Project acronym A-FRO
Project Actively Frozen - contextual modulation of freezing and its neuronal basis
Researcher (PI) Marta de Aragão Pacheco Moita
Host Institution (HI) FUNDACAO D. ANNA SOMMER CHAMPALIMAUD E DR. CARLOS MONTEZ CHAMPALIMAUD
Call Details Consolidator Grant (CoG), LS5, ERC-2018-COG
Summary When faced with a threat, an animal must decide whether to freeze, reducing its chances of being noticed, or to flee to the safety of a refuge. Animals from fish to primates choose between these two alternatives when confronted by an attacking predator, a choice that largely depends on the context in which the threat occurs. Recent work has made strides identifying the pre-motor circuits, and their inputs, which control freezing behavior in rodents, but how contextual information is integrated to guide this choice is still far from understood. We recently found that fruit flies in response to visual looming stimuli, simulating a large object on collision course, make rapid freeze/flee choices that depend on the social and spatial environment, and the fly’s internal state. Further, identification of looming detector neurons was recently reported and we identified the descending command neurons, DNp09, responsible for freezing in the fly. Knowing the sensory input and descending output for looming-evoked freezing, two environmental factors that modulate its expression, and using a genetically tractable system affording the use of large sample sizes, places us in an unique position to understand how a information about a threat is integrated with cues from the environment to guide the choice of whether to freeze (our goal). To assess how social information impinges on the circuit for freezing, we will examine the sensory inputs and neuromodulators that mediate this process, mapping their connections to DNp09 neurons (Aim 1). We ask whether learning is required for the spatial modulation of freezing, which cues flies are using to discriminate different places and which brain circuits mediate this process (Aim 2). Finally, we will study how activity of DNp09 neurons drives freezing (Aim 3). This project will provide a comprehensive understanding of the mechanism of freezing and its modulation by the environment, from single neurons to behaviour.
Summary
When faced with a threat, an animal must decide whether to freeze, reducing its chances of being noticed, or to flee to the safety of a refuge. Animals from fish to primates choose between these two alternatives when confronted by an attacking predator, a choice that largely depends on the context in which the threat occurs. Recent work has made strides identifying the pre-motor circuits, and their inputs, which control freezing behavior in rodents, but how contextual information is integrated to guide this choice is still far from understood. We recently found that fruit flies in response to visual looming stimuli, simulating a large object on collision course, make rapid freeze/flee choices that depend on the social and spatial environment, and the fly’s internal state. Further, identification of looming detector neurons was recently reported and we identified the descending command neurons, DNp09, responsible for freezing in the fly. Knowing the sensory input and descending output for looming-evoked freezing, two environmental factors that modulate its expression, and using a genetically tractable system affording the use of large sample sizes, places us in an unique position to understand how a information about a threat is integrated with cues from the environment to guide the choice of whether to freeze (our goal). To assess how social information impinges on the circuit for freezing, we will examine the sensory inputs and neuromodulators that mediate this process, mapping their connections to DNp09 neurons (Aim 1). We ask whether learning is required for the spatial modulation of freezing, which cues flies are using to discriminate different places and which brain circuits mediate this process (Aim 2). Finally, we will study how activity of DNp09 neurons drives freezing (Aim 3). This project will provide a comprehensive understanding of the mechanism of freezing and its modulation by the environment, from single neurons to behaviour.
Max ERC Funding
1 969 750 €
Duration
Start date: 2019-02-01, End date: 2024-01-31
Project acronym bECOMiNG
Project spontaneous Evolution and Clonal heterOgeneity in MoNoclonal Gammopathies: from mechanisms of progression to clinical management
Researcher (PI) Niccolo Bolli
Host Institution (HI) UNIVERSITA DEGLI STUDI DI MILANO
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary As an onco-hematologist with a strong expertise in genomics, I significantly contributed to the understanding of multiple myeloma (MM) heterogeneity and its evolution over time, driven by genotypic and phenotypic features carried by different subpopulations of cells. MM is preceded by prevalent, asymptomatic stages that may evolve with variable frequency, not accurately captured by current clinical prognostic scores. Supported by preliminary data, my hypothesis is that the same heterogeneity is present early on the disease course, and identification of the biological determinants of evolution at this stage will allow better prediction of its evolutionary trajectory, if not its control. In this proposal I will therefore make a sharp change from conventional approaches and move to early stages of MM using unique retrospective sample cohorts and ambitious prospective sampling. To identify clonal MM cells in the elderly before a monoclonal gammopathy can be detected, I will collect bone marrow (BM) from hundreds of hip replacement specimens, and analyze archive peripheral blood samples of thousands of healthy individuals with years of annotated clinical follow-up. This will identify early genomic alterations that are permissive to disease initiation/evolution and may serve as biomarkers for clinical screening. Through innovative, integrated single-cell genotyping and phenotyping of hundreds of asymptomatic MMs, I will functionally dissect heterogeneity and characterize the BM microenvironment to look for determinants of disease progression. Correlation with clinical outcome and mini-invasive serial sampling of circulating cell-free DNA will identify candidate biological markers to better predict evolution. Last, aggressive modelling of candidate early lesions and modifier screens will offer a list of vulnerabilities that could be exploited for rationale therapies. These methodologies will deliver a paradigm for the use of molecularly-driven precision medicine in cancer.
Summary
As an onco-hematologist with a strong expertise in genomics, I significantly contributed to the understanding of multiple myeloma (MM) heterogeneity and its evolution over time, driven by genotypic and phenotypic features carried by different subpopulations of cells. MM is preceded by prevalent, asymptomatic stages that may evolve with variable frequency, not accurately captured by current clinical prognostic scores. Supported by preliminary data, my hypothesis is that the same heterogeneity is present early on the disease course, and identification of the biological determinants of evolution at this stage will allow better prediction of its evolutionary trajectory, if not its control. In this proposal I will therefore make a sharp change from conventional approaches and move to early stages of MM using unique retrospective sample cohorts and ambitious prospective sampling. To identify clonal MM cells in the elderly before a monoclonal gammopathy can be detected, I will collect bone marrow (BM) from hundreds of hip replacement specimens, and analyze archive peripheral blood samples of thousands of healthy individuals with years of annotated clinical follow-up. This will identify early genomic alterations that are permissive to disease initiation/evolution and may serve as biomarkers for clinical screening. Through innovative, integrated single-cell genotyping and phenotyping of hundreds of asymptomatic MMs, I will functionally dissect heterogeneity and characterize the BM microenvironment to look for determinants of disease progression. Correlation with clinical outcome and mini-invasive serial sampling of circulating cell-free DNA will identify candidate biological markers to better predict evolution. Last, aggressive modelling of candidate early lesions and modifier screens will offer a list of vulnerabilities that could be exploited for rationale therapies. These methodologies will deliver a paradigm for the use of molecularly-driven precision medicine in cancer.
Max ERC Funding
1 998 781 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym CANCEREVO
Project Deciphering and predicting the evolution of cancer cell populations
Researcher (PI) Marco Helmut GERLINGER
Host Institution (HI) THE INSTITUTE OF CANCER RESEARCH: ROYAL CANCER HOSPITAL
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary The fundamental evolutionary nature of cancer is well recognized but an understanding of the dynamic evolutionary changes occurring throughout a tumour’s lifetime and their clinical implications is in its infancy. Current approaches to reveal cancer evolution by sequencing of multiple biopsies remain of limited use in the clinic due to sample access problems in multi-metastatic disease. Circulating tumour DNA (ctDNA) is thought to comprehensively sample subclones across metastatic sites. However, available technologies either have high sensitivity but are restricted to the analysis of small gene panels or they allow sequencing of large target regions such as exomes but with too limited sensitivity to detect rare subclones. We developed a novel error corrected sequencing technology that will be applied to perform deep exome sequencing on longitudinal ctDNA samples from highly heterogeneous metastatic gastro-oesophageal carcinomas. This will track the evolution of the entire cancer population over the lifetime of these tumours, from metastatic disease over drug therapy to end-stage disease and enable ground breaking insights into cancer population evolution rules and mechanisms. Specifically, we will: 1. Define the genomic landscape and drivers of metastatic and end stage disease. 2. Understand the rules of cancer evolutionary dynamics of entire cancer cell populations. 3. Predict cancer evolution and define the limits of predictability. 4. Rapidly identify drug resistance mechanisms to chemo- and immunotherapy based on signals of Darwinian selection such as parallel and convergent evolution. Our sequencing technology and analysis framework will also transform the way cancer evolution metrics can be accessed and interpreted in the clinic which will have major impacts, ranging from better biomarkers to predict cancer evolution to the identification of drug targets that drive disease progression and therapy resistance.
Summary
The fundamental evolutionary nature of cancer is well recognized but an understanding of the dynamic evolutionary changes occurring throughout a tumour’s lifetime and their clinical implications is in its infancy. Current approaches to reveal cancer evolution by sequencing of multiple biopsies remain of limited use in the clinic due to sample access problems in multi-metastatic disease. Circulating tumour DNA (ctDNA) is thought to comprehensively sample subclones across metastatic sites. However, available technologies either have high sensitivity but are restricted to the analysis of small gene panels or they allow sequencing of large target regions such as exomes but with too limited sensitivity to detect rare subclones. We developed a novel error corrected sequencing technology that will be applied to perform deep exome sequencing on longitudinal ctDNA samples from highly heterogeneous metastatic gastro-oesophageal carcinomas. This will track the evolution of the entire cancer population over the lifetime of these tumours, from metastatic disease over drug therapy to end-stage disease and enable ground breaking insights into cancer population evolution rules and mechanisms. Specifically, we will: 1. Define the genomic landscape and drivers of metastatic and end stage disease. 2. Understand the rules of cancer evolutionary dynamics of entire cancer cell populations. 3. Predict cancer evolution and define the limits of predictability. 4. Rapidly identify drug resistance mechanisms to chemo- and immunotherapy based on signals of Darwinian selection such as parallel and convergent evolution. Our sequencing technology and analysis framework will also transform the way cancer evolution metrics can be accessed and interpreted in the clinic which will have major impacts, ranging from better biomarkers to predict cancer evolution to the identification of drug targets that drive disease progression and therapy resistance.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym DNA Funs
Project DNA-based functional lattices
Researcher (PI) Tim LIEDL
Host Institution (HI) LUDWIG-MAXIMILIANS-UNIVERSITAET MUENCHEN
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary Nature has evolved astonishingly diverse structures where the nanoscale assembly of components is key to their functionality. Such nanostructures self-assemble at massive scales and at spatial resolutions surpassing top-down production techniques. The leaves of a single tree, e.g., can cover the area of 10.000 m^2 while every mm^2 contains more than 10^8 highly efficient light-harvesting complexes. For future photovoltaic devices, light-managing surfaces and photonic devices it will thus be beneficial to adopt principles of self-assembly. Advances in design and low-cost production of DNA nanostructures allow us to challenge nature. By combining the assembly power of bottom-up DNA origami with top-down lithography it will be possible to fabricate functional nanostructured materials designed on the molecular level while reaching macroscopic dimensions.
With the goal to boost energy conversion rates, I will design DNA structures that grow from pre-patterned surfaces and assemble into interpenetrating 3D networks that exhibit the highest possible contact area for electron donor and acceptor molecules in organic photovoltaic devices. Spectral tuning through carefully designed dye arrangements will complement these efforts.
Custom-tailored photonic crystals built from lattices of DNA origami structures will control the flow of light. By incorporating dynamic DNA reconfigurability and colloidal nanoparticles at freely chosen positions, intelligent materials that respond to external cues such as light or heat are projected.
Positioning accuracy of 1 nm renders possible the emergence of so-called “Dirac plasmons” in DNA-assembled particle lattices. Such topologically protected states are sought after for the coherent and loss-less propagation of energy and information in next-generation all-optical circuits.
These approaches have the potential to reduce production costs and increase efficiencies of light-harvesting devices, intelligent surfaces and future computing devices.
Summary
Nature has evolved astonishingly diverse structures where the nanoscale assembly of components is key to their functionality. Such nanostructures self-assemble at massive scales and at spatial resolutions surpassing top-down production techniques. The leaves of a single tree, e.g., can cover the area of 10.000 m^2 while every mm^2 contains more than 10^8 highly efficient light-harvesting complexes. For future photovoltaic devices, light-managing surfaces and photonic devices it will thus be beneficial to adopt principles of self-assembly. Advances in design and low-cost production of DNA nanostructures allow us to challenge nature. By combining the assembly power of bottom-up DNA origami with top-down lithography it will be possible to fabricate functional nanostructured materials designed on the molecular level while reaching macroscopic dimensions.
With the goal to boost energy conversion rates, I will design DNA structures that grow from pre-patterned surfaces and assemble into interpenetrating 3D networks that exhibit the highest possible contact area for electron donor and acceptor molecules in organic photovoltaic devices. Spectral tuning through carefully designed dye arrangements will complement these efforts.
Custom-tailored photonic crystals built from lattices of DNA origami structures will control the flow of light. By incorporating dynamic DNA reconfigurability and colloidal nanoparticles at freely chosen positions, intelligent materials that respond to external cues such as light or heat are projected.
Positioning accuracy of 1 nm renders possible the emergence of so-called “Dirac plasmons” in DNA-assembled particle lattices. Such topologically protected states are sought after for the coherent and loss-less propagation of energy and information in next-generation all-optical circuits.
These approaches have the potential to reduce production costs and increase efficiencies of light-harvesting devices, intelligent surfaces and future computing devices.
Max ERC Funding
1 997 500 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym EngineeringBAP
Project Engineering brain activity patterns for therapeutics of neuropsychiatric and neurological disorders
Researcher (PI) Mehmet Fatih YANIK
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Consolidator Grant (CoG), LS5, ERC-2018-COG
Summary Neuropsychiatric and neurological disorders are complex dysfunctions of neuronal circuits. Their treatment
has been limited by the lack of non-invasive methods for measuring the underlying circuit dysfunctions, and
for direct and localized modifications of these circuits. We propose minimally invasive technologies for
measuring brain activity and functional connectivity patterns, and for manipulating them directly in vivo to
correct the abnormal behavioural phenotypes (in rodents with potential scalability to non-human primates and
humans). First, we present a proof-of-principle study on mutant zebrafish, in which we correct whole-brain
level abnormal activity patterns and behaviours by using large-scale single-neuron resolution measurements,
and by simultaneously modulating multiple sub-networks via neuromodulator cocktails. Next, we present
strong preliminary data in rodents and our plan: (1) For manipulating brain circuits in rodents/primates noninvasively,
we will develop technologies that can deliver receptive-specific neuromodulators to spatially
precise brain targets without opening/damaging the blood brain barrier. These methods will employ engineered
ultrasound pulses and drug carrying microparticles we designed. (2) For reading out the brain circuits in
rodents/primates, we will develop flexible low-power neuromorphic μECoG circuits that can detect single
neuron signals from superficial cortical layers of many cortical areas simultaneously. (3) Finally, these novel
technologies will be comprehensively evaluated on a mouse model of obsessive compulsivity and anxiety
using a battery of behavioural tasks to reverse the pathological symptoms (beyond what is achievable by
existing approaches). This project constitutes a major step towards the development and testing of minimallyinvasive
and high-precision technologies for manipulating brain activity patterns, which can impact both our
understanding of the brain and treatment of intractable brain disorders.
Summary
Neuropsychiatric and neurological disorders are complex dysfunctions of neuronal circuits. Their treatment
has been limited by the lack of non-invasive methods for measuring the underlying circuit dysfunctions, and
for direct and localized modifications of these circuits. We propose minimally invasive technologies for
measuring brain activity and functional connectivity patterns, and for manipulating them directly in vivo to
correct the abnormal behavioural phenotypes (in rodents with potential scalability to non-human primates and
humans). First, we present a proof-of-principle study on mutant zebrafish, in which we correct whole-brain
level abnormal activity patterns and behaviours by using large-scale single-neuron resolution measurements,
and by simultaneously modulating multiple sub-networks via neuromodulator cocktails. Next, we present
strong preliminary data in rodents and our plan: (1) For manipulating brain circuits in rodents/primates noninvasively,
we will develop technologies that can deliver receptive-specific neuromodulators to spatially
precise brain targets without opening/damaging the blood brain barrier. These methods will employ engineered
ultrasound pulses and drug carrying microparticles we designed. (2) For reading out the brain circuits in
rodents/primates, we will develop flexible low-power neuromorphic μECoG circuits that can detect single
neuron signals from superficial cortical layers of many cortical areas simultaneously. (3) Finally, these novel
technologies will be comprehensively evaluated on a mouse model of obsessive compulsivity and anxiety
using a battery of behavioural tasks to reverse the pathological symptoms (beyond what is achievable by
existing approaches). This project constitutes a major step towards the development and testing of minimallyinvasive
and high-precision technologies for manipulating brain activity patterns, which can impact both our
understanding of the brain and treatment of intractable brain disorders.
Max ERC Funding
1 998 984 €
Duration
Start date: 2019-10-01, End date: 2024-09-30
Project acronym Epi4MS
Project Targeting the epigenome: towards a better understanding of disease pathogenesis and novel therapeutic strategies in Multiple Sclerosis
Researcher (PI) Maja JAGODIC
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary Multiple Sclerosis (MS) is a leading cause of unpredictable and incurable progressive disability in young adults. Although the exact cause remains unknown, this immune-mediated disease is likely triggered by environmental factors in genetically predisposed individuals. I propose that epigenetic mechanisms, which regulate gene expression without affecting the genetic code, mediate the processes that cause MS and that aberrant epigenetic states can be corrected, spearheading the development of alternative therapies. We will exploit the stable and reversible nature of epigenetic marks, in particular DNA methylation, to gain insights into the novel modifiable disease mechanisms by studying the target organ in a way that has not been possible before. This highly ambitious project comprises three synergistic facets formulated in specific aims to: (i) identify epigenetic states that characterize the pathogenesis of MS, (ii) prioritize functional epigenetic states using high-throughput epigenome-screens, and (iii) develop novel approaches for precision medicine based on correcting causal epigenetic states. Our unique MS biobank combined with cutting-edge methodologies to capture pathogenic cells and measure their functional states provides a rational starting point to identify MS targets. I will complement this approach with studies of the functional impact of MS targets using innovative in vitro screens, with the added value of unbiased discovery of robust regulators of specific MS pathways. Finally, my laboratory has extensive experience with animal models of MS and I will utilize these powerful systems to dissect molecular mechanisms of MS targets and test the therapeutic potential of targeted epigenome editing in vivo. Our findings will set the stage for a paradigm-shift in studying and treating chronic inflammatory diseases based on preventing and modulating aggressive immune responses by inducing self-sustained reversal of aberrant epigenetic states.
Summary
Multiple Sclerosis (MS) is a leading cause of unpredictable and incurable progressive disability in young adults. Although the exact cause remains unknown, this immune-mediated disease is likely triggered by environmental factors in genetically predisposed individuals. I propose that epigenetic mechanisms, which regulate gene expression without affecting the genetic code, mediate the processes that cause MS and that aberrant epigenetic states can be corrected, spearheading the development of alternative therapies. We will exploit the stable and reversible nature of epigenetic marks, in particular DNA methylation, to gain insights into the novel modifiable disease mechanisms by studying the target organ in a way that has not been possible before. This highly ambitious project comprises three synergistic facets formulated in specific aims to: (i) identify epigenetic states that characterize the pathogenesis of MS, (ii) prioritize functional epigenetic states using high-throughput epigenome-screens, and (iii) develop novel approaches for precision medicine based on correcting causal epigenetic states. Our unique MS biobank combined with cutting-edge methodologies to capture pathogenic cells and measure their functional states provides a rational starting point to identify MS targets. I will complement this approach with studies of the functional impact of MS targets using innovative in vitro screens, with the added value of unbiased discovery of robust regulators of specific MS pathways. Finally, my laboratory has extensive experience with animal models of MS and I will utilize these powerful systems to dissect molecular mechanisms of MS targets and test the therapeutic potential of targeted epigenome editing in vivo. Our findings will set the stage for a paradigm-shift in studying and treating chronic inflammatory diseases based on preventing and modulating aggressive immune responses by inducing self-sustained reversal of aberrant epigenetic states.
Max ERC Funding
1 998 798 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym F-ELEMENT_ARCHITECT
Project Building Precise Molecular Architectures to Unlock Remarkable f-Element Properties
Researcher (PI) David MILLS
Host Institution (HI) THE UNIVERSITY OF MANCHESTER
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary The astonishing properties of the f-elements have been exploited in numerous consumer technologies, despite their fundamental chemistry being poorly developed. It is now crucial to address this issue to provide the necessary insights to develop future applications. Design criteria exist to build f-element complexes with maximised physical attributes. This adventurous proposal targets the synthesis and thorough analysis of two complementary molecular f-element architectures that 1) optimise magnetic properties and 2) stabilise unusual oxidation states.
In Part 1, we target highly axial f-element complexes that lack equatorial ligand interactions. These molecules can exhibit maximised single-molecule magnet properties, including magnetic hysteresis, a memory effect and as a prerequisite of data storage, at liquid nitrogen temperatures. This is the necessary first step towards achieving high-density molecular data storage without expensive liquid helium cooling and future commercial applications.
In Part 2, we target trigonal f-element complexes that lack axial ligand interactions. These are optimal ligand fields for the stabilisation of low oxidation states, thus we aim for rare lanthanide/actinide(II) and unprecedented lanthanide/actinide(I) complexes. These compounds are ideal candidates for unique measurements of covalency by pulsed electron paramagnetic resonance spectroscopy, which will provide textbook data that can be transferable to nuclear fuel cycles.
An ERC CoG will provide the necessary resources to build a world-leading research team that will deliver landmark synthetic results and fresh insights into f-element electronic structure, whilst opening up new chemical space for future exploitation. These findings will underpin current technologies and will facilitate the discovery of future applications, supporting key Horizon 2020 priority areas including the Flagship on Quantum Technologies, and enhancing the scientific reputation and economy of the EU.
Summary
The astonishing properties of the f-elements have been exploited in numerous consumer technologies, despite their fundamental chemistry being poorly developed. It is now crucial to address this issue to provide the necessary insights to develop future applications. Design criteria exist to build f-element complexes with maximised physical attributes. This adventurous proposal targets the synthesis and thorough analysis of two complementary molecular f-element architectures that 1) optimise magnetic properties and 2) stabilise unusual oxidation states.
In Part 1, we target highly axial f-element complexes that lack equatorial ligand interactions. These molecules can exhibit maximised single-molecule magnet properties, including magnetic hysteresis, a memory effect and as a prerequisite of data storage, at liquid nitrogen temperatures. This is the necessary first step towards achieving high-density molecular data storage without expensive liquid helium cooling and future commercial applications.
In Part 2, we target trigonal f-element complexes that lack axial ligand interactions. These are optimal ligand fields for the stabilisation of low oxidation states, thus we aim for rare lanthanide/actinide(II) and unprecedented lanthanide/actinide(I) complexes. These compounds are ideal candidates for unique measurements of covalency by pulsed electron paramagnetic resonance spectroscopy, which will provide textbook data that can be transferable to nuclear fuel cycles.
An ERC CoG will provide the necessary resources to build a world-leading research team that will deliver landmark synthetic results and fresh insights into f-element electronic structure, whilst opening up new chemical space for future exploitation. These findings will underpin current technologies and will facilitate the discovery of future applications, supporting key Horizon 2020 priority areas including the Flagship on Quantum Technologies, and enhancing the scientific reputation and economy of the EU.
Max ERC Funding
1 990 801 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym HighPotOx
Project Exploring the Limits of High Potential OxidizersPrediction, Validation and Preparation of Unusual Molecules at the Edge of Stability
Researcher (PI) Sebastian HASENSTAB-RIEDEL
Host Institution (HI) FREIE UNIVERSITAET BERLIN
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary The very well-known concept of formal oxidation states, used e. g. for redox reactions is one of the most fundamental ones in general chemistry. However, in the area of very strong oxidizers even the familiar oxido(-II) ligand becomes redox-innocent and assigning oxidation states becomes ambiguous. Very strong (super-) oxidizers are compounds whose oxidizing strength exceeds that of elemental F2. Anyhow, not only molecular oxidizer but also their interaction with the environment in different media needs to be considered, as these dramatically affect their intrinsic oxidizing strength. Here we propose novel conjugate oxidizer/Lewis or Brønsted acid systems with extremely high ox. power. These new ox. media make use of the alliance of high ox. strength and Lewis /Brønsted super acidity. The investigation and development of oxidizers is of essential interest in all areas of chemistry and beyond. Unfortunately a detailed understanding of this fundamental chemistry is still lacking. Here we describe based on three work strands PV, MI, and BP, how we aim at a more fundamental understanding of such systems. The undertaken research, which includes qc investigations, molecular characterizations in matrices and synthetic fluorine chemistry as well as oxido complexes is summarized in five work packages describing different prototype areas (organigram). Based on the gained knowledge, the project will rank and specify such oxidizers and the mechanism leading to ox. media. By using the threefold work strand approach, our project will guide us in a systematic discovery of the systems with high application potential in terms of selectivity and disposability, and oxidizing systems with high to ultrahigh oxidation potentials, and into the chemical terra incognita of fragile molecules at the edge of stability. We envision to highlight that the outcome of the project will be extremely useful for scientists from almost all fields of chemistry and related disciplines.
Summary
The very well-known concept of formal oxidation states, used e. g. for redox reactions is one of the most fundamental ones in general chemistry. However, in the area of very strong oxidizers even the familiar oxido(-II) ligand becomes redox-innocent and assigning oxidation states becomes ambiguous. Very strong (super-) oxidizers are compounds whose oxidizing strength exceeds that of elemental F2. Anyhow, not only molecular oxidizer but also their interaction with the environment in different media needs to be considered, as these dramatically affect their intrinsic oxidizing strength. Here we propose novel conjugate oxidizer/Lewis or Brønsted acid systems with extremely high ox. power. These new ox. media make use of the alliance of high ox. strength and Lewis /Brønsted super acidity. The investigation and development of oxidizers is of essential interest in all areas of chemistry and beyond. Unfortunately a detailed understanding of this fundamental chemistry is still lacking. Here we describe based on three work strands PV, MI, and BP, how we aim at a more fundamental understanding of such systems. The undertaken research, which includes qc investigations, molecular characterizations in matrices and synthetic fluorine chemistry as well as oxido complexes is summarized in five work packages describing different prototype areas (organigram). Based on the gained knowledge, the project will rank and specify such oxidizers and the mechanism leading to ox. media. By using the threefold work strand approach, our project will guide us in a systematic discovery of the systems with high application potential in terms of selectivity and disposability, and oxidizing systems with high to ultrahigh oxidation potentials, and into the chemical terra incognita of fragile molecules at the edge of stability. We envision to highlight that the outcome of the project will be extremely useful for scientists from almost all fields of chemistry and related disciplines.
Max ERC Funding
1 988 280 €
Duration
Start date: 2020-01-01, End date: 2024-12-31
Project acronym iHEAR
Project Gene therapy of inherited and acquired hearing loss
Researcher (PI) Axel Rainer Schambach
Host Institution (HI) MEDIZINISCHE HOCHSCHULE HANNOVER
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary To address the substantial financial and social burden caused by hearing loss in 360 million people world-wide, I aim to improve hearing via gene therapy to correct inherited and protect from acquired hearing loss. In vitro experiments will establish the best vector configurations for transfer of therapeutic genes and miRNAs into inner ear hair cells (HC) and spiral ganglion neurons (SGN). The efficiency of the best-performing vector designs will then be explored in vivo using fluorescent marker proteins. Cell-type specific and inducible promoters as well as receptor-targeted vectors will be employed as a safety measure and to ensure transgene expression in HC and SGN target cells. Once efficient transduction of appropriate target cells and proper expression of therapeutic proteins are demonstrated, I will perform proof-of-concept studies in hearing loss models, incl. established mouse models, to correct (WP1) or protect (WP2) from impaired hearing. To ensure translatability of these findings, I will generate human induced pluripotent stem cells (iPSC) from patients with hearing loss (WP3), so that I can test optimized constructs in human otic cells. Moreover, I have access to a collection of well-characterized samples from over 600 hearing loss patients, including children with congenital hearing loss in whom many novel monogenetic alterations were identified. These resources provide the unique opportunity to generate a novel toolbox for the treatment of hearing loss. In addition to lentiviral and adeno-associated viral (AAV) vector delivery of corrective or protective genes to treat hearing loss, I will apply state-of-the-art genome editing tools to model and correct mutations causative for hearing loss in cell lines, primary cells from murine models, human patients and patient-derived iPSC. This work will contribute to development of clinically translatable approaches for precision medicine strategies to improve hearing loss treatment.
Summary
To address the substantial financial and social burden caused by hearing loss in 360 million people world-wide, I aim to improve hearing via gene therapy to correct inherited and protect from acquired hearing loss. In vitro experiments will establish the best vector configurations for transfer of therapeutic genes and miRNAs into inner ear hair cells (HC) and spiral ganglion neurons (SGN). The efficiency of the best-performing vector designs will then be explored in vivo using fluorescent marker proteins. Cell-type specific and inducible promoters as well as receptor-targeted vectors will be employed as a safety measure and to ensure transgene expression in HC and SGN target cells. Once efficient transduction of appropriate target cells and proper expression of therapeutic proteins are demonstrated, I will perform proof-of-concept studies in hearing loss models, incl. established mouse models, to correct (WP1) or protect (WP2) from impaired hearing. To ensure translatability of these findings, I will generate human induced pluripotent stem cells (iPSC) from patients with hearing loss (WP3), so that I can test optimized constructs in human otic cells. Moreover, I have access to a collection of well-characterized samples from over 600 hearing loss patients, including children with congenital hearing loss in whom many novel monogenetic alterations were identified. These resources provide the unique opportunity to generate a novel toolbox for the treatment of hearing loss. In addition to lentiviral and adeno-associated viral (AAV) vector delivery of corrective or protective genes to treat hearing loss, I will apply state-of-the-art genome editing tools to model and correct mutations causative for hearing loss in cell lines, primary cells from murine models, human patients and patient-derived iPSC. This work will contribute to development of clinically translatable approaches for precision medicine strategies to improve hearing loss treatment.
Max ERC Funding
1 999 500 €
Duration
Start date: 2019-05-01, End date: 2024-04-30
Project acronym ImmunoBioSynth
Project Synergistic engineering of anti-tumor immunity by synthetic biomaterials
Researcher (PI) Bruno DE GEEST
Host Institution (HI) UNIVERSITEIT GENT
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary Immunotherapy holds the potential to dramatically improve the curative prognosis of cancer patients. However, despite significant progress, a huge gap remains to be bridged to gain board success in the clinic. A first limiting factor in cancer immunotherapy is the low response rate in large fraction of the patients and an unmet need exists for more efficient - potentially synergistic - immunotherapies that improve upon or complement existing strategies. The second limiting factor is immune-related toxicity that can cause live-threatening situations as well as seriously impair the quality of life of patients. Therefore, there is an urgent need for safer immunotherapies that allow for a more target-specific engineering of the immune system. Strategies to engineer the immune system via a materials chemistry approach, i.e. immuno-engineering, have gathered major attention over the past decade and could complement or replace biologicals, and holds promise to contribute to resolving the current issues faced by the immunotherapy field. I hypothesize that synthetic biomaterials can play an important role in anti-cancer immunotherapy with regard to synergistic, safe, but potent, instruction of innate and adaptive anti-cancer immunity and to revert the tumor microenvironment from an immune-suppressive into an immune-susceptible state. Hereto, the overall scientific objective of this proposal is to fully embrace the potential of immuno-engineering and develop several highly synergistic biomaterials strategies to engineer the immune system to fight cancer. I will develop a series of biomaterials and address a number of fundamental questions with regard to optimal biomaterial design for immuno-engineering. Based on these findings, I will elucidate those therapeutic strategies that lead to synergistic engineering of innate and adaptive immunity in combination with remodeling the tumor microenvironment from an immune-suppressive into an immune-susceptible state.
Summary
Immunotherapy holds the potential to dramatically improve the curative prognosis of cancer patients. However, despite significant progress, a huge gap remains to be bridged to gain board success in the clinic. A first limiting factor in cancer immunotherapy is the low response rate in large fraction of the patients and an unmet need exists for more efficient - potentially synergistic - immunotherapies that improve upon or complement existing strategies. The second limiting factor is immune-related toxicity that can cause live-threatening situations as well as seriously impair the quality of life of patients. Therefore, there is an urgent need for safer immunotherapies that allow for a more target-specific engineering of the immune system. Strategies to engineer the immune system via a materials chemistry approach, i.e. immuno-engineering, have gathered major attention over the past decade and could complement or replace biologicals, and holds promise to contribute to resolving the current issues faced by the immunotherapy field. I hypothesize that synthetic biomaterials can play an important role in anti-cancer immunotherapy with regard to synergistic, safe, but potent, instruction of innate and adaptive anti-cancer immunity and to revert the tumor microenvironment from an immune-suppressive into an immune-susceptible state. Hereto, the overall scientific objective of this proposal is to fully embrace the potential of immuno-engineering and develop several highly synergistic biomaterials strategies to engineer the immune system to fight cancer. I will develop a series of biomaterials and address a number of fundamental questions with regard to optimal biomaterial design for immuno-engineering. Based on these findings, I will elucidate those therapeutic strategies that lead to synergistic engineering of innate and adaptive immunity in combination with remodeling the tumor microenvironment from an immune-suppressive into an immune-susceptible state.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym ImmunoStem
Project Dissecting and Overcoming Innate Immune Barriers for Therapeutically Efficient Hematopoietic Stem Cell Gene Engineering
Researcher (PI) Anna Christina Kajaste-Rudnitski
Host Institution (HI) OSPEDALE SAN RAFFAELE SRL
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary The low gene manipulation efficiency of human hematopoietic stem cells (HSC) remains a major hurdle for sustainable and broad clinical application of innovative therapies for a wide range of disorders. Indeed, high vector doses and prolonged ex vivo culture are still required for clinically relevant levels of gene transfer even with the most established lentiviral vector-based delivery platforms.
Current and emerging gene transfer and editing technologies expose HSC to components potentially recognized by host antiviral factors and nucleic acid sensors that likely restrict their genetic engineering and contribute to broad individual variability in clinical outcomes observed in recent gene therapy trials. Nevertheless, specific effectors are yet to be identified in HSC. We have recently identified an antiviral factor that potently blocks gene transfer in HSC and have discovered small molecules that efficiently counteract it. This is the first example of how manipulating a single host factor can significantly impact gene transfer efficiencies in HSC but likely represents the mere tip of the iceberg of the plethora of innate sensing mechanisms potentially hampering genetic manipulation of this primitive cell compartment.
This proposal aims to identify the antiviral factors and innate sensing pathways that prevent efficient modification of HSC and to mitigate their effects using methods developed through a thorough understanding of their mechanisms of action. My approach builds on the innovative concept that understanding the crosstalk between HSC and viral vectors will instruct us on which immune sensors and effectors to avoid and how, with direct implications for all gene engineering technologies. Successful completion of this project will deliver broadly exportable novel paradigms of innate pathogen recognition that will allow ground-breaking progress in the development of cutting-edge cell and gene therapies and to fight infectious and autoimmune diseases.
Summary
The low gene manipulation efficiency of human hematopoietic stem cells (HSC) remains a major hurdle for sustainable and broad clinical application of innovative therapies for a wide range of disorders. Indeed, high vector doses and prolonged ex vivo culture are still required for clinically relevant levels of gene transfer even with the most established lentiviral vector-based delivery platforms.
Current and emerging gene transfer and editing technologies expose HSC to components potentially recognized by host antiviral factors and nucleic acid sensors that likely restrict their genetic engineering and contribute to broad individual variability in clinical outcomes observed in recent gene therapy trials. Nevertheless, specific effectors are yet to be identified in HSC. We have recently identified an antiviral factor that potently blocks gene transfer in HSC and have discovered small molecules that efficiently counteract it. This is the first example of how manipulating a single host factor can significantly impact gene transfer efficiencies in HSC but likely represents the mere tip of the iceberg of the plethora of innate sensing mechanisms potentially hampering genetic manipulation of this primitive cell compartment.
This proposal aims to identify the antiviral factors and innate sensing pathways that prevent efficient modification of HSC and to mitigate their effects using methods developed through a thorough understanding of their mechanisms of action. My approach builds on the innovative concept that understanding the crosstalk between HSC and viral vectors will instruct us on which immune sensors and effectors to avoid and how, with direct implications for all gene engineering technologies. Successful completion of this project will deliver broadly exportable novel paradigms of innate pathogen recognition that will allow ground-breaking progress in the development of cutting-edge cell and gene therapies and to fight infectious and autoimmune diseases.
Max ERC Funding
1 994 375 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym InOutBioLight
Project Advanced biohybrid lighting and photovoltaic devices
Researcher (PI) Rubén Darío COSTA
Host Institution (HI) FUNDACION IMDEA MATERIALES
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary InOutBioLight aims to design multifunctional rubbers with enhanced mechanical, thermal, color-converting, and light-guiding features towards advanced biohybrid lighting and photovoltaic technologies. The latter are placed at the forefront of the EU efforts for low-cost production and efficient consumption of electricity, a critical issue for a sustainable development.
In this context, the use of biomolecules as functional components in lighting and photovoltaic devices is still a challenge, as they quickly denature under storage and device operation conditions. This paradigm has changed using an innovative rubber-like material, in which the biofunctionality is long preserved. As a proof-of-concept, color down-converting rubbers based on fluorescent proteins were used to design the first biohybrid white light-emitting diode (bio-HWLED). To develop a new generation of biohybrid devices, InOutBioLight will address the following critical issues, namely i) the nature of the protein-matrix stabilization, ii) how to enhance the thermal/mechanical features, iii) how to design multifunctional rubbers, iv) how to mimic natural patterns for light-guiding, and v) how to expand the technological use of the rubber approach.
To achieve these goals, InOutBioLight involves comprehensive spectroscopic, microscopic, and mechanical studies to investigate the protein-matrix interaction using new polymer matrices, additives, and protein-based nanoparticles. In addition, the mechanical, thermal, and light-coupling features will be enhanced using structural biocompounds and reproducing biomorphic patterns. As such, InOutBioLight offers three major advances: i) a thorough scientific basis for the rubber approach, ii) a significant thrust of the emerging bio-HWLEDs, and iii) innovative breakthroughs beyond state-of-the-art biohybrid solar cells.
Summary
InOutBioLight aims to design multifunctional rubbers with enhanced mechanical, thermal, color-converting, and light-guiding features towards advanced biohybrid lighting and photovoltaic technologies. The latter are placed at the forefront of the EU efforts for low-cost production and efficient consumption of electricity, a critical issue for a sustainable development.
In this context, the use of biomolecules as functional components in lighting and photovoltaic devices is still a challenge, as they quickly denature under storage and device operation conditions. This paradigm has changed using an innovative rubber-like material, in which the biofunctionality is long preserved. As a proof-of-concept, color down-converting rubbers based on fluorescent proteins were used to design the first biohybrid white light-emitting diode (bio-HWLED). To develop a new generation of biohybrid devices, InOutBioLight will address the following critical issues, namely i) the nature of the protein-matrix stabilization, ii) how to enhance the thermal/mechanical features, iii) how to design multifunctional rubbers, iv) how to mimic natural patterns for light-guiding, and v) how to expand the technological use of the rubber approach.
To achieve these goals, InOutBioLight involves comprehensive spectroscopic, microscopic, and mechanical studies to investigate the protein-matrix interaction using new polymer matrices, additives, and protein-based nanoparticles. In addition, the mechanical, thermal, and light-coupling features will be enhanced using structural biocompounds and reproducing biomorphic patterns. As such, InOutBioLight offers three major advances: i) a thorough scientific basis for the rubber approach, ii) a significant thrust of the emerging bio-HWLEDs, and iii) innovative breakthroughs beyond state-of-the-art biohybrid solar cells.
Max ERC Funding
1 999 188 €
Duration
Start date: 2020-01-01, End date: 2024-12-31
Project acronym LeaRNN
Project Principles of Learning in a Recurrent Neural Network
Researcher (PI) Marta Zlatic
Host Institution (HI) THE CHANCELLOR MASTERS AND SCHOLARS OF THE UNIVERSITY OF CAMBRIDGE
Call Details Consolidator Grant (CoG), LS5, ERC-2018-COG
Summary Forming memories, generating predictions based on memories, and updating memories when predictions no longer match actual experience are fundamental brain functions. Dopaminergic neurons provide a so-called “teaching signal” that drives the formation and updates of associative memories across the animal kingdom. Many theoretical models propose how neural circuits could compute the teaching signals, but the actual implementation of this computation in real nervous systems is unknown.
This project will discover the basic principles by which neural circuits compute the teaching signals that drive memory formation and updates using a tractable insect model system, the Drosophila larva. We will generate, for the first time in any animal, the following essential datasets for a distributed, multilayered, recurrent learning circuit, the mushroom body-related circuitry in the larval brain. First, building on our preliminary work that provides the synaptic-resolution connectome of the circuit, including all feedforward and feedback pathways upstream of all dopaminergic neurons, we will generate a map of functional monosynaptic connections. Second, we will obtain cellular-resolution whole-nervous system activity maps in intact living animals, as they form, extinguish, or consolidate memories to discover the features represented in each layer of the circuit (e.g. predictions, actual reinforcement, and prediction errors), the learning algorithms, and the candidate circuit motifs that implement them. Finally, we will develop a model of the circuit constrained by these datasets and test the predictions about the necessity and sufficiency of uniquely identified circuit elements for implementing learning algorithms by selectively manipulating their activity.
Understanding the basic functional principles of an entire multilayered recurrent learning circuit in an animal has the potential to revolutionize, not only neuroscience and medicine, but also machine-learning and robotics.
Summary
Forming memories, generating predictions based on memories, and updating memories when predictions no longer match actual experience are fundamental brain functions. Dopaminergic neurons provide a so-called “teaching signal” that drives the formation and updates of associative memories across the animal kingdom. Many theoretical models propose how neural circuits could compute the teaching signals, but the actual implementation of this computation in real nervous systems is unknown.
This project will discover the basic principles by which neural circuits compute the teaching signals that drive memory formation and updates using a tractable insect model system, the Drosophila larva. We will generate, for the first time in any animal, the following essential datasets for a distributed, multilayered, recurrent learning circuit, the mushroom body-related circuitry in the larval brain. First, building on our preliminary work that provides the synaptic-resolution connectome of the circuit, including all feedforward and feedback pathways upstream of all dopaminergic neurons, we will generate a map of functional monosynaptic connections. Second, we will obtain cellular-resolution whole-nervous system activity maps in intact living animals, as they form, extinguish, or consolidate memories to discover the features represented in each layer of the circuit (e.g. predictions, actual reinforcement, and prediction errors), the learning algorithms, and the candidate circuit motifs that implement them. Finally, we will develop a model of the circuit constrained by these datasets and test the predictions about the necessity and sufficiency of uniquely identified circuit elements for implementing learning algorithms by selectively manipulating their activity.
Understanding the basic functional principles of an entire multilayered recurrent learning circuit in an animal has the potential to revolutionize, not only neuroscience and medicine, but also machine-learning and robotics.
Max ERC Funding
2 350 000 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym LifeLikeMat
Project Dissipative self-assembly in synthetic systems: Towards life-like materials
Researcher (PI) Rafal KLAJN
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary "Living organisms are sophisticated self-assembled structures that exist and operate far from thermodynamic equilibrium and, as such, represent the ultimate example of dissipative self-assembly. They remain stable at highly organized (low-entropy) states owing to the continuous consumption of energy stored in ""chemical fuels"", which they convert into low-energy waste. Dissipative self-assembly is ubiquitous in nature, where it gives rise to complex structures and properties such as self-healing, homeostasis, and camouflage. In sharp contrast, nearly all man-made materials are static: they are designed to serve a given purpose rather than to exhibit different properties dependent on external conditions. Developing the means to rationally design dissipative self-assembly constructs will greatly impact a range of industries, including the pharmaceutical and energy sectors.
The goal of the proposed research program is to develop novel principles for designing dissipative self-assembly systems and to fabricate a range of dissipative materials based on these principles. To achieve this goal, we will employ novel, unconventional approaches based predominantly on integrating organic and colloidal-inorganic building blocks.
Specifically, we will (WP1) drive dissipative self-assembly using chemical reactions such as polymerization, oxidation of sugars, and CO2-to-methanol conversion, (WP2) develop new modes of intrinsically dissipative self-assembly, whereby the activated building blocks are inherently unstable, and (WP3&4) conceive systems whereby self-assembly is spontaneously followed by disassembly.
The proposed studies will lead to new classes of ""driven"" materials with features such as tunable lifetimes, time-dependent electrical conductivity, and dynamic exchange of building blocks. Overall, this project will lay the foundations for developing new synthetic dissipative materials, bringing us closer to the rich and varied functionality of materials found in nature."
Summary
"Living organisms are sophisticated self-assembled structures that exist and operate far from thermodynamic equilibrium and, as such, represent the ultimate example of dissipative self-assembly. They remain stable at highly organized (low-entropy) states owing to the continuous consumption of energy stored in ""chemical fuels"", which they convert into low-energy waste. Dissipative self-assembly is ubiquitous in nature, where it gives rise to complex structures and properties such as self-healing, homeostasis, and camouflage. In sharp contrast, nearly all man-made materials are static: they are designed to serve a given purpose rather than to exhibit different properties dependent on external conditions. Developing the means to rationally design dissipative self-assembly constructs will greatly impact a range of industries, including the pharmaceutical and energy sectors.
The goal of the proposed research program is to develop novel principles for designing dissipative self-assembly systems and to fabricate a range of dissipative materials based on these principles. To achieve this goal, we will employ novel, unconventional approaches based predominantly on integrating organic and colloidal-inorganic building blocks.
Specifically, we will (WP1) drive dissipative self-assembly using chemical reactions such as polymerization, oxidation of sugars, and CO2-to-methanol conversion, (WP2) develop new modes of intrinsically dissipative self-assembly, whereby the activated building blocks are inherently unstable, and (WP3&4) conceive systems whereby self-assembly is spontaneously followed by disassembly.
The proposed studies will lead to new classes of ""driven"" materials with features such as tunable lifetimes, time-dependent electrical conductivity, and dynamic exchange of building blocks. Overall, this project will lay the foundations for developing new synthetic dissipative materials, bringing us closer to the rich and varied functionality of materials found in nature."
Max ERC Funding
1 999 572 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym makingtheretina
Project Principles of retinal neuronal lamination from zebrafish to humans
Researcher (PI) Caren NORDEN
Host Institution (HI) MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN EV
Call Details Consolidator Grant (CoG), LS5, ERC-2018-COG
Summary Neuronal lamination is a hallmark of many diverse brain areas where it is important for efficient circuit formation and neuronal wiring. Despite this significance, the cellular and tissue scale principles that ensure successful and robust lamination are not fully understood. In particular, how cell-tissue interactions and biomechanics influence neuronal lamination is only scarcely explored. To fill this gap, we will use the vertebrate retina with its five neuronal cell types arranged in a highly ordered pattern to investigate the emergence of neuronal lamination.
We will initially use the zebrafish system and employ long term light sheet imaging to reveal the migration behaviour of the different retinal neurons. Based on this, transcriptomics approaches will enable the dissection of cellular pathways and extracellular cues involved in neuronal migration and overall lamination. To dissect how biomechanics influence lamination, we will use Brillouin microscopy to explore the influence of changing tissue stiffness on lamination and test the role of differential adhesion. These combined results will be the basis to expand studies to the human system and ex vivo human organoids to generate insights into human retinal development.
To date, systematic studies investigating molecular pathways in combination with biophysical parameters to understand brain formation across model systems are rare. Due to our previous expertise, we are in an excellent position to perform such interdisciplinary, integrative and interspecies approach. This will unveil common denominators of retinal neuronal lamination in zebrafish, humans and human organoids and thereby reveal the similarities of retinal development in different species and how developmental programs compare in vivo versus ex vivo.
In addition, while this proposal focuses on neural lamination in the retina, findings will also inspire future cross-disciplinary studies investigating neuronal lamination in other parts of the brain.
Summary
Neuronal lamination is a hallmark of many diverse brain areas where it is important for efficient circuit formation and neuronal wiring. Despite this significance, the cellular and tissue scale principles that ensure successful and robust lamination are not fully understood. In particular, how cell-tissue interactions and biomechanics influence neuronal lamination is only scarcely explored. To fill this gap, we will use the vertebrate retina with its five neuronal cell types arranged in a highly ordered pattern to investigate the emergence of neuronal lamination.
We will initially use the zebrafish system and employ long term light sheet imaging to reveal the migration behaviour of the different retinal neurons. Based on this, transcriptomics approaches will enable the dissection of cellular pathways and extracellular cues involved in neuronal migration and overall lamination. To dissect how biomechanics influence lamination, we will use Brillouin microscopy to explore the influence of changing tissue stiffness on lamination and test the role of differential adhesion. These combined results will be the basis to expand studies to the human system and ex vivo human organoids to generate insights into human retinal development.
To date, systematic studies investigating molecular pathways in combination with biophysical parameters to understand brain formation across model systems are rare. Due to our previous expertise, we are in an excellent position to perform such interdisciplinary, integrative and interspecies approach. This will unveil common denominators of retinal neuronal lamination in zebrafish, humans and human organoids and thereby reveal the similarities of retinal development in different species and how developmental programs compare in vivo versus ex vivo.
In addition, while this proposal focuses on neural lamination in the retina, findings will also inspire future cross-disciplinary studies investigating neuronal lamination in other parts of the brain.
Max ERC Funding
1 923 750 €
Duration
Start date: 2019-10-01, End date: 2024-09-30
Project acronym MATRIX
Project Novel mitochondria-targeted therapies for cancer treatment-induced cardiotoxicity
Researcher (PI) Borja Ibáñez Cabeza
Host Institution (HI) CENTRO NACIONAL DE INVESTIGACIONESCARDIOVASCULARES CARLOS III (F.S.P.)
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary Cardiac toxicity is one of the most frequent serious side effects of cancer therapy, affecting up to 30% of treated patients. Cancer treatment-induced cardiotoxicity (CTiCT) can result in severe heart failure. The trade-off between cancer and chronic heart failure is an immense personal burden with physical and psychological consequences. Current therapies for CTiCT are suboptimal, featuring poor early detection algorithms and nonspecific heart failure treatments. Based on our recently published results and additional preliminary data presented here, we propose that CTiCT is associated with altered mitochondrial dynamics, triggering a cardiomyocyte metabolic reprogramming. MATRIX represents a holistic approach to tackling mitochondrial dysfunction in CTiCT. Our hypothesis is that reverting metabolic reprogramming by shifting mitochondrial substrate utilization could represent a new paradigm in the treatment of early-stage CTiCT. By refining a novel imaging-based algorithm recently developed in our group, we will achieve very early detection of myocardial damage in patients treated with commonly prescribed cancer therapies, long before clinically used parameters become abnormal. Such early detection, not available currently, is crucial for implementation of early therapies. We also hypothesize that in end-stage CTiCT, mitochondrial dysfunction has passed a no-return point, and the failing heart will only be rescued by a strategy to replenish the myocardium with fresh healthy mitochondria. This will be achieved with a radical new therapeutic option: in-vivo mitochondrial transplantation. The MATRIX project has broad translational potential, including a new therapeutic approach to a clinically relevant condition, the development of technology for early diagnosis, and advances in knowledge of basic disease mechanisms.
Summary
Cardiac toxicity is one of the most frequent serious side effects of cancer therapy, affecting up to 30% of treated patients. Cancer treatment-induced cardiotoxicity (CTiCT) can result in severe heart failure. The trade-off between cancer and chronic heart failure is an immense personal burden with physical and psychological consequences. Current therapies for CTiCT are suboptimal, featuring poor early detection algorithms and nonspecific heart failure treatments. Based on our recently published results and additional preliminary data presented here, we propose that CTiCT is associated with altered mitochondrial dynamics, triggering a cardiomyocyte metabolic reprogramming. MATRIX represents a holistic approach to tackling mitochondrial dysfunction in CTiCT. Our hypothesis is that reverting metabolic reprogramming by shifting mitochondrial substrate utilization could represent a new paradigm in the treatment of early-stage CTiCT. By refining a novel imaging-based algorithm recently developed in our group, we will achieve very early detection of myocardial damage in patients treated with commonly prescribed cancer therapies, long before clinically used parameters become abnormal. Such early detection, not available currently, is crucial for implementation of early therapies. We also hypothesize that in end-stage CTiCT, mitochondrial dysfunction has passed a no-return point, and the failing heart will only be rescued by a strategy to replenish the myocardium with fresh healthy mitochondria. This will be achieved with a radical new therapeutic option: in-vivo mitochondrial transplantation. The MATRIX project has broad translational potential, including a new therapeutic approach to a clinically relevant condition, the development of technology for early diagnosis, and advances in knowledge of basic disease mechanisms.
Max ERC Funding
1 999 375 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym MechanoTubes
Project Supramolecular machineries with life-like mechanical functions
Researcher (PI) Tibor Kudernac
Host Institution (HI) UNIVERSITEIT TWENTE
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary Artificial molecular motors and switches have the potential to become a core part of nanotechnology. However, a wide gap in length scales still remains unaccounted for, between the operation of these molecules in solution, where their individual mechanical action is randomly dispersed in the Brownian storm, and on the other hand their action at the macroscopic level, e.g. in polymer networks and crystals.
This proposal is about bridging this gap, by developing chemo-mechanical transduction strategies that will allow dynamic molecules to perform a range of unprecedented tasks, e.g. by generating strong directional forces at the nanoscale, and through shape-shifting microscopic formations.
This project aims to harness the mechanically-purposeful motion of dynamic molecules as to generate measurable forces from the nanoscale, and ultimately establish operational principles for chemo-mechanical transduction in supramolecular systems.
In my wholly synthetic approach, I draw inspiration from the operational principles of microtubules. I will incorporate molecular photo-switches into supramolecular tubes, and enable the controlled growth and disassembly of the tubes by using light as the energy input. Thus, I will: (i) Synthesize stiff supramolecular tubes that grow actively under continuous illumination, and disassemble with a power stroke as soon as illumination stops; (ii) Measure, and harvest the forces generated by the tubes to manipulate individual nanoparticles with a sense of directionality; and (iii) Encapsulate the tubes into water droplets and vesicles, to yield shape-shifting, and eventually rudimentary splitting models for cells.
This project reaches beyond the state of the art in adaptive molecular nano-systems, by pioneering strategies to engineer and harness strain in supramolecular assemblies. It thus lays the foundations for machineries that are capable of manipulating matter at length scales that are also those at which the cytoskeleton operates.
Summary
Artificial molecular motors and switches have the potential to become a core part of nanotechnology. However, a wide gap in length scales still remains unaccounted for, between the operation of these molecules in solution, where their individual mechanical action is randomly dispersed in the Brownian storm, and on the other hand their action at the macroscopic level, e.g. in polymer networks and crystals.
This proposal is about bridging this gap, by developing chemo-mechanical transduction strategies that will allow dynamic molecules to perform a range of unprecedented tasks, e.g. by generating strong directional forces at the nanoscale, and through shape-shifting microscopic formations.
This project aims to harness the mechanically-purposeful motion of dynamic molecules as to generate measurable forces from the nanoscale, and ultimately establish operational principles for chemo-mechanical transduction in supramolecular systems.
In my wholly synthetic approach, I draw inspiration from the operational principles of microtubules. I will incorporate molecular photo-switches into supramolecular tubes, and enable the controlled growth and disassembly of the tubes by using light as the energy input. Thus, I will: (i) Synthesize stiff supramolecular tubes that grow actively under continuous illumination, and disassemble with a power stroke as soon as illumination stops; (ii) Measure, and harvest the forces generated by the tubes to manipulate individual nanoparticles with a sense of directionality; and (iii) Encapsulate the tubes into water droplets and vesicles, to yield shape-shifting, and eventually rudimentary splitting models for cells.
This project reaches beyond the state of the art in adaptive molecular nano-systems, by pioneering strategies to engineer and harness strain in supramolecular assemblies. It thus lays the foundations for machineries that are capable of manipulating matter at length scales that are also those at which the cytoskeleton operates.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym METAPTPs
Project PROTEIN TYROSINE PHOSPHATASES IN METABOLIC DISEASES: OXIDATION, DYSFUNCTION AND THERAPEUTIC POTENTIAL
Researcher (PI) Esteban GURZOV AMARELO
Host Institution (HI) UNIVERSITE LIBRE DE BRUXELLES
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary Diabetes mellitus is characterised by hyperglycaemia caused by an absolute or relative insulin deficiency. The global prevalence of diabetes has reached more than 410 million individuals, underscoring the need for novel therapeutic strategies targeting the pathology as a multi-organ disease. Protein tyrosine phosphatases (PTPs) constitute a superfamily of enzymes that dephosphorylate tyrosine-phosphorylated proteins and oppose the actions of protein tyrosine kinases. My previous studies and preliminary data suggest that PTPs act as molecular switches for key signalling events in the development of diabetes, i.e. insulin/glucose/cytokine signalling. Dysregulation of these pathways results in metabolic consequences that are cell-specific. Oxidative stress abrogates the nucleophilic properties of the PTP active site and induces conformational changes that inhibit PTP activity and prevent substrate-binding. I have recently developed an innovative proteomic approach to quantify PTP oxidation in vivo and demonstrated that this occurs in liver/pancreas under pathological conditions, including obesity and inflammation. In this proposal, I aim to fully characterise the activity and oxidation status of PTPs in dysfunctional metabolic relevant cells in obesity and diabetes. Importantly, the crucial role of PTPs make them promising candidates for the treatment of metabolic disorders. I hypothesise that specific antioxidants, diets and/or adenovirus will restore PTP function and ameliorate the metabolic deleterious defects in pre-clinical studies. Over the next 5 years, I aim to:
• Identify the major oxidised PTPs in metabolic relevant tissues/cells in both obesity and diabetes.
• Determine the contribution of PTP inactivation in cellular responses to metabolic signalling in human samples.
• Assess the impact of tissue-specific PTP deficiency on the development of obesity and diabetes.
• Test novel therapeutic approaches targeting PTPs to prevent/reverse metabolic disorders.
Summary
Diabetes mellitus is characterised by hyperglycaemia caused by an absolute or relative insulin deficiency. The global prevalence of diabetes has reached more than 410 million individuals, underscoring the need for novel therapeutic strategies targeting the pathology as a multi-organ disease. Protein tyrosine phosphatases (PTPs) constitute a superfamily of enzymes that dephosphorylate tyrosine-phosphorylated proteins and oppose the actions of protein tyrosine kinases. My previous studies and preliminary data suggest that PTPs act as molecular switches for key signalling events in the development of diabetes, i.e. insulin/glucose/cytokine signalling. Dysregulation of these pathways results in metabolic consequences that are cell-specific. Oxidative stress abrogates the nucleophilic properties of the PTP active site and induces conformational changes that inhibit PTP activity and prevent substrate-binding. I have recently developed an innovative proteomic approach to quantify PTP oxidation in vivo and demonstrated that this occurs in liver/pancreas under pathological conditions, including obesity and inflammation. In this proposal, I aim to fully characterise the activity and oxidation status of PTPs in dysfunctional metabolic relevant cells in obesity and diabetes. Importantly, the crucial role of PTPs make them promising candidates for the treatment of metabolic disorders. I hypothesise that specific antioxidants, diets and/or adenovirus will restore PTP function and ameliorate the metabolic deleterious defects in pre-clinical studies. Over the next 5 years, I aim to:
• Identify the major oxidised PTPs in metabolic relevant tissues/cells in both obesity and diabetes.
• Determine the contribution of PTP inactivation in cellular responses to metabolic signalling in human samples.
• Assess the impact of tissue-specific PTP deficiency on the development of obesity and diabetes.
• Test novel therapeutic approaches targeting PTPs to prevent/reverse metabolic disorders.
Max ERC Funding
1 966 906 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym MOF-reactors
Project Metal-Organic Frameworks as Chemical Reactors for the Synthesis of Well-Defined Sub-Nanometer Metal Clusters
Researcher (PI) Emilio PARDO
Host Institution (HI) UNIVERSITAT DE VALENCIA
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary Humankind advancement is connected to the use and development of metal forms. Recent works have unveiled exceptional properties –such as luminescence, biocompatibility, antitumoral activity or a superlative catalytic activity– for small aggregations of metal atoms, so–called sub–nanometer metal clusters (SNMCs). Despite this importance, the gram-scale synthesis of structurally and electronically well–defined SNMCs is still far from being a reality.
The present proposal situates at the centre of such weakness and aims at making a breakthrough step-change on the use of metal-organic frameworks (MOFs) as chemical reactors for the in–situ synthesis of stable ligand-free SNMCs with such unique properties. This challenging synthetic strategy, which is assisted by striking published and inedited preliminary results, has solid foundations. Firstly, the design and large-scale preparation of cheap and novel families of highly robust and crystalline MOFs with tailor-made functional channels to be used as chemical reactors. Secondly, the application of solid-state post-synthetic methods to drive the multigram-scale preparation of unique ligand-free homo- and heterometallic SNMCs, which are, in the best-case scenario, very difficult to be obtained and stabilised outside the channels. Last but not least, single-crystal X-Ray diffraction will be used as the definitive tool for the characterisation, at the atomic level, of such ultrasmall species offering unprecedented snapshots about their real structures and formation mechanisms.
The ultimate goal will be upscaling this synthetic strategy aiming at the large-scale fabrication of SNMCs and their industrial application will be then evaluated. A successful achievement of all the aforementioned objectives of this ground-breaking project would open new routes for the use of MOFs as chemical reactors to manufacture, at competitive prices, MOF-driven, structurally and electronically well–defined, ligand–free SNMCs in a multigram-scale.
Summary
Humankind advancement is connected to the use and development of metal forms. Recent works have unveiled exceptional properties –such as luminescence, biocompatibility, antitumoral activity or a superlative catalytic activity– for small aggregations of metal atoms, so–called sub–nanometer metal clusters (SNMCs). Despite this importance, the gram-scale synthesis of structurally and electronically well–defined SNMCs is still far from being a reality.
The present proposal situates at the centre of such weakness and aims at making a breakthrough step-change on the use of metal-organic frameworks (MOFs) as chemical reactors for the in–situ synthesis of stable ligand-free SNMCs with such unique properties. This challenging synthetic strategy, which is assisted by striking published and inedited preliminary results, has solid foundations. Firstly, the design and large-scale preparation of cheap and novel families of highly robust and crystalline MOFs with tailor-made functional channels to be used as chemical reactors. Secondly, the application of solid-state post-synthetic methods to drive the multigram-scale preparation of unique ligand-free homo- and heterometallic SNMCs, which are, in the best-case scenario, very difficult to be obtained and stabilised outside the channels. Last but not least, single-crystal X-Ray diffraction will be used as the definitive tool for the characterisation, at the atomic level, of such ultrasmall species offering unprecedented snapshots about their real structures and formation mechanisms.
The ultimate goal will be upscaling this synthetic strategy aiming at the large-scale fabrication of SNMCs and their industrial application will be then evaluated. A successful achievement of all the aforementioned objectives of this ground-breaking project would open new routes for the use of MOFs as chemical reactors to manufacture, at competitive prices, MOF-driven, structurally and electronically well–defined, ligand–free SNMCs in a multigram-scale.
Max ERC Funding
1 886 000 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym NeuroMag
Project The Neurological Basis of the Magnetic Sense
Researcher (PI) David KEAYS
Host Institution (HI) FORSCHUNGSINSTITUT FUR MOLEKULARE PATHOLOGIE GESELLSCHAFT MBH
Call Details Consolidator Grant (CoG), LS5, ERC-2018-COG
Summary Each year millions of animals undertake remarkable migratory journeys, across oceans and through hemispheres, guided by the Earth’s magnetic field. While there is unequivocal behavioural evidence demonstrating the existence of the magnetic sense, it is the least understood of all sensory faculties. The biophysical, molecular, cellular, and neurological underpinnings of the sense remain opaque. In this application we aim to remedy this situation, exploiting an established assay, our unique infrastructure, and state-of-the-art methodology, using pigeons as a model system. The proposal will address three questions:
1) Where are the primary magnetosensors?
2) Where is magnetic information processed in the brain?
3) How is magnetic information encoded in the brain?
In Aim 1 we will explore whether inner ear hair cells are the primary sensors, and if the detection of magnetic stimuli depends on the presence of magnetic crystals or electromagnetic induction. We will employ a range of physical methods to locate magnetite, and a molecular approach to identify putative electroreceptors. In Aim 2 we will use light sheet microscopy coupled with clearing methods to undertake whole brain mapping of magnetically-induced neuronal activation in the pigeon. We will complement these studies with transcriptomic methods to molecularly and anatomically define magnetosensitive circuits within the pigeon brain. We will build on this work in Aim 3 utilising in vivo 2-photon microscopy to investigate how cells within the pigeon brain encode magnetic information. We will determine whether neurons encode for specific components of the magnetic field (i.e. inclination, intensity, and polarity) and explore whether there are spatially restricted ensembles, providing a dynamic picture of magnetically induced neuronal activity. We anticipate that these experiments will reveal a secret that nature has kept hidden for millennia; How do animals detect magnetic fields?
Summary
Each year millions of animals undertake remarkable migratory journeys, across oceans and through hemispheres, guided by the Earth’s magnetic field. While there is unequivocal behavioural evidence demonstrating the existence of the magnetic sense, it is the least understood of all sensory faculties. The biophysical, molecular, cellular, and neurological underpinnings of the sense remain opaque. In this application we aim to remedy this situation, exploiting an established assay, our unique infrastructure, and state-of-the-art methodology, using pigeons as a model system. The proposal will address three questions:
1) Where are the primary magnetosensors?
2) Where is magnetic information processed in the brain?
3) How is magnetic information encoded in the brain?
In Aim 1 we will explore whether inner ear hair cells are the primary sensors, and if the detection of magnetic stimuli depends on the presence of magnetic crystals or electromagnetic induction. We will employ a range of physical methods to locate magnetite, and a molecular approach to identify putative electroreceptors. In Aim 2 we will use light sheet microscopy coupled with clearing methods to undertake whole brain mapping of magnetically-induced neuronal activation in the pigeon. We will complement these studies with transcriptomic methods to molecularly and anatomically define magnetosensitive circuits within the pigeon brain. We will build on this work in Aim 3 utilising in vivo 2-photon microscopy to investigate how cells within the pigeon brain encode magnetic information. We will determine whether neurons encode for specific components of the magnetic field (i.e. inclination, intensity, and polarity) and explore whether there are spatially restricted ensembles, providing a dynamic picture of magnetically induced neuronal activity. We anticipate that these experiments will reveal a secret that nature has kept hidden for millennia; How do animals detect magnetic fields?
Max ERC Funding
1 990 376 €
Duration
Start date: 2019-10-01, End date: 2024-09-30
Project acronym PrefrontalMap
Project Organization and learning-associated dynamics of prefrontal synaptic connectivity
Researcher (PI) Ofer YIZHAR
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS5, ERC-2018-COG
Summary How does experience alter the functional architecture of synaptic connections in neural circuits? This question is particularly pertinent for the complex circuits of the medial prefrontal cortex (mPFC), a high-order associative neocortical area that plays a crucial role in flexible, goal-directed behavior. The mPFC is densely interconnected with cortical and subcortical circuits, and its neurons were shown to undergo substantial experience-dependent structural remodeling that is thought to support learning and memory consolidation. However, little is known regarding the synaptic organization of this complex circuit, and of the functional implications of its experience-dependent structural remodeling. In this proposal, we aim to uncover the organization and learning-associated dynamics of functional connectivity in the mouse mPFC.
To obtain high-resolution maps of cell type-specific synaptic connectivity in the mPFC, we will combine single-cell optogenetic manipulation with calcium imaging and electrophysiology in vitro, and establish the circuit-wide organization of connectivity within and between defined projecting neuron populations. We will test the hypothesis that pyramidal neurons projecting to subcortical targets form tightly interconnected subnetworks, and that inhibitory inputs to these networks, through selective innervation, can modulate information output from the mPFC.
To understand how learning changes the functional synaptic organization of the mPFC, we will establish an all-optical system for interrogation of synaptic connectivity in vivo. We will utilize this powerful platform to test the hypothesis that prefrontal-dependent learning is associated with reorganization of local-circuit functional connectivity among identified subcortically-projecting cell assemblies.
Our innovative technology will be widely applicable for neural circuit analysis in a variety of systems, and allow us to gain new insights into the complex circuitry of the mPFC.
Summary
How does experience alter the functional architecture of synaptic connections in neural circuits? This question is particularly pertinent for the complex circuits of the medial prefrontal cortex (mPFC), a high-order associative neocortical area that plays a crucial role in flexible, goal-directed behavior. The mPFC is densely interconnected with cortical and subcortical circuits, and its neurons were shown to undergo substantial experience-dependent structural remodeling that is thought to support learning and memory consolidation. However, little is known regarding the synaptic organization of this complex circuit, and of the functional implications of its experience-dependent structural remodeling. In this proposal, we aim to uncover the organization and learning-associated dynamics of functional connectivity in the mouse mPFC.
To obtain high-resolution maps of cell type-specific synaptic connectivity in the mPFC, we will combine single-cell optogenetic manipulation with calcium imaging and electrophysiology in vitro, and establish the circuit-wide organization of connectivity within and between defined projecting neuron populations. We will test the hypothesis that pyramidal neurons projecting to subcortical targets form tightly interconnected subnetworks, and that inhibitory inputs to these networks, through selective innervation, can modulate information output from the mPFC.
To understand how learning changes the functional synaptic organization of the mPFC, we will establish an all-optical system for interrogation of synaptic connectivity in vivo. We will utilize this powerful platform to test the hypothesis that prefrontal-dependent learning is associated with reorganization of local-circuit functional connectivity among identified subcortically-projecting cell assemblies.
Our innovative technology will be widely applicable for neural circuit analysis in a variety of systems, and allow us to gain new insights into the complex circuitry of the mPFC.
Max ERC Funding
1 880 003 €
Duration
Start date: 2019-02-01, End date: 2024-01-31
Project acronym PROTONMBRT
Project Spatial fractionation of the dose in proton therapy: a novel therapeutic approach
Researcher (PI) Yolanda PREZADO ALONSO
Host Institution (HI) CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE CNRS
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary Radiotherapy (RT) is one of the most frequently used methods for cancer treatment (above 50% of patients will receive RT). Despite remarkable advancements, the dose tolerances of normal tissues continue to be the main limitation in RT. Finding novel approaches that allow increasing normal tissue resistance is of utmost importance. This would make it possible to escalate tumour dose, resulting in an improvement in cure rate. With this aim, I propose a new approach, called proton minibeam radiation therapy (PROTONMBRT), which combines the prominent advantages of protons for RT and the remarkable tissue preservation provided by the use of submillimetric field sizes and a spatial fractionation of the dose, as in minibeam radiation therapy (MBRT). The main objectives of this project are to explore the gain of therapeutic index for radioresistant tumors, to disentangle the biological mechanisms involved and to evaluate the clinical potential of this novel approach. For this purpose, a method for minibeam generation adequate for patient treatments and a complete set of dosimetric tools will be developed. Then, tumour control effectiveness will be evaluated, and the possible biological mechanisms involved both in tumour and normal tissue responses will be disentangled. The gain in normal tissue recovery can foster one of the main applications of proton therapy, paediatric oncology, as well as open the door to an effective treatment of very radioresistant tumours, such as high-grade gliomas, which are currently mostly treated palliatively.
Summary
Radiotherapy (RT) is one of the most frequently used methods for cancer treatment (above 50% of patients will receive RT). Despite remarkable advancements, the dose tolerances of normal tissues continue to be the main limitation in RT. Finding novel approaches that allow increasing normal tissue resistance is of utmost importance. This would make it possible to escalate tumour dose, resulting in an improvement in cure rate. With this aim, I propose a new approach, called proton minibeam radiation therapy (PROTONMBRT), which combines the prominent advantages of protons for RT and the remarkable tissue preservation provided by the use of submillimetric field sizes and a spatial fractionation of the dose, as in minibeam radiation therapy (MBRT). The main objectives of this project are to explore the gain of therapeutic index for radioresistant tumors, to disentangle the biological mechanisms involved and to evaluate the clinical potential of this novel approach. For this purpose, a method for minibeam generation adequate for patient treatments and a complete set of dosimetric tools will be developed. Then, tumour control effectiveness will be evaluated, and the possible biological mechanisms involved both in tumour and normal tissue responses will be disentangled. The gain in normal tissue recovery can foster one of the main applications of proton therapy, paediatric oncology, as well as open the door to an effective treatment of very radioresistant tumours, such as high-grade gliomas, which are currently mostly treated palliatively.
Max ERC Funding
1 997 870 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym REALNANO
Project 3D Structure of Nanomaterials under Realistic Conditions
Researcher (PI) Sara BALS
Host Institution (HI) UNIVERSITEIT ANTWERPEN
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary The properties of nanomaterials are essentially determined by their 3D structure. Electron tomography enables one to measure the morphology and composition of nanostructures in 3D, even at atomic resolution. Unfortunately, all these measurements are performed at room temperature and in ultra-high vacuum, which are conditions that are completely irrelevant for the use of nanoparticles in real applications! Moreover, nanoparticles often have ligands at their surface, which form the interface to the environment. These ligands are mostly neglected in imaging, although they strongly influence the growth, thermal stability and drive self-assembly.
I will develop innovative and quantitative 3D characterisation tools, compatible with the fast changes of nanomaterials that occur in a realistic thermal and gaseous environment. To visualise surface ligands, I will combine direct electron detection with novel exit wave reconstruction techniques.
Tracking the 3D structure of nanomaterials in a relevant environment is extremely challenging and ambitious. However, our preliminary experiments demonstrate the enormous impact. We will be able to perform a dynamic characterisation of shape changes of nanoparticles. This is important to improve thermal stability during drug delivery, sensing, data storage or hyperthermic cancer treatment. We will provide quantitative 3D measurements of the coordination numbers of the surface atoms of catalytic nanoparticles and follow the motion of individual atoms live during catalysis. By visualising surface ligands, we will understand their fundamental influence on particle shape and during self-assembly.
This program will be the start of a completely new research line in the field of 3D imaging at the atomic scale. The outcome will certainly boost the design and performance of nanomaterials. This is not only of importance at a fundamental level, but is a prerequisite for the incorporation of nanomaterials in our future technology.
Summary
The properties of nanomaterials are essentially determined by their 3D structure. Electron tomography enables one to measure the morphology and composition of nanostructures in 3D, even at atomic resolution. Unfortunately, all these measurements are performed at room temperature and in ultra-high vacuum, which are conditions that are completely irrelevant for the use of nanoparticles in real applications! Moreover, nanoparticles often have ligands at their surface, which form the interface to the environment. These ligands are mostly neglected in imaging, although they strongly influence the growth, thermal stability and drive self-assembly.
I will develop innovative and quantitative 3D characterisation tools, compatible with the fast changes of nanomaterials that occur in a realistic thermal and gaseous environment. To visualise surface ligands, I will combine direct electron detection with novel exit wave reconstruction techniques.
Tracking the 3D structure of nanomaterials in a relevant environment is extremely challenging and ambitious. However, our preliminary experiments demonstrate the enormous impact. We will be able to perform a dynamic characterisation of shape changes of nanoparticles. This is important to improve thermal stability during drug delivery, sensing, data storage or hyperthermic cancer treatment. We will provide quantitative 3D measurements of the coordination numbers of the surface atoms of catalytic nanoparticles and follow the motion of individual atoms live during catalysis. By visualising surface ligands, we will understand their fundamental influence on particle shape and during self-assembly.
This program will be the start of a completely new research line in the field of 3D imaging at the atomic scale. The outcome will certainly boost the design and performance of nanomaterials. This is not only of importance at a fundamental level, but is a prerequisite for the incorporation of nanomaterials in our future technology.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-05-01, End date: 2024-04-30
Project acronym RECON
Project Reprogramming Conformation by Fluorination: Exploring New Areas of Chemical Space
Researcher (PI) Ryan GILMOUR
Host Institution (HI) WESTFAELISCHE WILHELMS-UNIVERSITAET MUENSTER
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary Despite the abundance of organic compounds in Nature, only 12 contain fluorine. In contrast, fluorinated organic materials account for over 40% of all pharmaceuticals and agrochemicals. Closer inspection of the fluorination patterns in these functional molecules reveals striking extremes towards perfluorination (in both 2D and 3D scaffolds) or single site fluorination predominantly in aryl substituents. Consequently, most fluorinated moieties in functional materials lack stereochemical information and are thus achiral. This disparity between the paucity of naturally occurring organofluorine compounds and their venerable history in functional molecule design confirms the enormous potential of fluorinated materials in the discovery of novel properties. That progress has largely been confined to 3 dimensional achiral and 2 dimensional achiral architectures reflects the synthetic challenges associated with preparing stereochemical defined multiply fluorinated systems. A major limitation in the construction of C(sp3)-F units remains the need for substrate pre-functionalisation via oxidation and the competing substitution/elimination scenario that compromises efficiency in the deoxyfluorination. This problem is magnified in the synthesis of optically active fluorides where the deoxyfluorination can compromise the enantiopurity of the starting materials. The principle aim of RECON is to facilitate exploration of 3D, chiral space by providing access to multiply fluorinated, stereochemically complex organofluorine materials from simple feedstock using inexpensive, commercially available fluoride sources. In providing a modular platform to rationally place function on a structural basis, exploration of uncharted chemical space will accelerate the discovery of next generation materials for medicinal and agrochemistry, material sciences and bio-medicine.
Summary
Despite the abundance of organic compounds in Nature, only 12 contain fluorine. In contrast, fluorinated organic materials account for over 40% of all pharmaceuticals and agrochemicals. Closer inspection of the fluorination patterns in these functional molecules reveals striking extremes towards perfluorination (in both 2D and 3D scaffolds) or single site fluorination predominantly in aryl substituents. Consequently, most fluorinated moieties in functional materials lack stereochemical information and are thus achiral. This disparity between the paucity of naturally occurring organofluorine compounds and their venerable history in functional molecule design confirms the enormous potential of fluorinated materials in the discovery of novel properties. That progress has largely been confined to 3 dimensional achiral and 2 dimensional achiral architectures reflects the synthetic challenges associated with preparing stereochemical defined multiply fluorinated systems. A major limitation in the construction of C(sp3)-F units remains the need for substrate pre-functionalisation via oxidation and the competing substitution/elimination scenario that compromises efficiency in the deoxyfluorination. This problem is magnified in the synthesis of optically active fluorides where the deoxyfluorination can compromise the enantiopurity of the starting materials. The principle aim of RECON is to facilitate exploration of 3D, chiral space by providing access to multiply fluorinated, stereochemically complex organofluorine materials from simple feedstock using inexpensive, commercially available fluoride sources. In providing a modular platform to rationally place function on a structural basis, exploration of uncharted chemical space will accelerate the discovery of next generation materials for medicinal and agrochemistry, material sciences and bio-medicine.
Max ERC Funding
1 999 375 €
Duration
Start date: 2019-02-01, End date: 2024-01-31
Project acronym RememberEx
Project Human Subcortical-Cortical Circuit Dynamics for Remembering the Exceptional
Researcher (PI) Bryan STRANGE
Host Institution (HI) UNIVERSIDAD POLITECNICA DE MADRID
Call Details Consolidator Grant (CoG), LS5, ERC-2018-COG
Summary Our memory system is optimised for remembering the exceptional over the mundane. We remember better those events that violate predictions generated by the prevailing context, particularly because of surprise or emotional impact. Understanding how we form and retrieve long-term memories for important or salient events is critical for combating the rapidly growing incidence of pathologies associated with memory dysfunction with huge socio-econonomic burden. Human lesion and non-invasive functional imaging data, motivated by findings from animal models, have identified subcortical structures that are critical for upregulating hippocampal function during salient event memory. However, mechanistic understanding of these processes in humans remains scarce, and requires better experimental approaches such as direct intracranial recordings from, and focal electrical stimulation of, these subcortical structures.
This project will characterise human subcortico-cortical neuronal circuit dynamics associated with enhanced episodic memory for salient stimuli by studying direct recordings from human hippocampus, amygdala, nucleus accumbens, ventral midbrain and cortex. Within this framework, I will elucidate the electrophysiological mechanisms underlying amygdala-hippocampal-cortical coupling that lead to better memory for emotional stimuli, extend the hippocampal role in detecting unpredicted stimuli to define its role in orchestrating cortical dynamics in unpredictable contexts, and discover the neuronal response profile of the human mesolimbic dopamine system during salient stimulus encoding. The predicted results, based on my own preliminary data, will offer several conceptual breakthroughs, particularly regarding hippocampal function and the role of dopaminergic ventral midbrain in memory. The knowledge gained from this project is a fundamental requirement for designing therapeutic interventions for patients with memory deficits and other neuropsychiatric disorders.
Summary
Our memory system is optimised for remembering the exceptional over the mundane. We remember better those events that violate predictions generated by the prevailing context, particularly because of surprise or emotional impact. Understanding how we form and retrieve long-term memories for important or salient events is critical for combating the rapidly growing incidence of pathologies associated with memory dysfunction with huge socio-econonomic burden. Human lesion and non-invasive functional imaging data, motivated by findings from animal models, have identified subcortical structures that are critical for upregulating hippocampal function during salient event memory. However, mechanistic understanding of these processes in humans remains scarce, and requires better experimental approaches such as direct intracranial recordings from, and focal electrical stimulation of, these subcortical structures.
This project will characterise human subcortico-cortical neuronal circuit dynamics associated with enhanced episodic memory for salient stimuli by studying direct recordings from human hippocampus, amygdala, nucleus accumbens, ventral midbrain and cortex. Within this framework, I will elucidate the electrophysiological mechanisms underlying amygdala-hippocampal-cortical coupling that lead to better memory for emotional stimuli, extend the hippocampal role in detecting unpredicted stimuli to define its role in orchestrating cortical dynamics in unpredictable contexts, and discover the neuronal response profile of the human mesolimbic dopamine system during salient stimulus encoding. The predicted results, based on my own preliminary data, will offer several conceptual breakthroughs, particularly regarding hippocampal function and the role of dopaminergic ventral midbrain in memory. The knowledge gained from this project is a fundamental requirement for designing therapeutic interventions for patients with memory deficits and other neuropsychiatric disorders.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-05-01, End date: 2024-04-30
Project acronym ReNewHydrides
Project Renewable Hydride Donors and Their Utilization in Catalytic Reduction and Deoxygenation Reactions
Researcher (PI) Thibault CANTAT
Host Institution (HI) COMMISSARIAT A L ENERGIE ATOMIQUE ET AUX ENERGIES ALTERNATIVES
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary The production of chemicals, plastics, solvents, etc., contributes to 20 % of the Gross Value Added in the EU, where sales have doubled over the last 20 years. Despite this dynamism, the chemical industry is energy intensive and 95 % of organic chemicals derive from fossil oil and natural gas. To sustain the growth of this industry, the replacement of fossil feedstocks with renewable carbon, phosphorus and silicon sources should be encouraged. Nonetheless, such a sourcing shift represents a paradigm shift: while the development of petrochemistry has relied on the selective oxidation of hydrocarbons, the conversion of renewable feedstocks (e.g. CO2, phosphates, silicates or biomass) requires efficient reduction methods and catalysts to overcome their oxidized nature.
Today, no reduction method meets the criteria for a versatile and energy efficient reduction of oxidized feedstocks and the aim of the ReNewHydrides project is to design novel reductants and catalytic reactions to achieve this important aim. At the crossroads of main group element chemistry, organometallic chemistry, electrochemistry and homogenous catalysis, I propose to develop innovative and recyclable reductants based on silicon and boron compounds, and to utilize them to tackle catalytic challenges in the reduction of C–O, P–O and Si–O bonds. The overarching principle is to build a balanced synthetic cycle, where the electrochemical reduction of functionalized and oxidized substrates is ensured by silicon and boron based hydride donors, with a high energy efficiency and selectivity.
This project will foster innovative routes in the utilization of renewable carbon, phosphorus and silicon feedstocks. It is therefore of high risk, but ultimately extremely rewarding. The results will also also open-up new horizons in silicon and boron chemistry and they will finally serve the scientific community involved in the fields of organic and inorganic chemistry, sustainable chemistry and energy storage.
Summary
The production of chemicals, plastics, solvents, etc., contributes to 20 % of the Gross Value Added in the EU, where sales have doubled over the last 20 years. Despite this dynamism, the chemical industry is energy intensive and 95 % of organic chemicals derive from fossil oil and natural gas. To sustain the growth of this industry, the replacement of fossil feedstocks with renewable carbon, phosphorus and silicon sources should be encouraged. Nonetheless, such a sourcing shift represents a paradigm shift: while the development of petrochemistry has relied on the selective oxidation of hydrocarbons, the conversion of renewable feedstocks (e.g. CO2, phosphates, silicates or biomass) requires efficient reduction methods and catalysts to overcome their oxidized nature.
Today, no reduction method meets the criteria for a versatile and energy efficient reduction of oxidized feedstocks and the aim of the ReNewHydrides project is to design novel reductants and catalytic reactions to achieve this important aim. At the crossroads of main group element chemistry, organometallic chemistry, electrochemistry and homogenous catalysis, I propose to develop innovative and recyclable reductants based on silicon and boron compounds, and to utilize them to tackle catalytic challenges in the reduction of C–O, P–O and Si–O bonds. The overarching principle is to build a balanced synthetic cycle, where the electrochemical reduction of functionalized and oxidized substrates is ensured by silicon and boron based hydride donors, with a high energy efficiency and selectivity.
This project will foster innovative routes in the utilization of renewable carbon, phosphorus and silicon feedstocks. It is therefore of high risk, but ultimately extremely rewarding. The results will also also open-up new horizons in silicon and boron chemistry and they will finally serve the scientific community involved in the fields of organic and inorganic chemistry, sustainable chemistry and energy storage.
Max ERC Funding
1 999 838 €
Duration
Start date: 2019-10-01, End date: 2024-09-30
Project acronym SCALE-HALO
Project Multiscale chemical engineering of functional metal halides
Researcher (PI) Maksym KOVALENKO
Host Institution (HI) EIDGENOESSISCHE TECHNISCHE HOCHSCHULE ZUERICH
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary SCALE-HALO proposes a research program that will advance the development of highly luminescent molecular and solid-state compounds by focusing on the emerging, vast, and rather underexplored compositional and structural spaces comprised of metals and halogens, i.e., metal halides (MHs). SCALE-HALO is motivated by the eventual utility of MHs as versatile photonic sources in modern appliances (e.g., displays and lighting) and in future quantum technologies. The recent success of lead halide perovskites in optoelectronics inspires broader exploration of the chemistry and photophysics of MHs. The clear objective is to determine factors controlling the spectral widths and emission peak wavelengths, Stokes shifts, radiative lifetimes, and quantum efficiencies. In addition to the need to discover new chemically robust and nontoxic MH emitters, there is also a critical need to engineer material morphologies suitable for specific applications (e.g., thin films, nanocrystals, composites, etc.) Ensuring the thermal and environmental stabilities are especially important efforts. SCALE-HALO will therefore encompass the chemical engineering of MHs at the atomic scale (e.g., new compounds), nanoscale (e.g., synthesis of nanostructures and their surface chemistry), and mesoscale (e.g., nanostructure superlattices and composites). Furthermore, modern exploratory syntheses will be accelerated with automated high-throughput methods (e.g., robotics and microfluidics). The characterization toolbox for probing the local atomistic structure will be expanded with multinuclear NMR spectroscopy. The individual and collective optical properties of MH nanostructures and their periodic assemblies will be established and rationalized. Toward diverse real-world applications, first trials will be undertaken to evaluate the potentials of novel MH materials for LCD displays, solid-state lighting and light-emitting diodes.
Summary
SCALE-HALO proposes a research program that will advance the development of highly luminescent molecular and solid-state compounds by focusing on the emerging, vast, and rather underexplored compositional and structural spaces comprised of metals and halogens, i.e., metal halides (MHs). SCALE-HALO is motivated by the eventual utility of MHs as versatile photonic sources in modern appliances (e.g., displays and lighting) and in future quantum technologies. The recent success of lead halide perovskites in optoelectronics inspires broader exploration of the chemistry and photophysics of MHs. The clear objective is to determine factors controlling the spectral widths and emission peak wavelengths, Stokes shifts, radiative lifetimes, and quantum efficiencies. In addition to the need to discover new chemically robust and nontoxic MH emitters, there is also a critical need to engineer material morphologies suitable for specific applications (e.g., thin films, nanocrystals, composites, etc.) Ensuring the thermal and environmental stabilities are especially important efforts. SCALE-HALO will therefore encompass the chemical engineering of MHs at the atomic scale (e.g., new compounds), nanoscale (e.g., synthesis of nanostructures and their surface chemistry), and mesoscale (e.g., nanostructure superlattices and composites). Furthermore, modern exploratory syntheses will be accelerated with automated high-throughput methods (e.g., robotics and microfluidics). The characterization toolbox for probing the local atomistic structure will be expanded with multinuclear NMR spectroscopy. The individual and collective optical properties of MH nanostructures and their periodic assemblies will be established and rationalized. Toward diverse real-world applications, first trials will be undertaken to evaluate the potentials of novel MH materials for LCD displays, solid-state lighting and light-emitting diodes.
Max ERC Funding
1 999 950 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym SCHIZTYPE
Project Brain cell type-specific interactions and schizophrenia
Researcher (PI) Jens Gunnar Hakan HJERLING LEFFLER
Host Institution (HI) KAROLINSKA INSTITUTET
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary Schizophrenia is a heritable but genetically complex disease. Pathological and epidemiological data fit a model of SCZ as a network disease with perturbations during brain development leading to early-adulthood onset clinical symptomatology. Our present understanding is based on single markers or arrays of gene expression from tissue samples containing multiple cell types. As a consequence, pathological changes in the function of inhibitory or excitatory neurons, microglia, or oligodendrocytes have variously been proposed to be the cause of symptoms. In light of recent data I hypothesize that it is unlikely that the various cellular SCZ-pathologies all arise independently from genetic alterations in multiple cell types. Recent findings from my lab show that in the cortex the expression of risk genes for SCZ are enriched in excitatory neurons, and that this set of risk-genes is largely non-overlapping with those expressed in other cell types. I propose that pathological genetic changes in excitatory cells ultimately initiates pathological changes in other cell types contributing to the multiple cellular pathologies observed in SCZ. We will:
1. Identify cell type-specific gene regulatory networks involved in SCZ (SCZ-GRNs) in prefrontal cortical excitatory cells by analysis of four distinct SCZ mouse models.
2. Confirm putative SCZ-GRNs in patient material using in situ transcriptomics on postmortem brains and connect to clinical features via collaboration with genomic studies in Sweden and Denmark.
3. Functionally investigate the effects of perturbing excitatory cell SCZ-GRNs on other cell types.
Single-cell RNA-seq, providing insights into the molecular properties of individual cells, and modern molecular tools for perturbing transcription in a cell type-specific way opens up for new knowledge of mechanisms underlying SCZ pathology. My work will identify causal relationships that can be exploited for the development of strategies for personalized treatment.
Summary
Schizophrenia is a heritable but genetically complex disease. Pathological and epidemiological data fit a model of SCZ as a network disease with perturbations during brain development leading to early-adulthood onset clinical symptomatology. Our present understanding is based on single markers or arrays of gene expression from tissue samples containing multiple cell types. As a consequence, pathological changes in the function of inhibitory or excitatory neurons, microglia, or oligodendrocytes have variously been proposed to be the cause of symptoms. In light of recent data I hypothesize that it is unlikely that the various cellular SCZ-pathologies all arise independently from genetic alterations in multiple cell types. Recent findings from my lab show that in the cortex the expression of risk genes for SCZ are enriched in excitatory neurons, and that this set of risk-genes is largely non-overlapping with those expressed in other cell types. I propose that pathological genetic changes in excitatory cells ultimately initiates pathological changes in other cell types contributing to the multiple cellular pathologies observed in SCZ. We will:
1. Identify cell type-specific gene regulatory networks involved in SCZ (SCZ-GRNs) in prefrontal cortical excitatory cells by analysis of four distinct SCZ mouse models.
2. Confirm putative SCZ-GRNs in patient material using in situ transcriptomics on postmortem brains and connect to clinical features via collaboration with genomic studies in Sweden and Denmark.
3. Functionally investigate the effects of perturbing excitatory cell SCZ-GRNs on other cell types.
Single-cell RNA-seq, providing insights into the molecular properties of individual cells, and modern molecular tools for perturbing transcription in a cell type-specific way opens up for new knowledge of mechanisms underlying SCZ pathology. My work will identify causal relationships that can be exploited for the development of strategies for personalized treatment.
Max ERC Funding
2 064 414 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym SECRETE-HF
Project SECRETED FACTORS IN CARDIAC REMODELING PROVOKE TUMORIGENESIS AND END ORGAN DAMAGE IN HEART FAILURE
Researcher (PI) Rudolf Allert DE BOER
Host Institution (HI) ACADEMISCH ZIEKENHUIS GRONINGEN
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary The objective of SECRETE-HF is to demonstrate the effects of secreted factors from failing hearts to explain the etiology of multimorbidity in heart failure (HF). The project focuses on two co-morbid patterns: 1) the emerging susceptibility of HF patients for incident cancer, 2) the more established co-morbid conditions of renal, liver and pulmonary disease in HF. The rationale is:
• HF treatment has improved, yet morbidity and mortality remain high, which can be attributed to co-morbid conditions rather than pump failure alone.
• HF treatment is heart-oriented, neglecting the systemic effects that come with HF, and the associated morbidity and mortality.
• Using innovative experimental approaches such as organ transplant models, target finding, and deep phenotyping of clinical databases I will dissect HF-derived effects on tumor growth and organ damage.
OBJECTIVES
1. To establish the effects of HF, due to different etiologies, using the state-of-the-art heart transplantation murine model with (spontaneous) formation of colon and renal tumors, and phenotype tumor growth, as well as the main HF-affected organs: kidney, liver and lungs.
2. Identification of the cardiac secretome using unbiased approaches.
3. Integrate the results and identify overlapping and diverse factors from different HF forms, and their consequences for tumor growth and kidney/liver/lung remodeling.
4. Validate discoveries in human cohorts with data on incident HF, cancer and organ function.
5. Create clinical algorithms to detect, monitor and act on extra-cardiac disease.
WORKPACKAGES
WP 1: Create HF, murine heart transplantation models; phenotype tumor growth and organ involvement.
WP 2: Explore the proteomic, metabolomic and extracellular vesicle profiles from HF subforms.
WP 3: Validate secreted factors in vitro and in vivo.
WP 4: Validate human relevance in large population-based cohorts with unique phenotyping.
WP 5: Describe added value of novel markers and design clinical
Summary
The objective of SECRETE-HF is to demonstrate the effects of secreted factors from failing hearts to explain the etiology of multimorbidity in heart failure (HF). The project focuses on two co-morbid patterns: 1) the emerging susceptibility of HF patients for incident cancer, 2) the more established co-morbid conditions of renal, liver and pulmonary disease in HF. The rationale is:
• HF treatment has improved, yet morbidity and mortality remain high, which can be attributed to co-morbid conditions rather than pump failure alone.
• HF treatment is heart-oriented, neglecting the systemic effects that come with HF, and the associated morbidity and mortality.
• Using innovative experimental approaches such as organ transplant models, target finding, and deep phenotyping of clinical databases I will dissect HF-derived effects on tumor growth and organ damage.
OBJECTIVES
1. To establish the effects of HF, due to different etiologies, using the state-of-the-art heart transplantation murine model with (spontaneous) formation of colon and renal tumors, and phenotype tumor growth, as well as the main HF-affected organs: kidney, liver and lungs.
2. Identification of the cardiac secretome using unbiased approaches.
3. Integrate the results and identify overlapping and diverse factors from different HF forms, and their consequences for tumor growth and kidney/liver/lung remodeling.
4. Validate discoveries in human cohorts with data on incident HF, cancer and organ function.
5. Create clinical algorithms to detect, monitor and act on extra-cardiac disease.
WORKPACKAGES
WP 1: Create HF, murine heart transplantation models; phenotype tumor growth and organ involvement.
WP 2: Explore the proteomic, metabolomic and extracellular vesicle profiles from HF subforms.
WP 3: Validate secreted factors in vitro and in vivo.
WP 4: Validate human relevance in large population-based cohorts with unique phenotyping.
WP 5: Describe added value of novel markers and design clinical
Max ERC Funding
1 999 861 €
Duration
Start date: 2019-07-01, End date: 2024-06-30
Project acronym SUPRAVACC
Project Supramolecular engineering of glycan-decorated peptides as synthetic vaccines
Researcher (PI) Pol BESENIUS
Host Institution (HI) JOHANNES GUTENBERG-UNIVERSITAT MAINZ
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary The main and most important feature of vaccines is the induction of an immunological memory response, which is key to providing long-term protection against pathogens. The current strategies for potent antibacterial and antiviral vaccines employ conjugation of pathogen specific entities onto carrier proteins, and are limited to formulations that suffer from low stability and short shelf-lives, and are thus not viable in developing countries. Strategies for the development of new vaccinations against endogenous diseases like cancer further remain an unmet challenge, since current methodologies suffer from a lack of a modular and tailored vaccine-specific functionalisation. I therefore propose a radically new design approach in the development of fully synthetic molecular vaccines. My team will synthesise carbohydrate and glycopeptide appended epitopes that are grafted onto supramolecular building blocks. These units can be individually designed to attach disease specific antigens and immunostimulants. Due to their self-assembling properties into nanoscaled pathogen mimetic particles, they serve as a supramolecular subunit vaccine toolbox. By developing a universal supramolecular polymer platform, we will construct multipotent vaccines from glycan-decorated peptides, that combine the activity of protein conjugates with the facile handling, precise composition and increased stability of traditional small molecule pharmaceutical compounds.
SUPRAVACC will pioneer the design of minimalistic and broadly applicable vaccines, and will evaluate the supramolecular engineering approach for immunisations against antibacterial diseases, as well as for applications as antitumour vaccine candidates. The fundamental insights gained will drive a paradigm shift in the design and preparation of vaccine candidates in academic and industrial research laboratories.
Summary
The main and most important feature of vaccines is the induction of an immunological memory response, which is key to providing long-term protection against pathogens. The current strategies for potent antibacterial and antiviral vaccines employ conjugation of pathogen specific entities onto carrier proteins, and are limited to formulations that suffer from low stability and short shelf-lives, and are thus not viable in developing countries. Strategies for the development of new vaccinations against endogenous diseases like cancer further remain an unmet challenge, since current methodologies suffer from a lack of a modular and tailored vaccine-specific functionalisation. I therefore propose a radically new design approach in the development of fully synthetic molecular vaccines. My team will synthesise carbohydrate and glycopeptide appended epitopes that are grafted onto supramolecular building blocks. These units can be individually designed to attach disease specific antigens and immunostimulants. Due to their self-assembling properties into nanoscaled pathogen mimetic particles, they serve as a supramolecular subunit vaccine toolbox. By developing a universal supramolecular polymer platform, we will construct multipotent vaccines from glycan-decorated peptides, that combine the activity of protein conjugates with the facile handling, precise composition and increased stability of traditional small molecule pharmaceutical compounds.
SUPRAVACC will pioneer the design of minimalistic and broadly applicable vaccines, and will evaluate the supramolecular engineering approach for immunisations against antibacterial diseases, as well as for applications as antitumour vaccine candidates. The fundamental insights gained will drive a paradigm shift in the design and preparation of vaccine candidates in academic and industrial research laboratories.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym SynapSeek
Project Learning the shape of synaptic plasticity rules for neuronal architectures and function through machine learning.
Researcher (PI) Tim VOGELS
Host Institution (HI) THE CHANCELLOR, MASTERS AND SCHOLARS OF THE UNIVERSITY OF OXFORD
Call Details Consolidator Grant (CoG), LS5, ERC-2018-COG
Summary How do we learn to dance, play an instrument, or a game as complex as chess or go? How do we make a memory? The common answer to these questions is “through synaptic plasticity”, through changing the synaptic connectivity of neural circuits so that representative brain activity can be reliably triggered. Such connectivity changes are governed by rules, i.e., synaptic mechanisms which monitor the activity of their environment and stereotypically strengthen or weaken synapses accordingly. The shape and mode of operation of these rules is still largely unknown: For the more than hundred different connection types in cortical circuits, only a handful of rules has been described at all. Similarly, testing observed rules in simulations of cortical function has only seen limited success. Our slow progress is due to the extraordinary difficulty of measuring and observing synapses without interference.
Here, we propose a new approach. By utilizing the growing power of machine learning methods we can deduce synaptic plasticity rules directly. Newly developed search algorithms and sheer computational power allow us to integrate published data and infer synaptic rules in silico. We aim to (1) develop a new mathematical expression of synaptic plasticity rules, experimentally appropriate and flexible enough to be implemented in a Machine Learning framework, dubbed SYNAPSEEK. Next (2), we will apply SYNAPSEEK to deduce the rules for building various neural structures with increasing complexity. Finally (3), we will incorporate additional constraints to SYNAPSEEK to develop synaptic rules that shape network function as much as its structure. Our work will establish, for the first time, canonical sets of synaptic plasticity rules, based on the circuit structure they must produce, and the function they are meant to support. SYNAPSEEK will have immediate and wide ranging applications, from a basic understanding of cortical development to better protocols for Deep Brain Stimulation.
Summary
How do we learn to dance, play an instrument, or a game as complex as chess or go? How do we make a memory? The common answer to these questions is “through synaptic plasticity”, through changing the synaptic connectivity of neural circuits so that representative brain activity can be reliably triggered. Such connectivity changes are governed by rules, i.e., synaptic mechanisms which monitor the activity of their environment and stereotypically strengthen or weaken synapses accordingly. The shape and mode of operation of these rules is still largely unknown: For the more than hundred different connection types in cortical circuits, only a handful of rules has been described at all. Similarly, testing observed rules in simulations of cortical function has only seen limited success. Our slow progress is due to the extraordinary difficulty of measuring and observing synapses without interference.
Here, we propose a new approach. By utilizing the growing power of machine learning methods we can deduce synaptic plasticity rules directly. Newly developed search algorithms and sheer computational power allow us to integrate published data and infer synaptic rules in silico. We aim to (1) develop a new mathematical expression of synaptic plasticity rules, experimentally appropriate and flexible enough to be implemented in a Machine Learning framework, dubbed SYNAPSEEK. Next (2), we will apply SYNAPSEEK to deduce the rules for building various neural structures with increasing complexity. Finally (3), we will incorporate additional constraints to SYNAPSEEK to develop synaptic rules that shape network function as much as its structure. Our work will establish, for the first time, canonical sets of synaptic plasticity rules, based on the circuit structure they must produce, and the function they are meant to support. SYNAPSEEK will have immediate and wide ranging applications, from a basic understanding of cortical development to better protocols for Deep Brain Stimulation.
Max ERC Funding
1 798 605 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym T2DCP
Project Development of Thiophene Based Conjugated Polymers in Two Dimensions
Researcher (PI) Xinliang FENG
Host Institution (HI) TECHNISCHE UNIVERSITAET DRESDEN
Call Details Consolidator Grant (CoG), PE5, ERC-2018-COG
Summary The proceeding inexorable digitalisation of modern economics and society creates a steadily increasing demand on smart devices in the context of the industrial internet and the internet of things. To meet future requirements, organic electronics is a disruptive technology featuring low-cost, robust, lightweight, flexible and affordable devices based on organic small molecules and polymers. In contrast to the boosting development of linear conjugated polymers and their applications in organic electronics, the successive increase of dimensionality by connecting multiple strands towards two-dimensional (2D) conjugated polymers remains largely unexplored. In this project, we will develop unprecedented thiophene-based double- and triple-strand conjugated polymers to 2D conjugated polymers (T2DCPs) for organic electronics with tailorable electronic band gap at the molecular level for superior performance in terms of charge carrier mobility, and defect tolerance enabled by the increased dimensionality. In this respect, we aim to establish versatile but also reliable solution-based synthesis strategies (one-pot solvothermal, two-step metal-templating reaction and interfacial soft-templating route) employing thiophene monomers rendering T2DCPs with entirely C=C/Ar-Ar backbone. We will further establish ground-breaking one-pot synthesis of donor-acceptor type T2DCPs featuring lower band gap and unique charge transport behavior. By employing designed thiophene-based monomers and linkage topologies, we will accomplish optical and energy gap engineering, control of the molecular weight (or crystalline domain size), and conjugation channel densities. The consequence is that we will explore the key functions of this intriguing class of semiconducting polymers. As the key achievements, we expect to establish a novel solution-based chemistry, delineation of reliable structure-property relationships and superior device performance of T2DCPs for organic field effect transistors.
Summary
The proceeding inexorable digitalisation of modern economics and society creates a steadily increasing demand on smart devices in the context of the industrial internet and the internet of things. To meet future requirements, organic electronics is a disruptive technology featuring low-cost, robust, lightweight, flexible and affordable devices based on organic small molecules and polymers. In contrast to the boosting development of linear conjugated polymers and their applications in organic electronics, the successive increase of dimensionality by connecting multiple strands towards two-dimensional (2D) conjugated polymers remains largely unexplored. In this project, we will develop unprecedented thiophene-based double- and triple-strand conjugated polymers to 2D conjugated polymers (T2DCPs) for organic electronics with tailorable electronic band gap at the molecular level for superior performance in terms of charge carrier mobility, and defect tolerance enabled by the increased dimensionality. In this respect, we aim to establish versatile but also reliable solution-based synthesis strategies (one-pot solvothermal, two-step metal-templating reaction and interfacial soft-templating route) employing thiophene monomers rendering T2DCPs with entirely C=C/Ar-Ar backbone. We will further establish ground-breaking one-pot synthesis of donor-acceptor type T2DCPs featuring lower band gap and unique charge transport behavior. By employing designed thiophene-based monomers and linkage topologies, we will accomplish optical and energy gap engineering, control of the molecular weight (or crystalline domain size), and conjugation channel densities. The consequence is that we will explore the key functions of this intriguing class of semiconducting polymers. As the key achievements, we expect to establish a novel solution-based chemistry, delineation of reliable structure-property relationships and superior device performance of T2DCPs for organic field effect transistors.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-03-01, End date: 2024-02-29
Project acronym TALE
Project Therapeutic Allele Engineering: A novel technology for cell therapy
Researcher (PI) Lukas JEKER
Host Institution (HI) UNIVERSITAT BASEL
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary We are currently witnessing a revolution in cell therapies that are routed in decades of basic research in genetics, cell biology and immunology. A deep understanding of mammalian, and in particular immune, cells is currently being translated into highly efficient cell-based therapeutics. Technologic breakthroughs in genetic and genome engineering are further fueling the generation of customized, high precision therapies that are based on cells as “smart drugs”. For instance, reprogramming immune killer cells to recognize B cell leukemias resulted in unprecedented clinical responses in treatment-resistant and relapsed patients. However, currently only very few, highly selected patients benefit from these developments. A fundamental problem of today’s cell therapies is that transferred cells cannot be distinguished from host cells. We have developed “allele engineering”, a new technology that solves this challenge. Here, we outline how allele engineering will improve the safety and efficacy of cell therapies. We will 1) generate a non-viral, DNA-free safety/shielding switch 2) develop a radically new curative approach to acute myeloid leukemia 3) rationally design a safe allele engineering solution for human therapy and 4) use allele engineering as a curative therapy of scurfy syndrome, a lethal monogenic autoimmune disease. Allele engineering enables completely new treatment strategies and can be applied to any surface protein. Therefore, I anticipate that the results will have a major impact on the field.
Summary
We are currently witnessing a revolution in cell therapies that are routed in decades of basic research in genetics, cell biology and immunology. A deep understanding of mammalian, and in particular immune, cells is currently being translated into highly efficient cell-based therapeutics. Technologic breakthroughs in genetic and genome engineering are further fueling the generation of customized, high precision therapies that are based on cells as “smart drugs”. For instance, reprogramming immune killer cells to recognize B cell leukemias resulted in unprecedented clinical responses in treatment-resistant and relapsed patients. However, currently only very few, highly selected patients benefit from these developments. A fundamental problem of today’s cell therapies is that transferred cells cannot be distinguished from host cells. We have developed “allele engineering”, a new technology that solves this challenge. Here, we outline how allele engineering will improve the safety and efficacy of cell therapies. We will 1) generate a non-viral, DNA-free safety/shielding switch 2) develop a radically new curative approach to acute myeloid leukemia 3) rationally design a safe allele engineering solution for human therapy and 4) use allele engineering as a curative therapy of scurfy syndrome, a lethal monogenic autoimmune disease. Allele engineering enables completely new treatment strategies and can be applied to any surface protein. Therefore, I anticipate that the results will have a major impact on the field.
Max ERC Funding
2 397 082 €
Duration
Start date: 2019-06-01, End date: 2024-05-31
Project acronym UreaCa
Project Deciphering the metabolic roles of the urea-cycle pathway in carcinogenesis for improving diagnosis and therapy
Researcher (PI) Ayelet EREZ
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary Almost 100 years ago, Warburg described a metabolic change in energy flux that occurs during carcinogenesis. Since then, multiple studies have demonstrated how anabolic synthesis of macromolecules can be altered to support cancer cell progression. Yet, the potential effect of altered catabolic degradation of macromolecules on tumour carcinogenesis has been much less studied.
The urea cycle (UC) is the main catabolic pathway by which mammals excrete waste nitrogen. Although the complete UC pathway is liver-specific, most tissues express different combinations of UC enzymes according to the cellular needs. Surprisingly, we find that changes in expression of UC components causing UC dysregulation, (UCD) is a global phenomenon in cancer, metabolically augmenting net nitrogen usage for the synthesis of macromolecules by reducing nitrogen waste. This metabolic alteration is associated with poor patient prognosis. Thus, we hypothesise that UCD provides a major metabolic advantage to multiple aspects of carcinogenesis and as such, leads to specific, identifiable genomic and biochemical signatures, with implications for cancer diagnosis and therapy.
To pursue our hypothesis, we will incorporate state-of-the-art comparative genomic, peptidomic, metabolomic, and molecular approaches to explore this scientific “blind spot” of nitrogen metabolism in carcinogenesis. We will investigate how UCD causally affects carcinogenesis, by characterising tumour-specific functions of UC enzymes (Aim I), correlating tumour phenotypes with systemic biomarkers (Aim II), and testing the treatment efficacy of drug combinations targeting UCD in cancers (Aim III).
Our proposal, strengthened by my training as a physician scientist, harbours considerable potential for translational diagnostic and therapeutic utility of our findings, enabling us to i) identify new diagnostic biomarkers for monitoring cancer initiation and progression and ii) predict and enhance the therapeutic response.
Summary
Almost 100 years ago, Warburg described a metabolic change in energy flux that occurs during carcinogenesis. Since then, multiple studies have demonstrated how anabolic synthesis of macromolecules can be altered to support cancer cell progression. Yet, the potential effect of altered catabolic degradation of macromolecules on tumour carcinogenesis has been much less studied.
The urea cycle (UC) is the main catabolic pathway by which mammals excrete waste nitrogen. Although the complete UC pathway is liver-specific, most tissues express different combinations of UC enzymes according to the cellular needs. Surprisingly, we find that changes in expression of UC components causing UC dysregulation, (UCD) is a global phenomenon in cancer, metabolically augmenting net nitrogen usage for the synthesis of macromolecules by reducing nitrogen waste. This metabolic alteration is associated with poor patient prognosis. Thus, we hypothesise that UCD provides a major metabolic advantage to multiple aspects of carcinogenesis and as such, leads to specific, identifiable genomic and biochemical signatures, with implications for cancer diagnosis and therapy.
To pursue our hypothesis, we will incorporate state-of-the-art comparative genomic, peptidomic, metabolomic, and molecular approaches to explore this scientific “blind spot” of nitrogen metabolism in carcinogenesis. We will investigate how UCD causally affects carcinogenesis, by characterising tumour-specific functions of UC enzymes (Aim I), correlating tumour phenotypes with systemic biomarkers (Aim II), and testing the treatment efficacy of drug combinations targeting UCD in cancers (Aim III).
Our proposal, strengthened by my training as a physician scientist, harbours considerable potential for translational diagnostic and therapeutic utility of our findings, enabling us to i) identify new diagnostic biomarkers for monitoring cancer initiation and progression and ii) predict and enhance the therapeutic response.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-05-01, End date: 2024-04-30
Project acronym VesselNet
Project Engineering Composite Tissues for Facial Reconstruction
Researcher (PI) Shulamit Levenberg
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary Facial reconstruction usually involves the use of autologous grafts or composite tissue allografts, which are highly complex tissues that pose significant challenges to tissue engineering experts. Tissue engineering of independent facial elements, e.g., bone, adipose, skin and muscle tissues, has been demonstrated. However, to date, no composite soft tissues composed of multiple facial layers have been created. Composite facial tissue engineering will require proper innervation and vascularization, essential to support generation of large thick implants. However, techniques for effective innervation of engineered tissues are currently insufficient and generation of well-vascularized large and thick engineered tissues is still one of the major obstacles limiting their translation to the clinic. Our goal is to engineer thick, composite, human-scale, facial tissues (muscle-adipose-dermis composite, and bone) of a personally adaptable shape, that will be vascularized in-vitro, and innervated upon transplantation. Our concept is to create in-vitro a functional vascular network (VesselNet), composed of both large and small vessels, within engineered constructs, which will allow for the generation of thick engineered tissues under continuous flow conditions. 3D bio-printing techniques will be applied to create the engineered tissues. These tissues will serve as a model to study mechanisms involved in vessel anastomosis, and tissue organization and stabilization. The applicability of the engineered composite soft and bone tissues will be evaluated in facial, breast and abdominal wall defect reconstruction models, and in an open fracture model. Such engineered large-scale composite tissues are expected to have a major impact on reconstructive surgery and will shed light on yet unknown tissue organization mechanisms.
Summary
Facial reconstruction usually involves the use of autologous grafts or composite tissue allografts, which are highly complex tissues that pose significant challenges to tissue engineering experts. Tissue engineering of independent facial elements, e.g., bone, adipose, skin and muscle tissues, has been demonstrated. However, to date, no composite soft tissues composed of multiple facial layers have been created. Composite facial tissue engineering will require proper innervation and vascularization, essential to support generation of large thick implants. However, techniques for effective innervation of engineered tissues are currently insufficient and generation of well-vascularized large and thick engineered tissues is still one of the major obstacles limiting their translation to the clinic. Our goal is to engineer thick, composite, human-scale, facial tissues (muscle-adipose-dermis composite, and bone) of a personally adaptable shape, that will be vascularized in-vitro, and innervated upon transplantation. Our concept is to create in-vitro a functional vascular network (VesselNet), composed of both large and small vessels, within engineered constructs, which will allow for the generation of thick engineered tissues under continuous flow conditions. 3D bio-printing techniques will be applied to create the engineered tissues. These tissues will serve as a model to study mechanisms involved in vessel anastomosis, and tissue organization and stabilization. The applicability of the engineered composite soft and bone tissues will be evaluated in facial, breast and abdominal wall defect reconstruction models, and in an open fracture model. Such engineered large-scale composite tissues are expected to have a major impact on reconstructive surgery and will shed light on yet unknown tissue organization mechanisms.
Max ERC Funding
2 375 000 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym ViroPedTher
Project Oncolytic viruses for the treatment of pediatric brain tumors: An integrated clinical and lab approach
Researcher (PI) marta ALONSO-ROLDAN
Host Institution (HI) UNIVERSIDAD DE NAVARRA
Call Details Consolidator Grant (CoG), LS7, ERC-2018-COG
Summary The overreaching goal of my lab is to improve the prognosis of patients with high-risk pediatric brain tumors. To this end, I propose to integrate clinical and lab-based research to develop tumor-targeted oncolytic adenoviruses with the capacity to elicit a therapeutic immune response in those tumors. Our research will use novel and relevant models to accomplish the experimental aims. We have previously worked with Delta-24-RGD (DNX-2401) a replication-competent adenovirus that has been translated to the clinical scenario. In 2017, the first clinical trial phase I with DNX-2401 for newly diagnosed Diffuse Intrinsic Pontine Gliomas (DIPG; a lethal pediatric brain tumor) opened propelled by my team. Preliminary results from the first trials revealed that the intratumoral injection of the virus instigated an initial phase of oncolysis followed by a delayed inflammatory response that ultimately resulted in complete regression in a subset of the patients without associated toxicities. I hypothesized that enhancement of the immune component of the DNX-2401-based therapy will result in the complete regression of the vast majority of pediatric brain tumors. In our specific approach, we propose to understand the immune microenvironment of DIPGs and the response to viral therapy in the context of the trial. Moreover, that knowledge will leverage the design of Delta-24-based adenoviruses to recruit lymphocytes to the tumor with the competence of different type of ligands to activate the tumor infiltrating lymphocytes. I expect that this combinatorial innovative treatment will efficiently challenge the profound and inherent tumor immunosuppression and, in turn, will elicit a robust anti-tumor immune response resulting in the significant improvement of the prognosis and quality of life of patients with pediatric brain tumors. This project has the potential to produce a vertical advance in the field of pediatric oncology.
Summary
The overreaching goal of my lab is to improve the prognosis of patients with high-risk pediatric brain tumors. To this end, I propose to integrate clinical and lab-based research to develop tumor-targeted oncolytic adenoviruses with the capacity to elicit a therapeutic immune response in those tumors. Our research will use novel and relevant models to accomplish the experimental aims. We have previously worked with Delta-24-RGD (DNX-2401) a replication-competent adenovirus that has been translated to the clinical scenario. In 2017, the first clinical trial phase I with DNX-2401 for newly diagnosed Diffuse Intrinsic Pontine Gliomas (DIPG; a lethal pediatric brain tumor) opened propelled by my team. Preliminary results from the first trials revealed that the intratumoral injection of the virus instigated an initial phase of oncolysis followed by a delayed inflammatory response that ultimately resulted in complete regression in a subset of the patients without associated toxicities. I hypothesized that enhancement of the immune component of the DNX-2401-based therapy will result in the complete regression of the vast majority of pediatric brain tumors. In our specific approach, we propose to understand the immune microenvironment of DIPGs and the response to viral therapy in the context of the trial. Moreover, that knowledge will leverage the design of Delta-24-based adenoviruses to recruit lymphocytes to the tumor with the competence of different type of ligands to activate the tumor infiltrating lymphocytes. I expect that this combinatorial innovative treatment will efficiently challenge the profound and inherent tumor immunosuppression and, in turn, will elicit a robust anti-tumor immune response resulting in the significant improvement of the prognosis and quality of life of patients with pediatric brain tumors. This project has the potential to produce a vertical advance in the field of pediatric oncology.
Max ERC Funding
2 000 000 €
Duration
Start date: 2019-03-01, End date: 2024-02-29