Project acronym CHAI
Project Cardiovascular Health effects of Air pollution in Andhra Pradesh, India
Researcher (PI) Cathryn Tonne
Host Institution (HI) FUNDACION PRIVADA INSTITUTO DE SALUD GLOBAL BARCELONA
Call Details Starting Grant (StG), LS7, ERC-2013-StG
Summary While there is convincing evidence that exposure to particulate air pollution causes cardiovascular mortality and morbidity, nearly all of this evidence is based on populations in high-income countries where concentrations are relatively low. There is large uncertainty regarding the relationship between combustion particles and cardiovascular risk for concentrations higher than outdoor concentrations in urban areas of high-income countries and lower than active smoking. Exposures for our study population are likely to be in this range.
We will investigate the cardiovascular health effects of exposure to particles from outdoor and household sources within a prospective cohort in Andhra Pradesh, India. Firstly, we will characterise exposure of participants using an integrated approach utilising outdoor mobile monitoring, personal monitoring, and questionnaire data. We will then collect data on participants’ activities and location using ‘life-logging’ from which activities driving exposure can be identified. Finally, we will quantify the association between exposure to particles and biomarkers of atherosclerosis.
This research will shed light on the relationship between particles and cardiovascular risk at concentration ranges where there is the largest uncertainty. It will provide some of the first evidence of the cardiovascular health effects of medium-term exposure to particulate air pollution outside of a high-income country. The research will also provide evidence regarding the relative contribution of sources and activities linked to high exposure, forming the basis of recommendations for exposure reduction.
Summary
While there is convincing evidence that exposure to particulate air pollution causes cardiovascular mortality and morbidity, nearly all of this evidence is based on populations in high-income countries where concentrations are relatively low. There is large uncertainty regarding the relationship between combustion particles and cardiovascular risk for concentrations higher than outdoor concentrations in urban areas of high-income countries and lower than active smoking. Exposures for our study population are likely to be in this range.
We will investigate the cardiovascular health effects of exposure to particles from outdoor and household sources within a prospective cohort in Andhra Pradesh, India. Firstly, we will characterise exposure of participants using an integrated approach utilising outdoor mobile monitoring, personal monitoring, and questionnaire data. We will then collect data on participants’ activities and location using ‘life-logging’ from which activities driving exposure can be identified. Finally, we will quantify the association between exposure to particles and biomarkers of atherosclerosis.
This research will shed light on the relationship between particles and cardiovascular risk at concentration ranges where there is the largest uncertainty. It will provide some of the first evidence of the cardiovascular health effects of medium-term exposure to particulate air pollution outside of a high-income country. The research will also provide evidence regarding the relative contribution of sources and activities linked to high exposure, forming the basis of recommendations for exposure reduction.
Max ERC Funding
1 200 000 €
Duration
Start date: 2015-01-01, End date: 2018-12-31
Project acronym HIVBIOCHIP
Project A POINT-OF-CARE BIOCHIP FOR HIV MONITORING IN THE DEVELOPING WORLD
Researcher (PI) Nikolaos Chronis
Host Institution (HI) "NATIONAL CENTER FOR SCIENTIFIC RESEARCH ""DEMOKRITOS"""
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary HIV/AIDS is one of the most destructive pandemics in human history, responsible for more than 25 million deaths. More than 30 million people live with limited or no access to therapeutic treatments, mainly due to the high cost of highly active antiretroviral therapies (HAART) and current diagnostic tests as well as due to the lack of basic infrastructure (e.g. lack of electricity, no trained personnel) that can support these tests. The need for innovative, inexpensive diagnostic instrumentation technology that can be used in resource-limited settings is immediate.
While programs that offer free HAART are being implemented in resource-limited settings, no diagnostic tests are available for evaluating the efficacy of HAART provided for the reasons mentioned above. Efficient management of HAART requires monitoring the course of HIV infection over time. The World Health Organization (WHO) recommends the CD4 T-cell count test for monitoring the clinical status of HIV individuals in resource-limited settings.
We propose to develop a portable, inexpensive, MEMS (MicroElectroMechanical Systems)-based, imaging system for counting the absolute number of CD4 cells from 1 l of whole blood. We use the term ‘imaging system’ to denote the different approach we follow for counting CD4 cells: rather the reading one by one singles cells (as it is done with flow cytometry), our system can image simultaneously thousands of individual cells, pre-assembled on the surface of a biochip. Although the proposed imaging system can replace current expensive cell counting instrumentation, our goal is to develop a system that can reach the end-user wherever limited infrastructure is present and no access to a hospital or clinic is possible. Such technology will not only enable to monitor the efficacy of an individual’s HAART in the developing world, but it will make more medicines available by identifying patients who need a treatment from patients who do not need it.
Summary
HIV/AIDS is one of the most destructive pandemics in human history, responsible for more than 25 million deaths. More than 30 million people live with limited or no access to therapeutic treatments, mainly due to the high cost of highly active antiretroviral therapies (HAART) and current diagnostic tests as well as due to the lack of basic infrastructure (e.g. lack of electricity, no trained personnel) that can support these tests. The need for innovative, inexpensive diagnostic instrumentation technology that can be used in resource-limited settings is immediate.
While programs that offer free HAART are being implemented in resource-limited settings, no diagnostic tests are available for evaluating the efficacy of HAART provided for the reasons mentioned above. Efficient management of HAART requires monitoring the course of HIV infection over time. The World Health Organization (WHO) recommends the CD4 T-cell count test for monitoring the clinical status of HIV individuals in resource-limited settings.
We propose to develop a portable, inexpensive, MEMS (MicroElectroMechanical Systems)-based, imaging system for counting the absolute number of CD4 cells from 1 l of whole blood. We use the term ‘imaging system’ to denote the different approach we follow for counting CD4 cells: rather the reading one by one singles cells (as it is done with flow cytometry), our system can image simultaneously thousands of individual cells, pre-assembled on the surface of a biochip. Although the proposed imaging system can replace current expensive cell counting instrumentation, our goal is to develop a system that can reach the end-user wherever limited infrastructure is present and no access to a hospital or clinic is possible. Such technology will not only enable to monitor the efficacy of an individual’s HAART in the developing world, but it will make more medicines available by identifying patients who need a treatment from patients who do not need it.
Max ERC Funding
1 986 000 €
Duration
Start date: 2012-06-01, End date: 2017-05-31
Project acronym IMMA
Project Integrating the Multiple Meta-Analysis: a framework for evaluating and ranking multiple health care technologies
Researcher (PI) Georgia Salanti
Host Institution (HI) PANEPISTIMIO IOANNINON
Call Details Starting Grant (StG), LS7, ERC-2010-StG_20091118
Summary Health care practitioners face daily questions and make choices regarding the effectiveness and quality of several health technologies (e.g. alternative interventions). On this regard they usually consider meta-analysis; the statistical synthesis of results from relevant experiments. The main drawback of the current state of the art is that meta-analysis focuses on comparing only two alternatives. However, clinicians and scientists need to know the relative ranking of a set of alternative options and not only whether option A is better than B. There is an urgent need to establish and disseminate a robust framework for selecting among many treatment options, possibly after taking into account environmental and genetic interactions. The goal of the proposed project is to provide this by establishing and disseminating a revolutionary evidence synthesis toolkit. Its main methodological vehicle is a flexible statistical framework using Bayesian techniques for multiple-treatments meta-analysis. This will enable the relative ranking of all alternative health care options, will allow comprehensive use of all available data, will improve precision and confidence in the conclusions and will answer methodological questions related to bias. Once established in clinical epidemiology, the tool will be extended to genetic epidemiology to account for multiple genetic markers, environmental factors and effects of treatments. Based on ongoing collaborations with teams undertaking applied health care research I plan to evaluate the new tool empirically in real-life health care problems such as ranking the pharmacological treatments for osteoarthritis, indicating the best treatments for multiple sclerosis and ranking the vaccines for influenza.
Summary
Health care practitioners face daily questions and make choices regarding the effectiveness and quality of several health technologies (e.g. alternative interventions). On this regard they usually consider meta-analysis; the statistical synthesis of results from relevant experiments. The main drawback of the current state of the art is that meta-analysis focuses on comparing only two alternatives. However, clinicians and scientists need to know the relative ranking of a set of alternative options and not only whether option A is better than B. There is an urgent need to establish and disseminate a robust framework for selecting among many treatment options, possibly after taking into account environmental and genetic interactions. The goal of the proposed project is to provide this by establishing and disseminating a revolutionary evidence synthesis toolkit. Its main methodological vehicle is a flexible statistical framework using Bayesian techniques for multiple-treatments meta-analysis. This will enable the relative ranking of all alternative health care options, will allow comprehensive use of all available data, will improve precision and confidence in the conclusions and will answer methodological questions related to bias. Once established in clinical epidemiology, the tool will be extended to genetic epidemiology to account for multiple genetic markers, environmental factors and effects of treatments. Based on ongoing collaborations with teams undertaking applied health care research I plan to evaluate the new tool empirically in real-life health care problems such as ranking the pharmacological treatments for osteoarthritis, indicating the best treatments for multiple sclerosis and ranking the vaccines for influenza.
Max ERC Funding
592 500 €
Duration
Start date: 2010-10-01, End date: 2015-12-31
Project acronym MYELOMANEXT
Project Integrated next-generation flow cytometry and sequencing to uncover the pathway of curability in multiple myeloma
Researcher (PI) Bruno David Lourenço Paiva
Host Institution (HI) UNIVERSIDAD DE NAVARRA
Call Details Starting Grant (StG), LS7, ERC-2015-STG
Summary Multiple myeloma (MM) represents a unique model to investigate cancer stem cells (CSCs), circulating tumour cells (CTCs), and the mechanisms of malignant transformation and chemoresistance. Despite the substantial improvement in MM patients’ outcome, the vast majority of patients eventually relapse and the disease remains largely incurable. For those patients failing to achieve deep remissions, biologically targeted research on the ultra-chemoresistant minimal residual disease (MRD) clone may allow us to understand the cellular mechanisms driving chemoresistance, and design novel therapeutic to overcome; importantly, such effort should be equally performed on two additional key players: CSCs and CTCs. On the opposite side, it is unquestionable that a selected group of patients does experience long-term survival irrespectively of the depth of response achieved, but we fail to understand the mechanisms driving sustained disease control. Is it because of persistent residual benign clones? Is it because of immune surveillance? Here, we will integrate next-generation flow cytometry and sequencing to define i) the signature of CTCs and ultra-chemoresistant MRD cells, ii) the hierarchical place of putative CSCs, iii) the genomic landscape of benign vs. malignant clones; and iv) the role of immune surveillance to achieve functional cures. Hence, we will characterize for the first-time-ever the highly-professional subclones responsible for malignant transformation, disease dissemination, and dramatic relapses after optimal response to therapy. Noteworthy, the innovative approach of this scientific proposal strongly relies on the use and expertise of highly-sensitive next-generation flow cytometry, coupled with optimized DNA- and RNA-sequencing for low-cell-numbers, and prospective patient samples longitudinally available within the scope of well-controlled clinical trials. Herein, we believe that all requirements are met to conduct this ground-breaking research program.
Summary
Multiple myeloma (MM) represents a unique model to investigate cancer stem cells (CSCs), circulating tumour cells (CTCs), and the mechanisms of malignant transformation and chemoresistance. Despite the substantial improvement in MM patients’ outcome, the vast majority of patients eventually relapse and the disease remains largely incurable. For those patients failing to achieve deep remissions, biologically targeted research on the ultra-chemoresistant minimal residual disease (MRD) clone may allow us to understand the cellular mechanisms driving chemoresistance, and design novel therapeutic to overcome; importantly, such effort should be equally performed on two additional key players: CSCs and CTCs. On the opposite side, it is unquestionable that a selected group of patients does experience long-term survival irrespectively of the depth of response achieved, but we fail to understand the mechanisms driving sustained disease control. Is it because of persistent residual benign clones? Is it because of immune surveillance? Here, we will integrate next-generation flow cytometry and sequencing to define i) the signature of CTCs and ultra-chemoresistant MRD cells, ii) the hierarchical place of putative CSCs, iii) the genomic landscape of benign vs. malignant clones; and iv) the role of immune surveillance to achieve functional cures. Hence, we will characterize for the first-time-ever the highly-professional subclones responsible for malignant transformation, disease dissemination, and dramatic relapses after optimal response to therapy. Noteworthy, the innovative approach of this scientific proposal strongly relies on the use and expertise of highly-sensitive next-generation flow cytometry, coupled with optimized DNA- and RNA-sequencing for low-cell-numbers, and prospective patient samples longitudinally available within the scope of well-controlled clinical trials. Herein, we believe that all requirements are met to conduct this ground-breaking research program.
Max ERC Funding
1 468 606 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym NextGen IO
Project Exploiting the hypoxia response in T cells for Next-Generation Immuno-Oncology
Researcher (PI) Francisco de Asis PALAZON GARCIA
Host Institution (HI) ASOCIACION CENTRO DE INVESTIGACION COOPERATIVA EN BIOCIENCIAS
Call Details Starting Grant (StG), LS7, ERC-2018-STG
Summary NextGen_IO has a core focus on immuno-oncology, specifically on target discovery and drug development, to exploit several opportunities that the hypoxia pathway in T cells offers for the treatment of cancer. It is well recognised that the clinical response of immunotherapies depends on the ability of T-cells to mount an effective effector response, persist in treated patients and avoid exhaustion and toxicities. Several approaches to immunotherapy have shown promise in clinical trials, especially the use of immune checkpoint inhibitors and, more recently, autologous adoptive T-cell therapies. However, current state-of-the-art immunotherapies are only effective in a small fraction of patients, offering a medical need to be addressed in several cancer types. Importantly, the tumor microenvironment has specific features that impact the immune response, including decreased oxygenation, aberrant vascularization and altered nutrient availability; all these influence the success of immunotherapies. During the last 10 years, my research has been focused on elucidating the role of the oxygen sensing machinery in T cell function, and the link of hypoxia-driven metabolism and epigenetic modifications with T cell differentiation into effector and memory T cells within the context of cancer immunotherapy. The current proposal aims to exploit these previous findings with a multi-disciplinary strategy, to deliver several early-stage drug discovery outputs.
The main objectives are:
1. Development of a novel small molecule inhibitor to modulate the hypoxic response in T cells.
2. Therapeutic target discovery in T cells, focused on hypoxia-driven epigenetic modifications.
3. Development of hypoxia-inducible molecular switches for adoptive T cell therapy.
Successful completion of the project will allow me to further innovate and consolidate my position as a leader in this field, harness this pathway for therapeutic potential and explore potential combinatorial approaches.
Summary
NextGen_IO has a core focus on immuno-oncology, specifically on target discovery and drug development, to exploit several opportunities that the hypoxia pathway in T cells offers for the treatment of cancer. It is well recognised that the clinical response of immunotherapies depends on the ability of T-cells to mount an effective effector response, persist in treated patients and avoid exhaustion and toxicities. Several approaches to immunotherapy have shown promise in clinical trials, especially the use of immune checkpoint inhibitors and, more recently, autologous adoptive T-cell therapies. However, current state-of-the-art immunotherapies are only effective in a small fraction of patients, offering a medical need to be addressed in several cancer types. Importantly, the tumor microenvironment has specific features that impact the immune response, including decreased oxygenation, aberrant vascularization and altered nutrient availability; all these influence the success of immunotherapies. During the last 10 years, my research has been focused on elucidating the role of the oxygen sensing machinery in T cell function, and the link of hypoxia-driven metabolism and epigenetic modifications with T cell differentiation into effector and memory T cells within the context of cancer immunotherapy. The current proposal aims to exploit these previous findings with a multi-disciplinary strategy, to deliver several early-stage drug discovery outputs.
The main objectives are:
1. Development of a novel small molecule inhibitor to modulate the hypoxic response in T cells.
2. Therapeutic target discovery in T cells, focused on hypoxia-driven epigenetic modifications.
3. Development of hypoxia-inducible molecular switches for adoptive T cell therapy.
Successful completion of the project will allow me to further innovate and consolidate my position as a leader in this field, harness this pathway for therapeutic potential and explore potential combinatorial approaches.
Max ERC Funding
1 993 575 €
Duration
Start date: 2019-02-01, End date: 2024-01-31
Project acronym OPTICALBULLET
Project Studies of neurosecretion by remote control of exocytosis and endocytosis with ligt
Researcher (PI) Pau Gorostiza
Host Institution (HI) FUNDACIO INSTITUT DE BIOENGINYERIA DE CATALUNYA
Call Details Starting Grant (StG), LS7, ERC-2007-StG
Summary Optical switches are photoisomerizable compounds that allow to remotely controlling the activity of proteins, cells and entire organisms with light. These tools are revolutionizing research in biology with their high selectivity and spatiotemporal resolution. Here we propose to develop and apply optical switches to investigate the fundamental processes of secretion, exocytosis and endocytosis, in a way that is non-invasive, acute, and orthogonal to pharmacological and electrophysiological techniques. The optical control of exocytosis will be carried out by means of photoswitchable, Ca2+-permeable channels (LiGluR and Channelrhodopsin-2) which allow triggering vesicle fusion at single synaptic terminals. This procedure will allow studying vesicle release kinetics and the differences between synapses of the same neuron. The photocontrol of endocytosis will be carried out with: (1) inhibitory peptides of the clathrin pathway modified with an azobenzene crosslinker in order to photomodulate their structure and affinity, and (2) photoswitchable synthetic compounds based on chemical inhibitors of dynamin. Photomodulation of endocytosis in chromaffin cells and neurons will allow interfering with the internalisation of membrane receptors with an unprecedented spatial and temporal control. The use of photoswitchable inhibitors of endocytosis would allow for the first time to manipulate reversibly and with subcellular resolution, the vesicular trafficking of the endocytic pathway. In addition, these photoswitches could reveal how endocytosis regulates spatially receptor activation, controlling cell patterning and cell fate.
Summary
Optical switches are photoisomerizable compounds that allow to remotely controlling the activity of proteins, cells and entire organisms with light. These tools are revolutionizing research in biology with their high selectivity and spatiotemporal resolution. Here we propose to develop and apply optical switches to investigate the fundamental processes of secretion, exocytosis and endocytosis, in a way that is non-invasive, acute, and orthogonal to pharmacological and electrophysiological techniques. The optical control of exocytosis will be carried out by means of photoswitchable, Ca2+-permeable channels (LiGluR and Channelrhodopsin-2) which allow triggering vesicle fusion at single synaptic terminals. This procedure will allow studying vesicle release kinetics and the differences between synapses of the same neuron. The photocontrol of endocytosis will be carried out with: (1) inhibitory peptides of the clathrin pathway modified with an azobenzene crosslinker in order to photomodulate their structure and affinity, and (2) photoswitchable synthetic compounds based on chemical inhibitors of dynamin. Photomodulation of endocytosis in chromaffin cells and neurons will allow interfering with the internalisation of membrane receptors with an unprecedented spatial and temporal control. The use of photoswitchable inhibitors of endocytosis would allow for the first time to manipulate reversibly and with subcellular resolution, the vesicular trafficking of the endocytic pathway. In addition, these photoswitches could reveal how endocytosis regulates spatially receptor activation, controlling cell patterning and cell fate.
Max ERC Funding
1 338 000 €
Duration
Start date: 2008-09-01, End date: 2013-08-31
Project acronym PD-HUMMODEL
Project Elucidating early pathogenic mechanisms of neurodegeneration in Parkinson's disease through a humanized dynamic in vitro model
Researcher (PI) Antonella Consiglio
Host Institution (HI) UNIVERSITAT DE BARCELONA
Call Details Starting Grant (StG), LS7, ERC-2012-StG_20111109
Summary Our understanding of Parkinson’s disease (PD) pathogenesis is currently limited by difficulties in obtaining live neurons from patients and the inability to model the sporadic, most frequent, form of PD. It may be possible to overcome these challenges by reprogramming somatic cells from patients into induced pluripotent stem cells (iPSC). In preliminary studies, we have generated a collection of 50 iPSC lines representing both sporadic PD and familial PD patients, and identified distinct PD-related neurodegeneration phenotypes arising, upon long-term culture, in DAn differentiated from these PD-iPSC. Here, I propose to take advantage of this genuinely human PD model to investigate: i) mechanistic insights responsible for the PD phenotype identified in our model (by combining molecular and biochemical analyses to study mitochondrial function and redox profile, as well as genome-wide transcriptional profile of control versus PD-patient specific iPSC-derived DAn); ii) early functional alterations in patient-specific iPSC-derived DAn, which would predate neurodegeneration signs and provide valuable information as to ways to prevent, rather than rescue, neurodegeneration in PD patients (by electrophysiological recordings in in vitro reconstructed neuronal/glial networks to assess synaptic dynamics together with neuronal excitability); iii) further refinements in our iPSC-based PD model, including the generation of iPSC lines representing asymptomatic patients carrying pathogenic mutations, and the correction of known mutations by gene edition, all of which will allow exploring the relationship between pathogenic mutations and the genetic makeup of patients; and iv) whether DAn degeneration in PD is solely a cell-autonomous phenomenon, or whether it is influenced by an altered cross-talk between DAn and glial cells. These studies may impact significantly on our understanding of PD pathogenesis and on the development of new therapy strategy.
Summary
Our understanding of Parkinson’s disease (PD) pathogenesis is currently limited by difficulties in obtaining live neurons from patients and the inability to model the sporadic, most frequent, form of PD. It may be possible to overcome these challenges by reprogramming somatic cells from patients into induced pluripotent stem cells (iPSC). In preliminary studies, we have generated a collection of 50 iPSC lines representing both sporadic PD and familial PD patients, and identified distinct PD-related neurodegeneration phenotypes arising, upon long-term culture, in DAn differentiated from these PD-iPSC. Here, I propose to take advantage of this genuinely human PD model to investigate: i) mechanistic insights responsible for the PD phenotype identified in our model (by combining molecular and biochemical analyses to study mitochondrial function and redox profile, as well as genome-wide transcriptional profile of control versus PD-patient specific iPSC-derived DAn); ii) early functional alterations in patient-specific iPSC-derived DAn, which would predate neurodegeneration signs and provide valuable information as to ways to prevent, rather than rescue, neurodegeneration in PD patients (by electrophysiological recordings in in vitro reconstructed neuronal/glial networks to assess synaptic dynamics together with neuronal excitability); iii) further refinements in our iPSC-based PD model, including the generation of iPSC lines representing asymptomatic patients carrying pathogenic mutations, and the correction of known mutations by gene edition, all of which will allow exploring the relationship between pathogenic mutations and the genetic makeup of patients; and iv) whether DAn degeneration in PD is solely a cell-autonomous phenomenon, or whether it is influenced by an altered cross-talk between DAn and glial cells. These studies may impact significantly on our understanding of PD pathogenesis and on the development of new therapy strategy.
Max ERC Funding
1 324 802 €
Duration
Start date: 2013-07-01, End date: 2018-06-30
Project acronym REGMAMKID
Project How to regenerate the mammalian kidney
Researcher (PI) Nuria Montserrat Pulido
Host Institution (HI) FUNDACIO INSTITUT DE BIOENGINYERIA DE CATALUNYA
Call Details Starting Grant (StG), LS7, ERC-2014-STG
Summary Kidney non-endocrine functions are primarily performed by millions of individual integral units called nephrons. Although the adults of simple vertebrates (fish, amphibians and reptiles) have the ability to regenerate entire nephrons by a process called “nephron neogenesis”, this capacity is absent in birds and mammals. In this regard we have observed, for the first time, that the murine neonatal kidney is able to generate new nephrons after the resection of the 20 per cent of the kidney mass.
In the same manner, our recent published data on the generation of human kidney organoids ex vivo from kidney disease derived induced pluripotent stem cells (iPS), demonstrates the suitability of iPS technology to establish an unprecedented platform for drug screening, disease modelling and kidney regeneration.
Thus, with support from REGMAMKID we will carry out a dual strategy for kidney regeneration and therapy. For kidney regeneration, we will make use of our newly developed mouse model of neonatal kidney regeneration and identify the molecular and epigenetic drivers responsible for this process. For kidney therapy, we will generate specific kidney cell populations with therapeutic potential such as podocytes and tubular epithelial cells by combining emerging cutting-edge technologies from the fields of cellular reprogramming and pluripotent stem cells differentiation. Making use of the tools developed in REGMAMKID we will model two different genetic kidney disorders: one affecting the podocyte (Congenital Nephrotic Syndrome) and other leading to an excessive proliferation of tubular epithelial cells (Autosomal Dominant Polycistic Disease). These studies will significantly impact on our understanding of kidney disease and healing.
Summary
Kidney non-endocrine functions are primarily performed by millions of individual integral units called nephrons. Although the adults of simple vertebrates (fish, amphibians and reptiles) have the ability to regenerate entire nephrons by a process called “nephron neogenesis”, this capacity is absent in birds and mammals. In this regard we have observed, for the first time, that the murine neonatal kidney is able to generate new nephrons after the resection of the 20 per cent of the kidney mass.
In the same manner, our recent published data on the generation of human kidney organoids ex vivo from kidney disease derived induced pluripotent stem cells (iPS), demonstrates the suitability of iPS technology to establish an unprecedented platform for drug screening, disease modelling and kidney regeneration.
Thus, with support from REGMAMKID we will carry out a dual strategy for kidney regeneration and therapy. For kidney regeneration, we will make use of our newly developed mouse model of neonatal kidney regeneration and identify the molecular and epigenetic drivers responsible for this process. For kidney therapy, we will generate specific kidney cell populations with therapeutic potential such as podocytes and tubular epithelial cells by combining emerging cutting-edge technologies from the fields of cellular reprogramming and pluripotent stem cells differentiation. Making use of the tools developed in REGMAMKID we will model two different genetic kidney disorders: one affecting the podocyte (Congenital Nephrotic Syndrome) and other leading to an excessive proliferation of tubular epithelial cells (Autosomal Dominant Polycistic Disease). These studies will significantly impact on our understanding of kidney disease and healing.
Max ERC Funding
1 499 604 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym SACCO
Project Signals for accommodative responses in humans
Researcher (PI) Robert Montes-Mico
Host Institution (HI) UNIVERSITAT DE VALENCIA
Call Details Starting Grant (StG), LS7, ERC-2012-StG_20111109
Summary "Long-term objectives are to identify optical signals that control accommodation and emmetropization of the eye, and to identify the mechanisms that mediate the optical signals. A primary goal is to determine whether accommodation responds to the wavefront characteristics of light [defocus and higher-order aberrations (HOAs)], without feedback from change in defocus astigmatism or HOAs. Accommodation, pupillary responses and aberrations will be monitored by an imaging simulator that incorporates Hartmann-Shack wavefront sensing, adaptive optics and Hirschberg x-y eye tracking. Adaptive optics and Badal optics will compensate for the refractive error of the eye, alter or remove HOAs, and remove feedback from changes in accommodation and/or HOAs. Accommodation will be driven by defocus with normal HOAs present or removed. To determine whether accommodation responds to wavefront aberration per se, or simply to the shape or skewing of blur, aberrations will be removed while simulation of defocus targets are imaged on the retina, with and without the effects of HOAs on the blur. Standard model that accommodation operates as a closed-loop negative feedback system to maximize the contrast retinal image, will be tested by using adaptive optics to remove defocus, astigmatism and HOAs and by establishing a closed feedback loop between accommodation and target contrast. An alternative hypothesis that accommodation responds to wavefront defocus rather than blur of the retinal image, will be tested by driving accommodation with defocus without feedback from blur, in the absence of HOAs. A final experiment will determine whether the pupil changes independently of lenticular accommodation to provide better acuity in the presence of spherical aberration. Better understanding of the optical signals and mechanisms that the eye used to detect myopic and hyperopic defocus may lead to early detection of individual at risk for developing myopia, and new treatments to prevent/reduce myopia"
Summary
"Long-term objectives are to identify optical signals that control accommodation and emmetropization of the eye, and to identify the mechanisms that mediate the optical signals. A primary goal is to determine whether accommodation responds to the wavefront characteristics of light [defocus and higher-order aberrations (HOAs)], without feedback from change in defocus astigmatism or HOAs. Accommodation, pupillary responses and aberrations will be monitored by an imaging simulator that incorporates Hartmann-Shack wavefront sensing, adaptive optics and Hirschberg x-y eye tracking. Adaptive optics and Badal optics will compensate for the refractive error of the eye, alter or remove HOAs, and remove feedback from changes in accommodation and/or HOAs. Accommodation will be driven by defocus with normal HOAs present or removed. To determine whether accommodation responds to wavefront aberration per se, or simply to the shape or skewing of blur, aberrations will be removed while simulation of defocus targets are imaged on the retina, with and without the effects of HOAs on the blur. Standard model that accommodation operates as a closed-loop negative feedback system to maximize the contrast retinal image, will be tested by using adaptive optics to remove defocus, astigmatism and HOAs and by establishing a closed feedback loop between accommodation and target contrast. An alternative hypothesis that accommodation responds to wavefront defocus rather than blur of the retinal image, will be tested by driving accommodation with defocus without feedback from blur, in the absence of HOAs. A final experiment will determine whether the pupil changes independently of lenticular accommodation to provide better acuity in the presence of spherical aberration. Better understanding of the optical signals and mechanisms that the eye used to detect myopic and hyperopic defocus may lead to early detection of individual at risk for developing myopia, and new treatments to prevent/reduce myopia"
Max ERC Funding
1 384 200 €
Duration
Start date: 2012-11-01, End date: 2017-10-31
Project acronym SUMO
Project Study of the role of protein posttranslational modification by SUMO (Small Ubiquitin-like MOdifier) in abscisic acid signaling and stress responses in plants
Researcher (PI) Luisa Maria Lois
Host Institution (HI) CENTRE DE RECERCA EN AGRIGENOMICA CSIC-IRTA-UAB-UB
Call Details Starting Grant (StG), LS7, ERC-2007-StG
Summary Eukaryotic protein function is regulated in vivo by diverse mechanisms such as protein turnover, regulation of protein activity, localization and protein-protein interactions. These mechanisms involve constitutive or reversible post-translational modifications of specific amino-acid residues in the target protein by molecules of different nature. Ubiquitin and ubiquitin-like modifiers are polypeptides that are covalently attached to a lysine residue in the target protein. SUMO is a member of the ubiquitin family, but it differs from ubiquitin in its cellular function. Whereas protein ubiquitination results in degradation by the 26S proteasome, sumoylation is involved in regulation of protein activity, cellular localization, or protection from ubiquitination. In plants, SUMO plays an important role in biotic and abiotic stress responses, and regulates abscisic acid (ABA) signaling, plant hormone that mediates environmental stress responses, and flowering. In addition, we have found that a functional sumoylation system is essential during seed development, process that is also regulated by ABA at different stages. Our general goal is to investigate the biological role of SUMO in the context of ABA signaling and stress responses in Arabidopsis. For this purpose we will study different aspects of this novel posttranslational regulatory system involving the analysis of the SUMO biological role during seed development and germination, identification of new SUMO targets and dissection of the biological role of the catalase AtCAT3 sumoylation, and the study of molecular factors that could be responsible for recognition of SUMO conjugates. The data generated will contribute to better understanding of this biological process and, eventually, to a thoughtful design of plants with improved agronomical traits. Also, as sumoylation is an evolutionary conserved regulatory system, our work will greatly contribute to understand its mechanism of action in mammals.
Summary
Eukaryotic protein function is regulated in vivo by diverse mechanisms such as protein turnover, regulation of protein activity, localization and protein-protein interactions. These mechanisms involve constitutive or reversible post-translational modifications of specific amino-acid residues in the target protein by molecules of different nature. Ubiquitin and ubiquitin-like modifiers are polypeptides that are covalently attached to a lysine residue in the target protein. SUMO is a member of the ubiquitin family, but it differs from ubiquitin in its cellular function. Whereas protein ubiquitination results in degradation by the 26S proteasome, sumoylation is involved in regulation of protein activity, cellular localization, or protection from ubiquitination. In plants, SUMO plays an important role in biotic and abiotic stress responses, and regulates abscisic acid (ABA) signaling, plant hormone that mediates environmental stress responses, and flowering. In addition, we have found that a functional sumoylation system is essential during seed development, process that is also regulated by ABA at different stages. Our general goal is to investigate the biological role of SUMO in the context of ABA signaling and stress responses in Arabidopsis. For this purpose we will study different aspects of this novel posttranslational regulatory system involving the analysis of the SUMO biological role during seed development and germination, identification of new SUMO targets and dissection of the biological role of the catalase AtCAT3 sumoylation, and the study of molecular factors that could be responsible for recognition of SUMO conjugates. The data generated will contribute to better understanding of this biological process and, eventually, to a thoughtful design of plants with improved agronomical traits. Also, as sumoylation is an evolutionary conserved regulatory system, our work will greatly contribute to understand its mechanism of action in mammals.
Max ERC Funding
1 104 000 €
Duration
Start date: 2008-07-01, End date: 2014-06-30