster development of gene therapy for other inherited and acquired arrhythmias.
Max ERC Funding
2 314 029 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym EYEGET
Project Gene therapy of inherited retinal diseases
Researcher (PI) Alberto AURICCHIO
Host Institution (HI) FONDAZIONE TELETHON
Call Details Advanced Grant (AdG), LS7, ERC-2015-AdG
Summary Inherited retinal degenerations (IRDs) are a major cause of blindness worldwide. IRD patients witness inexorable progressive vision loss as no therapy is currently available. In the last decade my group has significantly contributed to a change of this scenario by developing efficient adeno-associated viral (AAV) vectors for retinal gene therapy that are safe and effective in humans. The objective of EYEGET (EYE GEne Therapy) is to overcome some of the current major limitations in the field of retinal gene therapy to expand initial therapeutic successes to a larger number of IRDs. To achieve this, we propose to use four parallel, highly innovative and complementary approaches: i. expansion of the limited AAV cargo capacity by a novel methodology based on co-administration of multiple AAVs that reassemble in target retinal cells and reconstitute large genes; ii. targeting of frequent dominant gain-of-function mutations that cause RP using state-of-the-art AAV-mediated genome editing technologies; iii. induction of retinal cells clearance of toxic IRD products by AAV-mediated activation of autophagy and lysosomal function; iv. development of methodologies to directly convert fibroblasts to photoreceptors that can be transplanted in retinas from IRD patients with advanced PR loss and for whom in vivo gene therapy is no longer an option. We will use a combination of in vitro and in vivo state-of-the-art technologies including novel AAV vector design, high content screening of drugs that enhance AAV transduction, genome editing, and advanced in vivo retinal phenotyping to obtain proof-of-concept for each of these therapeutic strategies. The results from this study may impact the quality of life of millions of people worldwide by providing a cure based on gene and/or cell therapy for a large group of IRDs.
Summary
Inherited retinal degenerations (IRDs) are a major cause of blindness worldwide. IRD patients witness inexorable progressive vision loss as no therapy is currently available. In the last decade my group has significantly contributed to a change of this scenario by developing efficient adeno-associated viral (AAV) vectors for retinal gene therapy that are safe and effective in humans. The objective of EYEGET (EYE GEne Therapy) is to overcome some of the current major limitations in the field of retinal gene therapy to expand initial therapeutic successes to a larger number of IRDs. To achieve this, we propose to use four parallel, highly innovative and complementary approaches: i. expansion of the limited AAV cargo capacity by a novel methodology based on co-administration of multiple AAVs that reassemble in target retinal cells and reconstitute large genes; ii. targeting of frequent dominant gain-of-function mutations that cause RP using state-of-the-art AAV-mediated genome editing technologies; iii. induction of retinal cells clearance of toxic IRD products by AAV-mediated activation of autophagy and lysosomal function; iv. development of methodologies to directly convert fibroblasts to photoreceptors that can be transplanted in retinas from IRD patients with advanced PR loss and for whom in vivo gene therapy is no longer an option. We will use a combination of in vitro and in vivo state-of-the-art technologies including novel AAV vector design, high content screening of drugs that enhance AAV transduction, genome editing, and advanced in vivo retinal phenotyping to obtain proof-of-concept for each of these therapeutic strategies. The results from this study may impact the quality of life of millions of people worldwide by providing a cure based on gene and/or cell therapy for a large group of IRDs.
Max ERC Funding
2 499 564 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym FAST
Project Investigating new therapeutic approaches to Friedreich's Ataxia
Researcher (PI) Roberto Testi
Host Institution (HI) UNIVERSITA DEGLI STUDI DI ROMA TOR VERGATA
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Friedreich’s Ataxia (FRDA) is a devastating degenerative disease with no specific therapy. It is passed by autosomal recessive inheritance and affects 1:30,000 individuals in Caucasian populations. Symptoms appear in the first decade of life and include progressive and unremitting lack of movement coordination, leading to complete inability, and dilated cardiomyopathy leading to congestive heart failure, the most common cause of premature death. FRDA is due to the insufficient transcription of the gene coding for the mitochondrial protein frataxin. Reduced cellular levels of frataxin cause impaired mitochondrial function and increased sensitivity to oxidative stress, leading to accelerated cell death in critical tissues.
Severity of the disease critically depends on residual frataxin levels. Therapeutic efforts are mostly focused on increasing cellular frataxin . We found that frataxin is normally degraded by the ubiquitin-proteasome system. We identified the lysine responsible for the ubiquitination of frataxin and, by computational screening followed by experimental validation, we identified and validated a series of small molecules, called ubiquitin-competing molecules (UCM), that prevent frataxin ubiquitination and induce frataxin accumulation in cells derived from FRDA patients. Moreover, treatment with UCM partially rescues aconitase and ATP production defects in cells derived from FRDA patients.
Our goal is two fold: 1) submit a set of leads we already identified, as well as their new and more complex derivatives, to preclinical testing in FRDA mice 2) identify the E3 ligase that is responsible for frataxin ubiquitination, and investigate the possibility to use it as a druggable target for small molecules to prevent frataxin degradation.
Summary
Friedreich’s Ataxia (FRDA) is a devastating degenerative disease with no specific therapy. It is passed by autosomal recessive inheritance and affects 1:30,000 individuals in Caucasian populations. Symptoms appear in the first decade of life and include progressive and unremitting lack of movement coordination, leading to complete inability, and dilated cardiomyopathy leading to congestive heart failure, the most common cause of premature death. FRDA is due to the insufficient transcription of the gene coding for the mitochondrial protein frataxin. Reduced cellular levels of frataxin cause impaired mitochondrial function and increased sensitivity to oxidative stress, leading to accelerated cell death in critical tissues.
Severity of the disease critically depends on residual frataxin levels. Therapeutic efforts are mostly focused on increasing cellular frataxin . We found that frataxin is normally degraded by the ubiquitin-proteasome system. We identified the lysine responsible for the ubiquitination of frataxin and, by computational screening followed by experimental validation, we identified and validated a series of small molecules, called ubiquitin-competing molecules (UCM), that prevent frataxin ubiquitination and induce frataxin accumulation in cells derived from FRDA patients. Moreover, treatment with UCM partially rescues aconitase and ATP production defects in cells derived from FRDA patients.
Our goal is two fold: 1) submit a set of leads we already identified, as well as their new and more complex derivatives, to preclinical testing in FRDA mice 2) identify the E3 ligase that is responsible for frataxin ubiquitination, and investigate the possibility to use it as a druggable target for small molecules to prevent frataxin degradation.
Max ERC Funding
1 496 200 €
Duration
Start date: 2012-03-01, End date: 2015-02-28
Project acronym FUNMETA
Project Metabolomics of fungal diseases: a systems biology approach for biomarkers discovery and therapy
Researcher (PI) Luigina Romani
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PERUGIA
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary Humans have evolved intimate symbiotic relationships with a consortium of gut microbes (including fungi) and individual variations in the microbiome influence host health and disease. The fact that fungi are capable of colonizing almost every niche within the human body suggests that they must possess particular immune adaptation mechanisms, the breakdown of which may result in fatal fungal infections and severe fungal diseases. Traditional reductionist approaches of the past have not been sufficient to address these new challenges in the pathogenesis of fungal diseases. Here, I propose an integrated, systems biology approach to understand the role of L-tryptophan (trp) metabolic pathways in multilevel host−fungus interactions. Present in mammals as well as in fungi, pathways of trp metabolic pathways are exploited by the host and the fungal biota for survival and immune adaptation. A variety of indole derivatives, generated through conversion from dietary trp by symbiotic bacteria, activate the aryl hydrocarbon receptor/IL-22 pathway that provides antifungal resistance and tissue repair. Harmful inflammatory responses to fungi are instead tamed by kynurenines generated via the enzyme indoleamine 2,3–dioxygenase (IDO) of the trp pathway. Through high-throughput wet-lab ‘omics’ techniques combined with computational techniques, the project aims at defining the molecular basis of mammalian and fungal IDO activity and a metabolic network linking the metabolic phenotype (metabotype) to immune adaptations and its possible breakdown in experimental and human fungal infections. The project will provide ideal post-graduate training focussed on the development of metabolomics for diagnosis of fungal diseases and optimization of current antifungal therapy and diet that are of relevance to public health care solutions.
Summary
Humans have evolved intimate symbiotic relationships with a consortium of gut microbes (including fungi) and individual variations in the microbiome influence host health and disease. The fact that fungi are capable of colonizing almost every niche within the human body suggests that they must possess particular immune adaptation mechanisms, the breakdown of which may result in fatal fungal infections and severe fungal diseases. Traditional reductionist approaches of the past have not been sufficient to address these new challenges in the pathogenesis of fungal diseases. Here, I propose an integrated, systems biology approach to understand the role of L-tryptophan (trp) metabolic pathways in multilevel host−fungus interactions. Present in mammals as well as in fungi, pathways of trp metabolic pathways are exploited by the host and the fungal biota for survival and immune adaptation. A variety of indole derivatives, generated through conversion from dietary trp by symbiotic bacteria, activate the aryl hydrocarbon receptor/IL-22 pathway that provides antifungal resistance and tissue repair. Harmful inflammatory responses to fungi are instead tamed by kynurenines generated via the enzyme indoleamine 2,3–dioxygenase (IDO) of the trp pathway. Through high-throughput wet-lab ‘omics’ techniques combined with computational techniques, the project aims at defining the molecular basis of mammalian and fungal IDO activity and a metabolic network linking the metabolic phenotype (metabotype) to immune adaptations and its possible breakdown in experimental and human fungal infections. The project will provide ideal post-graduate training focussed on the development of metabolomics for diagnosis of fungal diseases and optimization of current antifungal therapy and diet that are of relevance to public health care solutions.
Max ERC Funding
2 299 200 €
Duration
Start date: 2012-04-01, End date: 2018-03-31
Project acronym FUNSEL
Project Generation of AAV-based, arrayed genetic libraries for in vivo functional selection: an innovative approach to identify secreted factors and microRNAs against degenerative disorders
Researcher (PI) Mauro Giacca
Host Institution (HI) INTERNATIONAL CENTRE FOR GENETIC ENGINEERING AND BIOTECHNOLOGY
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary A foremost health problem stems from the burden of degenerative diseases, including heart failure, neurodegeneration, retinal degeneration and diabetes, essentially linked to the aging of the human population and the incapacity of post-mitotic tissues to undergo efficient repair. This is an ambitious, highly innovative project aimed at developing an in vivo selection procedure, based on gene transfer of two genetic libraries cloned into Adeno-Associated Virus (AAV)-based vectors, for the identification of novel secreted factors or microRNAs providing benefit against various degenerative diseases. Two arrayed libraries will be generated, one coding for ~1,300 cDNAs from the mouse secretome, the other for all known microRNAs (~800 genes). Pools of vectors from each library will be obtained with serotypes suitable for in vivo transduction of different organs. The vectors will be injected in a series of mouse models of degenerative disorders involving damage to cardiomyocytes,, neurodegeneration, retinal degeneration and loss of beta-cells in the pancreas. The degenerative conditions will drive the selection for secreted factors or miRNA putatively preventing cell apoptosis, enhancing residual cell function or, in the best possible scenario, promoting tissue regeneration. This in vivo selection approach, which is supported by very encouraging preliminary results, has never been attempted before and is rendered possible by the property of AAV vectors to be produced at high titers, infect tissues at high multiplicity, persist in the transduced cells for prolonged period of times and efficiently express their transgenes in vivo. In addition to its final goal of identifying novel biotherapeutics, the project entails the successful achievement of several intermediate objectives and is expected to extend both technology and knowledge beyond the state-of-the art.
Summary
A foremost health problem stems from the burden of degenerative diseases, including heart failure, neurodegeneration, retinal degeneration and diabetes, essentially linked to the aging of the human population and the incapacity of post-mitotic tissues to undergo efficient repair. This is an ambitious, highly innovative project aimed at developing an in vivo selection procedure, based on gene transfer of two genetic libraries cloned into Adeno-Associated Virus (AAV)-based vectors, for the identification of novel secreted factors or microRNAs providing benefit against various degenerative diseases. Two arrayed libraries will be generated, one coding for ~1,300 cDNAs from the mouse secretome, the other for all known microRNAs (~800 genes). Pools of vectors from each library will be obtained with serotypes suitable for in vivo transduction of different organs. The vectors will be injected in a series of mouse models of degenerative disorders involving damage to cardiomyocytes,, neurodegeneration, retinal degeneration and loss of beta-cells in the pancreas. The degenerative conditions will drive the selection for secreted factors or miRNA putatively preventing cell apoptosis, enhancing residual cell function or, in the best possible scenario, promoting tissue regeneration. This in vivo selection approach, which is supported by very encouraging preliminary results, has never been attempted before and is rendered possible by the property of AAV vectors to be produced at high titers, infect tissues at high multiplicity, persist in the transduced cells for prolonged period of times and efficiently express their transgenes in vivo. In addition to its final goal of identifying novel biotherapeutics, the project entails the successful achievement of several intermediate objectives and is expected to extend both technology and knowledge beyond the state-of-the art.
Max ERC Funding
1 824 000 €
Duration
Start date: 2010-04-01, End date: 2015-03-31
Project acronym HD-DittoGraph
Project HD-DittoGraph: a digital human Embryonic Stem Cell platform for Huntington's repeats
Researcher (PI) Elena CATTANEO
Host Institution (HI) UNIVERSITA DEGLI STUDI DI MILANO
Call Details Advanced Grant (AdG), LS5, ERC-2016-ADG
Summary This proposal is aimed at identifying the molecular mechanisms that have brought the human Huntington Disease-causing Huntingtin (Htt) exon 1, with its pure and unstable CAG repeat, to be shaped the way it is today. Specifically, we intend to screen for genetic elements affecting Htt repeat length instability in dividing and postmitotic neuronal cells. The novelty of our approach relies on the construction of a human embryonic stem (hES) cell platform that couples highly efficient CRISPR/Cas9 technology with genome-wide screenings and third generation sequencing, to test the contribution of thousands of unequivocally barcoded cis and trans modifiers on Htt exon 1 repeats instability.
In Aim 1, we will test the contribution of cis-modifiers to repeat instability during multiple mitotic divisions, by generating a hES cell platform where we will subsequently introduce a barcoded donor library of different Htt exon 1 constructs, with different CAG and flanking sequences, at the Htt locus.
In Aim 2 our hES cell platform will be implemented with inducible Cas9 elements and sgRNAs libraries to perform genome-wide loss and gain of function (LOF, GOF) screenings of trans-acting modifiers of repeat sequence and size. The sgRNAs will act as barcodes for the modifier genes, allowing to test their causative role on repeat size changes.
In Aim 3, we will exploit the neurogenic potential of hES cells in our LOF and GOF platforms to identify Htt exon 1 repeat modifiers in differentiating striatal neurons. Candidate modifier genes will be individually validated and tested for their functional impact on gene networks by transcriptome analysis.
In all approaches, third generation sequencing and ad hoc computational pipelines will allow the simultaneous identification of the repeat changes and their association to the corresponding modifiers. Overall, this research proposal is expected to provide key molecular and genetic insights into the process of Htt repeat expansion in human
Summary
This proposal is aimed at identifying the molecular mechanisms that have brought the human Huntington Disease-causing Huntingtin (Htt) exon 1, with its pure and unstable CAG repeat, to be shaped the way it is today. Specifically, we intend to screen for genetic elements affecting Htt repeat length instability in dividing and postmitotic neuronal cells. The novelty of our approach relies on the construction of a human embryonic stem (hES) cell platform that couples highly efficient CRISPR/Cas9 technology with genome-wide screenings and third generation sequencing, to test the contribution of thousands of unequivocally barcoded cis and trans modifiers on Htt exon 1 repeats instability.
In Aim 1, we will test the contribution of cis-modifiers to repeat instability during multiple mitotic divisions, by generating a hES cell platform where we will subsequently introduce a barcoded donor library of different Htt exon 1 constructs, with different CAG and flanking sequences, at the Htt locus.
In Aim 2 our hES cell platform will be implemented with inducible Cas9 elements and sgRNAs libraries to perform genome-wide loss and gain of function (LOF, GOF) screenings of trans-acting modifiers of repeat sequence and size. The sgRNAs will act as barcodes for the modifier genes, allowing to test their causative role on repeat size changes.
In Aim 3, we will exploit the neurogenic potential of hES cells in our LOF and GOF platforms to identify Htt exon 1 repeat modifiers in differentiating striatal neurons. Candidate modifier genes will be individually validated and tested for their functional impact on gene networks by transcriptome analysis.
In all approaches, third generation sequencing and ad hoc computational pipelines will allow the simultaneous identification of the repeat changes and their association to the corresponding modifiers. Overall, this research proposal is expected to provide key molecular and genetic insights into the process of Htt repeat expansion in human
Max ERC Funding
2 040 943 €
Duration
Start date: 2018-03-01, End date: 2023-02-28
Project acronym I-MIRNOME
Project Lymphocyte microRNAs in health and disease: Understanding lymphocyte functions through the identification of microRNA target genes and exploiting serum microRNA signatures to monitor immune responses
Researcher (PI) Sergio Abrignani
Host Institution (HI) UNIVERSITA DEGLI STUDI DI MILANO
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary Background: CD4+ T lymphocyte subsets orchestrate immune responses in health and disease. Little is known on control of T cell differentiation exerted by microRNA that affect mRNA translation. The identification of microRNA and their targets that regulate differentiation of T cell subsets may provide new therapeutic targets for immune-mediated diseases. Since microRNA are released in exosomes and circulate in blood, activities of tissue-derived lymphocytes could be assessed by microRNA signatures in the serum. We have defined microRNAs present in resting lymphocyte subsets from peripheral blood and measured lymphocyte-derived microRNAs in the serum. We have also solved important challenges for the identification of microRNA targets, the definition of signatures of activated T cells and their monitoring in the serum, which form the key topics of this application. Advancing State-of-the-Art and objectives: We will identify microRNA of CD4+ T cell subsets purified from inflamed organs and investigate microRNA target network that regulates T cell differentiation. We will exploit this knowledge to profile signatures of in vivo activated T cells and to map genes that could improve understanding of T cell commitment. We will also develop quantitative assays to monitor microRNA signatures in the serum and provide functional evidence of key genes targeted by microRNA which could be targets of immunomodulatory drugs. Significance: This application addresses important challenges at the frontiers of immunology and could lead to significant advances in immunotherapies and diagnostic tools for patients with immune mediated diseases. New ways of identifying microRNA targets and techniques to quantify microRNA signatures in the serum, could be widely applicable in biomedical research.
Summary
Background: CD4+ T lymphocyte subsets orchestrate immune responses in health and disease. Little is known on control of T cell differentiation exerted by microRNA that affect mRNA translation. The identification of microRNA and their targets that regulate differentiation of T cell subsets may provide new therapeutic targets for immune-mediated diseases. Since microRNA are released in exosomes and circulate in blood, activities of tissue-derived lymphocytes could be assessed by microRNA signatures in the serum. We have defined microRNAs present in resting lymphocyte subsets from peripheral blood and measured lymphocyte-derived microRNAs in the serum. We have also solved important challenges for the identification of microRNA targets, the definition of signatures of activated T cells and their monitoring in the serum, which form the key topics of this application. Advancing State-of-the-Art and objectives: We will identify microRNA of CD4+ T cell subsets purified from inflamed organs and investigate microRNA target network that regulates T cell differentiation. We will exploit this knowledge to profile signatures of in vivo activated T cells and to map genes that could improve understanding of T cell commitment. We will also develop quantitative assays to monitor microRNA signatures in the serum and provide functional evidence of key genes targeted by microRNA which could be targets of immunomodulatory drugs. Significance: This application addresses important challenges at the frontiers of immunology and could lead to significant advances in immunotherapies and diagnostic tools for patients with immune mediated diseases. New ways of identifying microRNA targets and techniques to quantify microRNA signatures in the serum, could be widely applicable in biomedical research.
Max ERC Funding
2 496 000 €
Duration
Start date: 2011-06-01, End date: 2016-05-31
Project acronym MEMEME
Project Randomized controlled trial of metformin and dietary restriction to prevent age-related morbid events in people with metabolic syndrome
Researcher (PI) Franco Berrino
Host Institution (HI) FONDAZIONE IRCCS ISTITUTO NAZIONALE DEI TUMORI
Call Details Advanced Grant (AdG), LS7, ERC-2012-ADG_20120314
Summary Age–related chronic diseases (ArCD) are complex non-linear processes that depends on a large number of interconnected genetic and metabolic pathways which should be tackled with a many faceted preventive strategy. Calorie-dense diet and sedentary lifestyle are responsible of the growing prevalence of metabolic syndrome (MetS), which, together with tobacco, is the major preventable cause of ArCD, mediated by the increased availability of insulin, growth factors, and inflammatory cytokines. In animals, calorie restriction (CR) is the most potent dietary intervention for preventing ArCD and prolonging life. In humans, we and others have shown that a sustainable CR, resulting in decreased prevalence of MetS, can be obtained through a comprehensive change in dietary habits, reducing animal food and refined carbohydrates, and increasing whole grain cereal products, legumes and vegetables, according to the Mediterranean and macrobiotic diet principles. Metformin (MET), an antidiabetic drug associated with decreased cancer incidence, activates the same gene pathways activated by CR, including AMPK/TSC, which reduces energy consuming processes, and cell proliferation (through the inhibition of mTOR). We propose a placebo controlled randomised trial to test whether treatment with MET, with or without associated CR, reduces the incidence of major chronic diseases, such as cancer, myocardial infarction, stroke, and diabetes. We estimated that randomizing 2,000 women and men aged 55-74 at high risk of developing ArCD because of MetS, shall provide over 90% power (at alpha = 0.05) to detect a significant 25 to 33% reduction of ArCD incidence in 5 years. In a subsample of the cohort we shall study the genetic and epigenetic mechanisms of the preventive action of metformin and CR.
Summary
Age–related chronic diseases (ArCD) are complex non-linear processes that depends on a large number of interconnected genetic and metabolic pathways which should be tackled with a many faceted preventive strategy. Calorie-dense diet and sedentary lifestyle are responsible of the growing prevalence of metabolic syndrome (MetS), which, together with tobacco, is the major preventable cause of ArCD, mediated by the increased availability of insulin, growth factors, and inflammatory cytokines. In animals, calorie restriction (CR) is the most potent dietary intervention for preventing ArCD and prolonging life. In humans, we and others have shown that a sustainable CR, resulting in decreased prevalence of MetS, can be obtained through a comprehensive change in dietary habits, reducing animal food and refined carbohydrates, and increasing whole grain cereal products, legumes and vegetables, according to the Mediterranean and macrobiotic diet principles. Metformin (MET), an antidiabetic drug associated with decreased cancer incidence, activates the same gene pathways activated by CR, including AMPK/TSC, which reduces energy consuming processes, and cell proliferation (through the inhibition of mTOR). We propose a placebo controlled randomised trial to test whether treatment with MET, with or without associated CR, reduces the incidence of major chronic diseases, such as cancer, myocardial infarction, stroke, and diabetes. We estimated that randomizing 2,000 women and men aged 55-74 at high risk of developing ArCD because of MetS, shall provide over 90% power (at alpha = 0.05) to detect a significant 25 to 33% reduction of ArCD incidence in 5 years. In a subsample of the cohort we shall study the genetic and epigenetic mechanisms of the preventive action of metformin and CR.
Max ERC Funding
2 500 000 €
Duration
Start date: 2013-08-01, End date: 2019-07-31
Project acronym OMVAC
Project Outer Membrane Vesicles (OMVs) from “Vaccinobacter”: A Synthetic Biology approach for effective vaccines against infectious diseases and cancer
Researcher (PI) Guido Grandi
Host Institution (HI) UNIVERSITA DEGLI STUDI DI TRENTO
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary This proposal intends to apply Synthetic Biology to create a new bacterial species, Vaccinobacter, devoted to the production of multivalent, highly effective vaccines. The project originates from the evidence that Outer membrane Vesicles (OMVs) naturally produced by all Gram-negative bacteria can induce remarkable protective immunity, a property already exploited to develop anti-Neisseria vaccines now available for human use. OMV protection is mediated by the abundance of Pathogen-Associated-Molecular Patterns (PAMPs), known to play a key role in stimulating innate immunity. Moreover, OMVs can be engineered by delivering recombinant proteins to bacterial periplasm and outer membrane. Intrinsic adjuvanticity and propensity to be manipulated potentially make OMVs an ideal vaccine platform, particularly indicated when antigen combinations (for pathogens with genetic variability) and strong potentiation of immunity (for the elderly and cancer) are needed. However, full exploitation of OMVs as vaccines is prevented by: i) presence of potentially reactogenic compounds such as LPS, virulence factors, and toxins, ii) presence of several irrelevant proteins, which dilute immune responses, iii) lack of broadly applicable molecular tools to load OMVs with foreign antigens. Scope of the project is to provide novel solutions to solve these limitations and demonstrate the unique performance OMVs as vaccines by testing them on complex pathogens and cancer. Main project activities are: 1) remodelling of E. coli genome to create “Vaccinobacter”, a “living factory” of OMVs deprived of all unnecessary components but carrying the relevant immune potentiators, 2) characterization and optimization of the immune stimulatory properties of OMVs, 3) development of novel methods to incorporate foreign antigens into Vaccinobacter-derived OMVs, 4) loading of OMVs with selected pathogen- and cancer-derived antigens and demonstration of their protective efficacy in appropriate animal models.
Summary
This proposal intends to apply Synthetic Biology to create a new bacterial species, Vaccinobacter, devoted to the production of multivalent, highly effective vaccines. The project originates from the evidence that Outer membrane Vesicles (OMVs) naturally produced by all Gram-negative bacteria can induce remarkable protective immunity, a property already exploited to develop anti-Neisseria vaccines now available for human use. OMV protection is mediated by the abundance of Pathogen-Associated-Molecular Patterns (PAMPs), known to play a key role in stimulating innate immunity. Moreover, OMVs can be engineered by delivering recombinant proteins to bacterial periplasm and outer membrane. Intrinsic adjuvanticity and propensity to be manipulated potentially make OMVs an ideal vaccine platform, particularly indicated when antigen combinations (for pathogens with genetic variability) and strong potentiation of immunity (for the elderly and cancer) are needed. However, full exploitation of OMVs as vaccines is prevented by: i) presence of potentially reactogenic compounds such as LPS, virulence factors, and toxins, ii) presence of several irrelevant proteins, which dilute immune responses, iii) lack of broadly applicable molecular tools to load OMVs with foreign antigens. Scope of the project is to provide novel solutions to solve these limitations and demonstrate the unique performance OMVs as vaccines by testing them on complex pathogens and cancer. Main project activities are: 1) remodelling of E. coli genome to create “Vaccinobacter”, a “living factory” of OMVs deprived of all unnecessary components but carrying the relevant immune potentiators, 2) characterization and optimization of the immune stimulatory properties of OMVs, 3) development of novel methods to incorporate foreign antigens into Vaccinobacter-derived OMVs, 4) loading of OMVs with selected pathogen- and cancer-derived antigens and demonstration of their protective efficacy in appropriate animal models.
Max ERC Funding
2 612 828 €
Duration
Start date: 2014-06-01, End date: 2019-05-31
Project acronym ONCOLYTIC-HERPES
Project ONCOLYTIC HERPESVIRUSES RETARGETED TO CANCER- SPECIFIC RECEPTORS
Researcher (PI) Maria Gabriella Campadelli
Host Institution (HI) ALMA MATER STUDIORUM - UNIVERSITA DI BOLOGNA
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary Cancer remains a major health burden worldwide. The aggressive targeting of metabolic pathways shared by normal and cancer cells results in prolonged survival and cures, but at a tremendous cost to patient life quality. What is missing is a therapeutic agent that clearly differentiates normal and cancer cells. This proposal delineates a process for killing exclusively cancer cells with no interference with normal cells. In the past two decades there has been considerable effort to develop attenuated viruses for killing cancer cells. Of the oncolytic viruses in clinical trials, attenuated herpes simplex viruses (HSV) are among the most promising because of safety, affinity for cancer cells, ability to treat patients multiple times without block by adaptive immunity. The shortcoming is that they do not discriminate between normal and cancer cells, are effective in a limited number of patients. The remarkable accomplishment by my laboratory at the basis of the proposal is the genetic engineering of HSVs that specifically infect and kill cancer cells and cannot infect normal cells. The prototype retargeted HSV targets HER2, a receptor in breast, ovary and other tumors. HER2-HSV ablates human breast and ovary cancers, and glioblastoma after intratumoral or intraperitoneal administration.
This proposal addresses basic research issues for the advancement of retargeted oncolytic HSVs. It is organized in 5 AIMS
• Engineer a non-cancer cell line for virus production acceptable to Health Authorities and re-engineer the retargeted-HSV accordingly. This will enable production of clinical grade retargeted-HSVs for clinical tests (AIM1)
• Engineer retargeted-HSVs suitable for systemic delivery and for boosting anti-tumor immunity (AIM2-3)
• Apply our platform to expand the repertoire of oncolytic HSVs that target glioblastomas, prostate, head-and-neck, colon carcinomas (AIM4)
• Determine the tumorigenic potential of cancer cells that escape killing by retargeted HSVs (AIM5)
Summary
Cancer remains a major health burden worldwide. The aggressive targeting of metabolic pathways shared by normal and cancer cells results in prolonged survival and cures, but at a tremendous cost to patient life quality. What is missing is a therapeutic agent that clearly differentiates normal and cancer cells. This proposal delineates a process for killing exclusively cancer cells with no interference with normal cells. In the past two decades there has been considerable effort to develop attenuated viruses for killing cancer cells. Of the oncolytic viruses in clinical trials, attenuated herpes simplex viruses (HSV) are among the most promising because of safety, affinity for cancer cells, ability to treat patients multiple times without block by adaptive immunity. The shortcoming is that they do not discriminate between normal and cancer cells, are effective in a limited number of patients. The remarkable accomplishment by my laboratory at the basis of the proposal is the genetic engineering of HSVs that specifically infect and kill cancer cells and cannot infect normal cells. The prototype retargeted HSV targets HER2, a receptor in breast, ovary and other tumors. HER2-HSV ablates human breast and ovary cancers, and glioblastoma after intratumoral or intraperitoneal administration.
This proposal addresses basic research issues for the advancement of retargeted oncolytic HSVs. It is organized in 5 AIMS
• Engineer a non-cancer cell line for virus production acceptable to Health Authorities and re-engineer the retargeted-HSV accordingly. This will enable production of clinical grade retargeted-HSVs for clinical tests (AIM1)
• Engineer retargeted-HSVs suitable for systemic delivery and for boosting anti-tumor immunity (AIM2-3)
• Apply our platform to expand the repertoire of oncolytic HSVs that target glioblastomas, prostate, head-and-neck, colon carcinomas (AIM4)
• Determine the tumorigenic potential of cancer cells that escape killing by retargeted HSVs (AIM5)
Max ERC Funding
2 477 346 €
Duration
Start date: 2014-03-01, End date: 2020-02-29
Project acronym QUIDPROQUO
Project Molecular Nanotechnology for Life Science Applications: QUantitative Interactomics for Diagnostics, PROteomics and QUantitative Oncology
Researcher (PI) Giacinto Scoles
Host Institution (HI) UNIVERSITA DEGLI STUDI DI UDINE
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary The main goal of this proposal is to introduce innovative devices and protocols (based on nano- manipulation, the response of micro-(nano-)mechanical oscillators and nano-fluidics) to carry out, precise, high throughput, high sensitivity, and low cost interactomic measurements. We aim at measuring, in parallel, the concentration of up to several proteins or non-coding RNA molecules, in samples down to the single cell level, following the real time concentrations of several biomarkers in patient-derived samples down to femto-molar concentrations, and single-circulating-tumor-cell resolution. We plan to develop our program a) by applying the principles and practice of intrinsically differential measurements, e.g. by building a self-assembled nano-devices that provide robust outputs measurable with topographic AFM imaging, electrochemical measurements, or gel electrophoresis, and b) by using the vertical equivalent of cantilever oscillators (pillars) that we plan to use as quartz “microbalances” that are 10,000 time more sensitive than cantilevers w. r. t. measurable min. mass and 100 time more sensitive w. r. t. dilution. The proposal’s core strategy is to exploit the PI’s expertise in innovative instrumentation and his integrated physical chemistry know-how, leading a highly multi-disciplinary staff to closely interact with first class medical staff in hospital settings to solve, and validate the solution of, relevant medical problems by generating innovative and versatile sensing devices. For instance, the sensitivity of our sensors will allow protein or miRNA analysis from very small and homogeneous samples of tumor cells, as well as the ease identification circulating tumor cells (CTCs) for, in addition to improved cancer diagnostics, also the prediction of patient response to treatment. The convergence between chemistry and biology, through nanotechnology, physics and computational approaches, with medical diagnostics will enable our team to come up with more versatile and reliable diagnostic tools, while stimulating fundamental research in fields as diverse as stem cells differentiation and the study of cell physiopathology.
Summary
The main goal of this proposal is to introduce innovative devices and protocols (based on nano- manipulation, the response of micro-(nano-)mechanical oscillators and nano-fluidics) to carry out, precise, high throughput, high sensitivity, and low cost interactomic measurements. We aim at measuring, in parallel, the concentration of up to several proteins or non-coding RNA molecules, in samples down to the single cell level, following the real time concentrations of several biomarkers in patient-derived samples down to femto-molar concentrations, and single-circulating-tumor-cell resolution. We plan to develop our program a) by applying the principles and practice of intrinsically differential measurements, e.g. by building a self-assembled nano-devices that provide robust outputs measurable with topographic AFM imaging, electrochemical measurements, or gel electrophoresis, and b) by using the vertical equivalent of cantilever oscillators (pillars) that we plan to use as quartz “microbalances” that are 10,000 time more sensitive than cantilevers w. r. t. measurable min. mass and 100 time more sensitive w. r. t. dilution. The proposal’s core strategy is to exploit the PI’s expertise in innovative instrumentation and his integrated physical chemistry know-how, leading a highly multi-disciplinary staff to closely interact with first class medical staff in hospital settings to solve, and validate the solution of, relevant medical problems by generating innovative and versatile sensing devices. For instance, the sensitivity of our sensors will allow protein or miRNA analysis from very small and homogeneous samples of tumor cells, as well as the ease identification circulating tumor cells (CTCs) for, in addition to improved cancer diagnostics, also the prediction of patient response to treatment. The convergence between chemistry and biology, through nanotechnology, physics and computational approaches, with medical diagnostics will enable our team to come up with more versatile and reliable diagnostic tools, while stimulating fundamental research in fields as diverse as stem cells differentiation and the study of cell physiopathology.
Max ERC Funding
2 979 700 €
Duration
Start date: 2011-07-01, End date: 2016-06-30
Project acronym REPROPARK
Project New experimental therapeutic approaches for Parkinson’s disease by direct DA neuronal reprogramming
Researcher (PI) Vania Broccoli
Host Institution (HI) OSPEDALE SAN RAFFAELE SRL
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary Neurodegenerative diseases cause a significant burden on the elderly population in Europe. Parkinson’s disease (PD) affects 1.2 million people in Europe and with the increasing life expectancy this number will rise, putting more pressure on health care. Treatment of PD is only symptomatic, and therefore, there is an urgent need for more efficient therapies. Degeneration of mesencephalic DA neurons triggers the initial phases of PD, which raises the concept that cell replacement might represent a long-term restorative option for this neuropathology. Indeed, previous studies in PD patients have indicated that cell therapy has the potential to significantly sustain an enduring symptomatic relief. However, these studies suffered for lacking an ideal source of transplantable human DA neurons. Only recently the generation of induced stem cells (iPSCs) by the reprogramming of somatic cells has disclosed the possibility to generate individual specific neurons with a high therapeutic potential. We have recently developed a methodology that promotes transdifferentiation of mouse and human fibroblasts into functional induced dopaminergic neuronal (iDAN) cells. iDAN cells display sophisticated neuronal properties including pacemaking firing activity, synaptic integration, activity-dependent dopamine release and D2 functional autoreceptors. Therefore, iDAN cells offer an unprecedented cellular source with ideal features for cell therapy in PD, since they can be generated from the patients in high amounts. Here, we propose to strengthen the technology in the human setting and decipher the molecular events. Next, we will elaborate methods of in vivo reprogramming promoting neuronal transdifferentiation locally in the mouse brain. Finally, autologous transplantations of iDAN cells in parkinsonian monkeys will be attempted. Overall, this project will enhance cell reprogramming technologies with the ambition to generate a superior cellular source for transplantations in PD patients.
Summary
Neurodegenerative diseases cause a significant burden on the elderly population in Europe. Parkinson’s disease (PD) affects 1.2 million people in Europe and with the increasing life expectancy this number will rise, putting more pressure on health care. Treatment of PD is only symptomatic, and therefore, there is an urgent need for more efficient therapies. Degeneration of mesencephalic DA neurons triggers the initial phases of PD, which raises the concept that cell replacement might represent a long-term restorative option for this neuropathology. Indeed, previous studies in PD patients have indicated that cell therapy has the potential to significantly sustain an enduring symptomatic relief. However, these studies suffered for lacking an ideal source of transplantable human DA neurons. Only recently the generation of induced stem cells (iPSCs) by the reprogramming of somatic cells has disclosed the possibility to generate individual specific neurons with a high therapeutic potential. We have recently developed a methodology that promotes transdifferentiation of mouse and human fibroblasts into functional induced dopaminergic neuronal (iDAN) cells. iDAN cells display sophisticated neuronal properties including pacemaking firing activity, synaptic integration, activity-dependent dopamine release and D2 functional autoreceptors. Therefore, iDAN cells offer an unprecedented cellular source with ideal features for cell therapy in PD, since they can be generated from the patients in high amounts. Here, we propose to strengthen the technology in the human setting and decipher the molecular events. Next, we will elaborate methods of in vivo reprogramming promoting neuronal transdifferentiation locally in the mouse brain. Finally, autologous transplantations of iDAN cells in parkinsonian monkeys will be attempted. Overall, this project will enhance cell reprogramming technologies with the ambition to generate a superior cellular source for transplantations in PD patients.
Max ERC Funding
2 415 767 €
Duration
Start date: 2014-06-01, End date: 2019-05-31
Project acronym RESET
Project Dreaming of no more renal dialysis: how self-derived tissue and cells can replace renal function
Researcher (PI) Giuseppe Remuzzi
Host Institution (HI) ISTITUTO DI RICERCHE FARMACOLOGICHE MARIO NEGRI
Call Details Advanced Grant (AdG), LS7, ERC-2010-AdG_20100317
Summary For chronic kidney diseases there is little chance that the vast majority of the world¿s population will have access to renal replacement therapy with dialysis. There is yet no adequate alternative with curative intent than allo-transplantation. This approach is seriously impaired by eventually limited graft survival and by the scarce availability of donors. We propose an innovative strategy that would help to simultaneously overwhelm the reduced access to dialysis, the need for organs for transplantation, the avoidance of patient exposure to immunosuppressants. The overall focus is based on the idea of generating new kidneys by tissue engineering technologies starting from a whole-kidney scaffold with intact three-dimensional geometry and vasculature created by de-cellularization of a kidney harvested from the patient with progressive chronic kidney disease (CKD). Repopulation of the kidney scaffold could be achieved with patient-specific induced pluripotent stem (iPS) cells generated by reprogramming to a pluripotent status of somatic cells. After regeneration, the kidney will be transplanted in the same patient. Thereafter, the same procedures will be repeated with the contralateral native injured kidney, after in vivo assessment of the proper functional performance of the newly generated transplanted organ.
This goal will be pursued through the three steps introductory to the development of the best strategy for translating the proposed frontier research to patients with CKD:
1.Create an intact whole-kidney scaffold by de-cellularization of a rodent kidney and validate the
procedure with a human kidney.
2. Attempt to reconstruct the organ by reseeding the rodent and human whole-kidney scaffolds
with iPS cells generated by reprogramming adult dermal fibroblasts to a pluripotent status.
3. Deeply characterize the function of the new rodent and human kidneys by in vivo and in vitro
techniques.
Summary
For chronic kidney diseases there is little chance that the vast majority of the world¿s population will have access to renal replacement therapy with dialysis. There is yet no adequate alternative with curative intent than allo-transplantation. This approach is seriously impaired by eventually limited graft survival and by the scarce availability of donors. We propose an innovative strategy that would help to simultaneously overwhelm the reduced access to dialysis, the need for organs for transplantation, the avoidance of patient exposure to immunosuppressants. The overall focus is based on the idea of generating new kidneys by tissue engineering technologies starting from a whole-kidney scaffold with intact three-dimensional geometry and vasculature created by de-cellularization of a kidney harvested from the patient with progressive chronic kidney disease (CKD). Repopulation of the kidney scaffold could be achieved with patient-specific induced pluripotent stem (iPS) cells generated by reprogramming to a pluripotent status of somatic cells. After regeneration, the kidney will be transplanted in the same patient. Thereafter, the same procedures will be repeated with the contralateral native injured kidney, after in vivo assessment of the proper functional performance of the newly generated transplanted organ.
This goal will be pursued through the three steps introductory to the development of the best strategy for translating the proposed frontier research to patients with CKD:
1.Create an intact whole-kidney scaffold by de-cellularization of a rodent kidney and validate the
procedure with a human kidney.
2. Attempt to reconstruct the organ by reseeding the rodent and human whole-kidney scaffolds
with iPS cells generated by reprogramming adult dermal fibroblasts to a pluripotent status.
3. Deeply characterize the function of the new rodent and human kidneys by in vivo and in vitro
techniques.
Max ERC Funding
2 493 000 €
Duration
Start date: 2011-05-01, End date: 2016-04-30
Project acronym SCOPE
Project Schwann Cell Options for chronic Pain Eradication
Researcher (PI) Pierangelo GEPPETTI
Host Institution (HI) UNIVERSITA DEGLI STUDI DI FIRENZE
Call Details Advanced Grant (AdG), LS7, ERC-2018-ADG
Summary Chronic pain, characterized by increased sensitivity to innocuous/mild stimuli (allodynia), afflicts 25% of the European adult population. Efficacy and/or safety of analgesic medicines is limited, and the treatment of chronic pain associated with inflammation, peripheral and central neuropathies and cancer remains unsatisfactory. Thus, identification of novel targets for better and safer analgesics is a major medical need. Transient receptor potential ankyrin 1 (TRPA1) channel, expressed by a subpopulation of primary sensory neurons (nociceptors), has been proposed as a major transducer of acute pain. We have, recently, identified that TRPA1 is expressed in Schwann cells that ensheath peripheral nerve fibres. In a prototypical model of neuropathic pain (sciatic nerve ligation in mice), we discovered that Schwann cell-TRPA1 exerts a hitherto unknown role that, via amplification of the oxidative stress message, sustains neuroinflammation and chronic pain (allodynia). Thus, Schwann cells, through their own repertoire of channels and enzymes orchestrate in the injured/inflamed tissue an autocrine/paracrine signalling pathway to sustain chronic pain. The purpose of the present project is to extend this observation to other models of inflammatory, neuropathic and cancer pain to identify a general paradigm based on Schwann cell/TRPA1/oxidative stress as the pathway that sustains chronic pain. We aim also at identifying in oligodendrocytes (the Schwann cells of the brain) whether the TRPA1/oxidative stress pathway sustains pain in the central nervous system. In mouse, rat and human Schwann cells/oligodendrocytes we aim at identifying biomarkers and combine them into biosignatures predictive of the susceptibility to the development of chronic pain. We anticipate that each molecular step that entails the TRPA1/oxidative stress pathway in Schwann cell lineages is an eligible target for discovering new effective and safer medicines for the treatment of chronic pain.
Summary
Chronic pain, characterized by increased sensitivity to innocuous/mild stimuli (allodynia), afflicts 25% of the European adult population. Efficacy and/or safety of analgesic medicines is limited, and the treatment of chronic pain associated with inflammation, peripheral and central neuropathies and cancer remains unsatisfactory. Thus, identification of novel targets for better and safer analgesics is a major medical need. Transient receptor potential ankyrin 1 (TRPA1) channel, expressed by a subpopulation of primary sensory neurons (nociceptors), has been proposed as a major transducer of acute pain. We have, recently, identified that TRPA1 is expressed in Schwann cells that ensheath peripheral nerve fibres. In a prototypical model of neuropathic pain (sciatic nerve ligation in mice), we discovered that Schwann cell-TRPA1 exerts a hitherto unknown role that, via amplification of the oxidative stress message, sustains neuroinflammation and chronic pain (allodynia). Thus, Schwann cells, through their own repertoire of channels and enzymes orchestrate in the injured/inflamed tissue an autocrine/paracrine signalling pathway to sustain chronic pain. The purpose of the present project is to extend this observation to other models of inflammatory, neuropathic and cancer pain to identify a general paradigm based on Schwann cell/TRPA1/oxidative stress as the pathway that sustains chronic pain. We aim also at identifying in oligodendrocytes (the Schwann cells of the brain) whether the TRPA1/oxidative stress pathway sustains pain in the central nervous system. In mouse, rat and human Schwann cells/oligodendrocytes we aim at identifying biomarkers and combine them into biosignatures predictive of the susceptibility to the development of chronic pain. We anticipate that each molecular step that entails the TRPA1/oxidative stress pathway in Schwann cell lineages is an eligible target for discovering new effective and safer medicines for the treatment of chronic pain.
Max ERC Funding
2 185 921 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym TARGETINGGENETHERAPY
Project Towards Safe and Effective Hematopoietic Stem Cell Gene Therapy: Targeting Integration to Genomic Safe Harbors and Exploiting Endogenous microRNA to Regulate Transgene Expression
Researcher (PI) Luigi Naldini
Host Institution (HI) UNIVERSITA VITA-SALUTE SAN RAFFAELE
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary Hematopoietic stem cell gene therapy has a tremendous potential to treat human disease. Yet, in conjunction with the first successful results in the clinic, severe adverse events linked to the gene transfer protocol were reported. Recently, we provided proof-of-principle of two new powerful strategies to improve the efficacy and safety of gene transfer: 1) regulating transgene expression by exploiting cellular microRNAs; 2) targeting integration at predetermined sites of the genome by forcing homologous recombination with designer Zinc finger nucleases. Here we will investigate the microRNA network regulating hematopoiesis and exploit the new knowledge to develop vectors with stringently controlled expression throughout the hematopoietic lineages. We will develop Zinc finger nuclease-based vectors that insert the transgene with high efficiency and specificity either downstream to its own endogenous promoter or into a safe genomic harbor that allows for robust expression without interference on the neighboring genes. By combining these strategies we will provide radically improved gene transfer platforms. Furthermore, we will exploit these technologies for the generation and genetic correction of induced pluripotent stem cells, providing a potentially unlimited source of patient-derived vector free gene corrected multipotent stem cells for future applications of regenerative medicine. The new gene therapy strategies will be tested in pre-clinical models of leukodystrophies and immunodeficiencies, for which we have extensive experience, and should enter a clinical trial for at least one such disease by the end of the proposed funding period. If successfully validated, the new strategies may eventually broaden the scope of gene therapy in medicine.
Summary
Hematopoietic stem cell gene therapy has a tremendous potential to treat human disease. Yet, in conjunction with the first successful results in the clinic, severe adverse events linked to the gene transfer protocol were reported. Recently, we provided proof-of-principle of two new powerful strategies to improve the efficacy and safety of gene transfer: 1) regulating transgene expression by exploiting cellular microRNAs; 2) targeting integration at predetermined sites of the genome by forcing homologous recombination with designer Zinc finger nucleases. Here we will investigate the microRNA network regulating hematopoiesis and exploit the new knowledge to develop vectors with stringently controlled expression throughout the hematopoietic lineages. We will develop Zinc finger nuclease-based vectors that insert the transgene with high efficiency and specificity either downstream to its own endogenous promoter or into a safe genomic harbor that allows for robust expression without interference on the neighboring genes. By combining these strategies we will provide radically improved gene transfer platforms. Furthermore, we will exploit these technologies for the generation and genetic correction of induced pluripotent stem cells, providing a potentially unlimited source of patient-derived vector free gene corrected multipotent stem cells for future applications of regenerative medicine. The new gene therapy strategies will be tested in pre-clinical models of leukodystrophies and immunodeficiencies, for which we have extensive experience, and should enter a clinical trial for at least one such disease by the end of the proposed funding period. If successfully validated, the new strategies may eventually broaden the scope of gene therapy in medicine.
Max ERC Funding
2 500 000 €
Duration
Start date: 2010-05-01, End date: 2016-01-31
Project acronym TRANSLATE
Project Noncoding and Translational Modulation of Gene Expression and Epigenetic Changes
Researcher (PI) Stefano Biffo
Host Institution (HI) UNIVERSITA DEGLI STUDI DI MILANO
Call Details Advanced Grant (AdG), LS7, ERC-2013-ADG
Summary "Gene expression studies rely on high throughput techniques, which do not take in account conceptual limits. I will overcome this situation by exploiting two biological facts. First, RNAs that are important in tissue function are a subset of the global mass, but are always associated with the ribosomal machinery and as such should be identified. Second, gene expression is the outcome of dynamic fluctuations that with time create a unique expression pattern. We need to dynamically label cell populations that undergo stress and follow them to generate a gene expression signature. To achieve my goal, I will consider: 1. Translational stress generated by viral infection or accumulation of misfolded proteins; 2. human CD4+ T lymphocyte subsets which are key to orchestrate immune responses; 3. EIF6 model of metabolic reprogramming.
1. Activation of eIF2alpha phosphorylation by viral infection generates a translational response in which silent mRNAs containing upstream ORFs (uORF) are translated. I will exploit this observation to construct the first in vivo reporter model of translational stress. We will label genetically cells that have translational stress, to identify all the changes that a single cell undergoes after viral infection/accumulation of undegraded proteins.
2. I will selectively sequence for the first time mRNAs and ncRNAs associated with the ribosomal machinery in human cells with a defined functional status.
3. Spectacular data have shown that translation factor eIF6 regulates tumorigenesis by inducing a profound metabolic reprogramming. This observation suggests that, in vivo, translation acts upstream of transcription. We will model how a short translational input results in a complex epigenetic change.
Significance: a revolution in finding biomarkers/drug targets. Generate a map of predictors of the process from stress to disease. Dscriminate biologically active sequences from background. Define how transient translation reshapes gene expression."
Summary
"Gene expression studies rely on high throughput techniques, which do not take in account conceptual limits. I will overcome this situation by exploiting two biological facts. First, RNAs that are important in tissue function are a subset of the global mass, but are always associated with the ribosomal machinery and as such should be identified. Second, gene expression is the outcome of dynamic fluctuations that with time create a unique expression pattern. We need to dynamically label cell populations that undergo stress and follow them to generate a gene expression signature. To achieve my goal, I will consider: 1. Translational stress generated by viral infection or accumulation of misfolded proteins; 2. human CD4+ T lymphocyte subsets which are key to orchestrate immune responses; 3. EIF6 model of metabolic reprogramming.
1. Activation of eIF2alpha phosphorylation by viral infection generates a translational response in which silent mRNAs containing upstream ORFs (uORF) are translated. I will exploit this observation to construct the first in vivo reporter model of translational stress. We will label genetically cells that have translational stress, to identify all the changes that a single cell undergoes after viral infection/accumulation of undegraded proteins.
2. I will selectively sequence for the first time mRNAs and ncRNAs associated with the ribosomal machinery in human cells with a defined functional status.
3. Spectacular data have shown that translation factor eIF6 regulates tumorigenesis by inducing a profound metabolic reprogramming. This observation suggests that, in vivo, translation acts upstream of transcription. We will model how a short translational input results in a complex epigenetic change.
Significance: a revolution in finding biomarkers/drug targets. Generate a map of predictors of the process from stress to disease. Dscriminate biologically active sequences from background. Define how transient translation reshapes gene expression."
Max ERC Funding
2 496 000 €
Duration
Start date: 2014-05-01, End date: 2019-04-30
Project acronym TREAT-NPM1-AML
Project Improving therapy of NPM1-mutated AML
Researcher (PI) Brunangelo FALINI
Host Institution (HI) UNIVERSITA DEGLI STUDI DI PERUGIA
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Acute myeloid leukemia (AML) is the most common acute leukemia in adults accounting for approximately 15,000 new cases/year in Europe and 20,000 new cases/year in US. Currently, 40-50% of AML patients (age 18-60 years) and only 5-10% of older patients (who are usually more frequently affected by the disease) can be cured using conventional chemotherapy +/- allogeneic hematopoietic stem cell transplantation. Thus, AML still remains an urgent medical need which calls for new forms of molecular targeted therapies (similarly to those available for acute promyelocytic leukemia). The P.I. previously discovered the nucleophosmin (NPM1) mutations, the most common genetic lesion in AML (about one-third of cases) and gave fundamental contributions in the translation of this seminal discovery into the clinic (improved classification of myeloid neoplasms according to WHO, genetic-based risk- stratification of AML patients, monitoring of minimal residual disease and first demonstration of the anti-leukemic activity of actinomycin D). The present research proposal is focused on improving therapy of NPM1-mutated AML. Specifically, it is aimed to: i) identify novel chemical tools interfering with NPM1 functions by interacting with the N-terminal portion of the protein (objective 1); ii) conduct a clinical trial (AML-PG02; Eudract 2014-003490-41) with actinomycin D in older untreated and/or unfit patients with NPM1-mutated AML and to better understand in vitro and in mice models the mechanisms of action of this drug, used alone or in combination with other agents (objective 2); iii) develop compound mouse models aimed to investigate how NPM1 mutations cooperate with FLT3-ITD or DNMT3A mutations in promoting AML with the goal to better understand the characteristics of relapsed cases and to design new therapeutic strategies (objectives 3 and 4): and iv) generate a murine model for testing the feasibility of “in situ” vaccination in AML, especially in NPM1-mutated AML (objective 5).
Summary
Acute myeloid leukemia (AML) is the most common acute leukemia in adults accounting for approximately 15,000 new cases/year in Europe and 20,000 new cases/year in US. Currently, 40-50% of AML patients (age 18-60 years) and only 5-10% of older patients (who are usually more frequently affected by the disease) can be cured using conventional chemotherapy +/- allogeneic hematopoietic stem cell transplantation. Thus, AML still remains an urgent medical need which calls for new forms of molecular targeted therapies (similarly to those available for acute promyelocytic leukemia). The P.I. previously discovered the nucleophosmin (NPM1) mutations, the most common genetic lesion in AML (about one-third of cases) and gave fundamental contributions in the translation of this seminal discovery into the clinic (improved classification of myeloid neoplasms according to WHO, genetic-based risk- stratification of AML patients, monitoring of minimal residual disease and first demonstration of the anti-leukemic activity of actinomycin D). The present research proposal is focused on improving therapy of NPM1-mutated AML. Specifically, it is aimed to: i) identify novel chemical tools interfering with NPM1 functions by interacting with the N-terminal portion of the protein (objective 1); ii) conduct a clinical trial (AML-PG02; Eudract 2014-003490-41) with actinomycin D in older untreated and/or unfit patients with NPM1-mutated AML and to better understand in vitro and in mice models the mechanisms of action of this drug, used alone or in combination with other agents (objective 2); iii) develop compound mouse models aimed to investigate how NPM1 mutations cooperate with FLT3-ITD or DNMT3A mutations in promoting AML with the goal to better understand the characteristics of relapsed cases and to design new therapeutic strategies (objectives 3 and 4): and iv) generate a murine model for testing the feasibility of “in situ” vaccination in AML, especially in NPM1-mutated AML (objective 5).
Max ERC Funding
2 895 836 €
Duration
Start date: 2017-11-01, End date: 2022-10-31