Project acronym 20SComplexity
Project An integrative approach to uncover the multilevel regulation of 20S proteasome degradation
Researcher (PI) Michal Sharon
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS1, ERC-2014-STG
Summary For many years, the ubiquitin-26S proteasome degradation pathway was considered the primary route for proteasomal degradation. However, it is now becoming clear that proteins can also be targeted for degradation by a ubiquitin-independent mechanism mediated by the core 20S proteasome itself. Although initially believed to be limited to rare exceptions, degradation by the 20S proteasome is now understood to have a wide range of substrates, many of which are key regulatory proteins. Despite its importance, little is known about the mechanisms that control 20S proteasomal degradation, unlike the extensive knowledge acquired over the years concerning degradation by the 26S proteasome. Our overall aim is to reveal the multiple regulatory levels that coordinate the 20S proteasome degradation route.
To achieve this goal we will carry out a comprehensive research program characterizing three distinct levels of 20S proteasome regulation:
Intra-molecular regulation- Revealing the intrinsic molecular switch that activates the latent 20S proteasome.
Inter-molecular regulation- Identifying novel proteins that bind the 20S proteasome to regulate its activity and characterizing their mechanism of function.
Cellular regulatory networks- Unraveling the cellular cues and multiple pathways that influence 20S proteasome activity using a novel systematic and unbiased screening approach.
Our experimental strategy involves the combination of biochemical approaches with native mass spectrometry, cross-linking and fluorescence measurements, complemented by cell biology analyses and high-throughput screening. Such a multidisciplinary approach, integrating in vitro and in vivo findings, will likely provide the much needed knowledge on the 20S proteasome degradation route. When completed, we anticipate that this work will be part of a new paradigm – no longer perceiving the 20S proteasome mediated degradation as a simple and passive event but rather a tightly regulated and coordinated process.
Summary
For many years, the ubiquitin-26S proteasome degradation pathway was considered the primary route for proteasomal degradation. However, it is now becoming clear that proteins can also be targeted for degradation by a ubiquitin-independent mechanism mediated by the core 20S proteasome itself. Although initially believed to be limited to rare exceptions, degradation by the 20S proteasome is now understood to have a wide range of substrates, many of which are key regulatory proteins. Despite its importance, little is known about the mechanisms that control 20S proteasomal degradation, unlike the extensive knowledge acquired over the years concerning degradation by the 26S proteasome. Our overall aim is to reveal the multiple regulatory levels that coordinate the 20S proteasome degradation route.
To achieve this goal we will carry out a comprehensive research program characterizing three distinct levels of 20S proteasome regulation:
Intra-molecular regulation- Revealing the intrinsic molecular switch that activates the latent 20S proteasome.
Inter-molecular regulation- Identifying novel proteins that bind the 20S proteasome to regulate its activity and characterizing their mechanism of function.
Cellular regulatory networks- Unraveling the cellular cues and multiple pathways that influence 20S proteasome activity using a novel systematic and unbiased screening approach.
Our experimental strategy involves the combination of biochemical approaches with native mass spectrometry, cross-linking and fluorescence measurements, complemented by cell biology analyses and high-throughput screening. Such a multidisciplinary approach, integrating in vitro and in vivo findings, will likely provide the much needed knowledge on the 20S proteasome degradation route. When completed, we anticipate that this work will be part of a new paradigm – no longer perceiving the 20S proteasome mediated degradation as a simple and passive event but rather a tightly regulated and coordinated process.
Max ERC Funding
1 500 000 €
Duration
Start date: 2015-04-01, End date: 2020-03-31
Project acronym 3DBrainStrom
Project Brain metastases: Deciphering tumor-stroma interactions in three dimensions for the rational design of nanomedicines
Researcher (PI) Ronit Satchi Fainaro
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Advanced Grant (AdG), LS7, ERC-2018-ADG
Summary Brain metastases represent a major therapeutic challenge. Despite significant breakthroughs in targeted therapies, survival rates of patients with brain metastases remain poor. Nowadays, discovery, development and evaluation of new therapies are performed on human cancer cells grown in 2D on rigid plastic plates followed by in vivo testing in immunodeficient mice. These experimental settings are lacking and constitute a fundamental hurdle for the translation of preclinical discoveries into clinical practice. We propose to establish 3D-printed models of brain metastases (Aim 1), which include brain extracellular matrix, stroma and serum containing immune cells flowing in functional tumor vessels. Our unique models better capture the clinical physio-mechanical tissue properties, signaling pathways, hemodynamics and drug responsiveness. Using our 3D-printed models, we aim to develop two new fronts for identifying novel clinically-relevant molecular drivers (Aim 2) followed by the development of precision nanomedicines (Aim 3). We will exploit our vast experience in anticancer nanomedicines to design three therapeutic approaches that target various cellular compartments involved in brain metastases: 1) Prevention of brain metastatic colonization using targeted nano-vaccines, which elicit antitumor immune response; 2) Intervention of tumor-brain stroma cells crosstalk when brain micrometastases establish; 3) Regression of macrometastatic disease by selectively targeting tumor cells. These approaches will materialize using our libraries of polymeric nanocarriers that selectively accumulate in tumors.
This project will result in a paradigm shift by generating new preclinical cancer models that will bridge the translational gap in cancer therapeutics. The insights and tumor-stroma-targeted nanomedicines developed here will pave the way for prediction of patient outcome, revolutionizing our perception of tumor modelling and consequently the way we prevent and treat cancer.
Summary
Brain metastases represent a major therapeutic challenge. Despite significant breakthroughs in targeted therapies, survival rates of patients with brain metastases remain poor. Nowadays, discovery, development and evaluation of new therapies are performed on human cancer cells grown in 2D on rigid plastic plates followed by in vivo testing in immunodeficient mice. These experimental settings are lacking and constitute a fundamental hurdle for the translation of preclinical discoveries into clinical practice. We propose to establish 3D-printed models of brain metastases (Aim 1), which include brain extracellular matrix, stroma and serum containing immune cells flowing in functional tumor vessels. Our unique models better capture the clinical physio-mechanical tissue properties, signaling pathways, hemodynamics and drug responsiveness. Using our 3D-printed models, we aim to develop two new fronts for identifying novel clinically-relevant molecular drivers (Aim 2) followed by the development of precision nanomedicines (Aim 3). We will exploit our vast experience in anticancer nanomedicines to design three therapeutic approaches that target various cellular compartments involved in brain metastases: 1) Prevention of brain metastatic colonization using targeted nano-vaccines, which elicit antitumor immune response; 2) Intervention of tumor-brain stroma cells crosstalk when brain micrometastases establish; 3) Regression of macrometastatic disease by selectively targeting tumor cells. These approaches will materialize using our libraries of polymeric nanocarriers that selectively accumulate in tumors.
This project will result in a paradigm shift by generating new preclinical cancer models that will bridge the translational gap in cancer therapeutics. The insights and tumor-stroma-targeted nanomedicines developed here will pave the way for prediction of patient outcome, revolutionizing our perception of tumor modelling and consequently the way we prevent and treat cancer.
Max ERC Funding
2 353 125 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym ABC
Project Targeting Multidrug Resistant Cancer
Researcher (PI) Gergely Szakacs
Host Institution (HI) MAGYAR TUDOMANYOS AKADEMIA TERMESZETTUDOMANYI KUTATOKOZPONT
Call Details Starting Grant (StG), LS7, ERC-2010-StG_20091118
Summary Despite considerable advances in drug discovery, resistance to anticancer chemotherapy confounds the effective treatment of patients. Cancer cells can acquire broad cross-resistance to mechanistically and structurally unrelated drugs. P-glycoprotein (Pgp) actively extrudes many types of drugs from cancer cells, thereby conferring resistance to those agents. The central tenet of my work is that Pgp, a universally accepted biomarker of drug resistance, should in addition be considered as a molecular target of multidrug-resistant (MDR) cancer cells. Successful targeting of MDR cells would reduce the tumor burden and would also enable the elimination of ABC transporter-overexpressing cancer stem cells that are responsible for the replenishment of tumors. The proposed project is based on the following observations:
- First, by using a pharmacogenomic approach, I have revealed the hidden vulnerability of MDRcells (Szakács et al. 2004, Cancer Cell 6, 129-37);
- Second, I have identified a series of MDR-selective compounds with increased toxicity toPgp-expressing cells
(Turk et al.,Cancer Res, 2009. 69(21));
- Third, I have shown that MDR-selective compounds can be used to prevent theemergence of MDR (Ludwig, Szakács et al. 2006, Cancer Res 66, 4808-15);
- Fourth, we have generated initial pharmacophore models for cytotoxicity and MDR-selectivity (Hall et al. 2009, J Med Chem 52, 3191-3204).
I propose a comprehensive series of studies that will address thefollowing critical questions:
- First, what is the scope of MDR-selective compounds?
- Second, what is their mechanism of action?
- Third, what is the optimal therapeutic modality?
Extensive biological, pharmacological and bioinformatic analyses will be utilized to address four major specific aims. These aims address basic questions concerning the physiology of MDR ABC transporters in determining the mechanism of action of MDR-selective compounds, setting the stage for a fresh therapeutic approach that may eventually translate into improved patient care.
Summary
Despite considerable advances in drug discovery, resistance to anticancer chemotherapy confounds the effective treatment of patients. Cancer cells can acquire broad cross-resistance to mechanistically and structurally unrelated drugs. P-glycoprotein (Pgp) actively extrudes many types of drugs from cancer cells, thereby conferring resistance to those agents. The central tenet of my work is that Pgp, a universally accepted biomarker of drug resistance, should in addition be considered as a molecular target of multidrug-resistant (MDR) cancer cells. Successful targeting of MDR cells would reduce the tumor burden and would also enable the elimination of ABC transporter-overexpressing cancer stem cells that are responsible for the replenishment of tumors. The proposed project is based on the following observations:
- First, by using a pharmacogenomic approach, I have revealed the hidden vulnerability of MDRcells (Szakács et al. 2004, Cancer Cell 6, 129-37);
- Second, I have identified a series of MDR-selective compounds with increased toxicity toPgp-expressing cells
(Turk et al.,Cancer Res, 2009. 69(21));
- Third, I have shown that MDR-selective compounds can be used to prevent theemergence of MDR (Ludwig, Szakács et al. 2006, Cancer Res 66, 4808-15);
- Fourth, we have generated initial pharmacophore models for cytotoxicity and MDR-selectivity (Hall et al. 2009, J Med Chem 52, 3191-3204).
I propose a comprehensive series of studies that will address thefollowing critical questions:
- First, what is the scope of MDR-selective compounds?
- Second, what is their mechanism of action?
- Third, what is the optimal therapeutic modality?
Extensive biological, pharmacological and bioinformatic analyses will be utilized to address four major specific aims. These aims address basic questions concerning the physiology of MDR ABC transporters in determining the mechanism of action of MDR-selective compounds, setting the stage for a fresh therapeutic approach that may eventually translate into improved patient care.
Max ERC Funding
1 499 640 €
Duration
Start date: 2012-01-01, End date: 2016-12-31
Project acronym ABDESIGN
Project Computational design of novel protein function in antibodies
Researcher (PI) Sarel-Jacob Fleishman
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS1, ERC-2013-StG
Summary We propose to elucidate the structural design principles of naturally occurring antibody complementarity-determining regions (CDRs) and to computationally design novel antibody functions. Antibodies represent the most versatile known system for molecular recognition. Research has yielded many insights into antibody design principles and promising biotechnological and pharmaceutical applications. Still, our understanding of how CDRs encode specific loop conformations lags far behind our understanding of structure-function relationships in non-immunological scaffolds. Thus, design of antibodies from first principles has not been demonstrated. We propose a computational-experimental strategy to address this challenge. We will: (a) characterize the design principles and sequence elements that rigidify antibody CDRs. Natural antibody loops will be subjected to computational modeling, crystallography, and a combined in vitro evolution and deep-sequencing approach to isolate sequence features that rigidify loop backbones; (b) develop a novel computational-design strategy, which uses the >1000 solved structures of antibodies deposited in structure databases to realistically model CDRs and design them to recognize proteins that have not been co-crystallized with antibodies. For example, we will design novel antibodies targeting insulin, for which clinically useful diagnostics are needed. By accessing much larger sequence/structure spaces than are available to natural immune-system repertoires and experimental methods, computational antibody design could produce higher-specificity and higher-affinity binders, even to challenging targets; and (c) develop new strategies to program conformational change in CDRs, generating, e.g., the first allosteric antibodies. These will allow targeting, in principle, of any molecule, potentially revolutionizing how antibodies are generated for research and medicine, providing new insights on the design principles of protein functional sites.
Summary
We propose to elucidate the structural design principles of naturally occurring antibody complementarity-determining regions (CDRs) and to computationally design novel antibody functions. Antibodies represent the most versatile known system for molecular recognition. Research has yielded many insights into antibody design principles and promising biotechnological and pharmaceutical applications. Still, our understanding of how CDRs encode specific loop conformations lags far behind our understanding of structure-function relationships in non-immunological scaffolds. Thus, design of antibodies from first principles has not been demonstrated. We propose a computational-experimental strategy to address this challenge. We will: (a) characterize the design principles and sequence elements that rigidify antibody CDRs. Natural antibody loops will be subjected to computational modeling, crystallography, and a combined in vitro evolution and deep-sequencing approach to isolate sequence features that rigidify loop backbones; (b) develop a novel computational-design strategy, which uses the >1000 solved structures of antibodies deposited in structure databases to realistically model CDRs and design them to recognize proteins that have not been co-crystallized with antibodies. For example, we will design novel antibodies targeting insulin, for which clinically useful diagnostics are needed. By accessing much larger sequence/structure spaces than are available to natural immune-system repertoires and experimental methods, computational antibody design could produce higher-specificity and higher-affinity binders, even to challenging targets; and (c) develop new strategies to program conformational change in CDRs, generating, e.g., the first allosteric antibodies. These will allow targeting, in principle, of any molecule, potentially revolutionizing how antibodies are generated for research and medicine, providing new insights on the design principles of protein functional sites.
Max ERC Funding
1 499 930 €
Duration
Start date: 2013-09-01, End date: 2018-08-31
Project acronym BARCODE DIAGNOSTICS
Project Next-Generation Personalized Diagnostic Nanotechnologies for Predicting Response to Cancer Medicine
Researcher (PI) Avraham Dror Schroeder
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS7, ERC-2015-STG
Summary Cancer is the leading cause of death in the Western world and the second cause of death worldwide. Despite advances in medical research, 30% of cancer patients are prescribed a medication the tumor does not respond to, or, alternatively, drugs that induce adverse side effects patients' cannot tolerate.
Nanotechnologies are becoming impactful therapeutic tools, granting tissue-targeting and cellular precision that cannot be attained using systems of larger scale.
In this proposal, I plan to expand far beyond the state-of-the-art and develop a conceptually new approach in which diagnostic nanoparticles are designed to retrieve drug-sensitivity information from malignant tissue inside the body. The ultimate goal of this program is to be able to predict, ahead of time, which treatment will be best for each cancer patient – an emerging field called personalized medicine. This interdisciplinary research program will expand our understandings and capabilities in nanotechnology, cancer biology and medicine.
To achieve this goal, I will engineer novel nanotechnologies that autonomously maneuver, target and diagnose the various cells that compose the tumor microenvironment and its disseminated metastasis. Each nanometric system will contain a miniscule amount of a biologically-active agent, and will serve as a nano lab for testing the activity of the agents inside the tumor cells.
To distinguish between system to system, and to grant single-cell sensitivity in vivo, nanoparticles will be barcoded with unique DNA fragments.
We will enable nanoparticle' deep tissue penetration into primary tumors and metastatic microenvironments using enzyme-loaded particles, and study how different agents, including small-molecule drugs, proteins and RNA, interact with the malignant and stromal cells that compose the cancerous microenvironments. Finally, we will demonstrate the ability of barcoded nanoparticles to predict adverse, life-threatening, side effects, in a personalized manner.
Summary
Cancer is the leading cause of death in the Western world and the second cause of death worldwide. Despite advances in medical research, 30% of cancer patients are prescribed a medication the tumor does not respond to, or, alternatively, drugs that induce adverse side effects patients' cannot tolerate.
Nanotechnologies are becoming impactful therapeutic tools, granting tissue-targeting and cellular precision that cannot be attained using systems of larger scale.
In this proposal, I plan to expand far beyond the state-of-the-art and develop a conceptually new approach in which diagnostic nanoparticles are designed to retrieve drug-sensitivity information from malignant tissue inside the body. The ultimate goal of this program is to be able to predict, ahead of time, which treatment will be best for each cancer patient – an emerging field called personalized medicine. This interdisciplinary research program will expand our understandings and capabilities in nanotechnology, cancer biology and medicine.
To achieve this goal, I will engineer novel nanotechnologies that autonomously maneuver, target and diagnose the various cells that compose the tumor microenvironment and its disseminated metastasis. Each nanometric system will contain a miniscule amount of a biologically-active agent, and will serve as a nano lab for testing the activity of the agents inside the tumor cells.
To distinguish between system to system, and to grant single-cell sensitivity in vivo, nanoparticles will be barcoded with unique DNA fragments.
We will enable nanoparticle' deep tissue penetration into primary tumors and metastatic microenvironments using enzyme-loaded particles, and study how different agents, including small-molecule drugs, proteins and RNA, interact with the malignant and stromal cells that compose the cancerous microenvironments. Finally, we will demonstrate the ability of barcoded nanoparticles to predict adverse, life-threatening, side effects, in a personalized manner.
Max ERC Funding
1 499 250 €
Duration
Start date: 2016-04-01, End date: 2021-03-31
Project acronym BisProt
Project Developing Multispecific Biological Agents that Target Tumor Neovasculature for Cancer Imaging and Therapy
Researcher (PI) Niv Papo
Host Institution (HI) BEN-GURION UNIVERSITY OF THE NEGEV
Call Details Starting Grant (StG), LS7, ERC-2013-StG
Summary The dysregulation of signaling pathways that mediate cell proliferation, survival and migration is an underlying cause of many cancers. In particular, dysregulation and over-expression of avb3 integrin, membrane-type-1 matrix metalloproteinase (MT1-MMP; also known as matrix metalloproteinase-14, MMP14) and vascular endothelial growth factor receptor-2 (VEGFR2) correlate with poor prognosis in many human tumors, making these proteins attractive targets for therapeutic intervention. Numerous papers have demonstrated the cross-talk between biological processes mediated by αvβ3 integrins, MT1-MMP, VEGFR2, and their ligands, particularly pathways responsible for angiogenesis. Dual-specific proteins that can target and inhibit the activity of the above multiple receptors therefore have superior potential to single-targeted agents due to differential expression of these disease markers in different patients and the ability of this expression to change over time. Most currently available bispecific protein therapeutics comprise antibodies (Abs) or antibody fragments. The new approach proposed here entails rational and combinatorial methods for engineering multispecificity into small peptides and natural protein ligands to function as non-immunoglobulin alternatives to antibodies. In this innovative approach to creating dual-specific proteins, an additional functionality is introduced into a small peptide or into a natural protein ligand to complement its existing biological properties. We predict that this approach will form a major part of a highly effective strategy for creating ligand-based multispecific receptor inhibitors and molecular tools for protein recognition. We envision that protein variants generated from these efforts will promote the next generation of therapeutics including, but not limited to, molecular imaging agents, targeted drug delivery agents, and selective tissue targeting probes.
Summary
The dysregulation of signaling pathways that mediate cell proliferation, survival and migration is an underlying cause of many cancers. In particular, dysregulation and over-expression of avb3 integrin, membrane-type-1 matrix metalloproteinase (MT1-MMP; also known as matrix metalloproteinase-14, MMP14) and vascular endothelial growth factor receptor-2 (VEGFR2) correlate with poor prognosis in many human tumors, making these proteins attractive targets for therapeutic intervention. Numerous papers have demonstrated the cross-talk between biological processes mediated by αvβ3 integrins, MT1-MMP, VEGFR2, and their ligands, particularly pathways responsible for angiogenesis. Dual-specific proteins that can target and inhibit the activity of the above multiple receptors therefore have superior potential to single-targeted agents due to differential expression of these disease markers in different patients and the ability of this expression to change over time. Most currently available bispecific protein therapeutics comprise antibodies (Abs) or antibody fragments. The new approach proposed here entails rational and combinatorial methods for engineering multispecificity into small peptides and natural protein ligands to function as non-immunoglobulin alternatives to antibodies. In this innovative approach to creating dual-specific proteins, an additional functionality is introduced into a small peptide or into a natural protein ligand to complement its existing biological properties. We predict that this approach will form a major part of a highly effective strategy for creating ligand-based multispecific receptor inhibitors and molecular tools for protein recognition. We envision that protein variants generated from these efforts will promote the next generation of therapeutics including, but not limited to, molecular imaging agents, targeted drug delivery agents, and selective tissue targeting probes.
Max ERC Funding
1 625 000 €
Duration
Start date: 2014-01-01, End date: 2018-12-31
Project acronym BRuSH
Project Oral bacteria as determinants for respiratory health
Researcher (PI) Randi BERTELSEN
Host Institution (HI) UNIVERSITETET I BERGEN
Call Details Starting Grant (StG), LS7, ERC-2018-STG
Summary The oral cavity is the gateway to the lower respiratory tract, and oral bacteria are likely to play a role in lung health. This may be the case for pathogens as well as commensal bacteria and the balance between species. The oral bacterial community of patients with periodontitis is dominated by gram-negative bacteria and a higher lipopolysaccharide (LPS) activity than in healthy microbiota. Furthermore, bacteria with especially potent pro-inflammatory LPS have been shown to be more common in the lungs of asthmatic than in healthy individuals. The working hypothesis of BRuSH is that microbiome communities dominated by LPS-producing bacteria which induce a particularly strong pro-inflammatory immune response in the host, will have a negative effect on respiratory health. I will test this hypothesis in two longitudinally designed population-based lung health studies. I aim to identify whether specific bacterial composition and types of LPS producing bacteria in oral and dust samples predict lung function and respiratory health over time; and if the different types of LPS-producing bacteria affect LPS in saliva saliva and dust. BRuSH will apply functional genome annotation that can assign biological significance to raw bacterial DNA sequences. With this bioinformatics tool I will cluster microbiome data into various LPS-producers: bacteria with LPS with strong inflammatory effects and others with weak- or antagonistic effects. The epidemiological studies will be supported by mice-models of asthma and cell assays of human bronchial epithelial cells, by exposing mice and bronchial cells to chemically synthesized Lipid A (the component that drive the LPS-induced immune responses) of various potency. The goal of BRuSH is to prove a causal relationship between oral microbiome and lung health, and gain knowledge that will enable us to make oral health a feasible target for intervention programs aimed at optimizing lung health and preventing respiratory disease.
Summary
The oral cavity is the gateway to the lower respiratory tract, and oral bacteria are likely to play a role in lung health. This may be the case for pathogens as well as commensal bacteria and the balance between species. The oral bacterial community of patients with periodontitis is dominated by gram-negative bacteria and a higher lipopolysaccharide (LPS) activity than in healthy microbiota. Furthermore, bacteria with especially potent pro-inflammatory LPS have been shown to be more common in the lungs of asthmatic than in healthy individuals. The working hypothesis of BRuSH is that microbiome communities dominated by LPS-producing bacteria which induce a particularly strong pro-inflammatory immune response in the host, will have a negative effect on respiratory health. I will test this hypothesis in two longitudinally designed population-based lung health studies. I aim to identify whether specific bacterial composition and types of LPS producing bacteria in oral and dust samples predict lung function and respiratory health over time; and if the different types of LPS-producing bacteria affect LPS in saliva saliva and dust. BRuSH will apply functional genome annotation that can assign biological significance to raw bacterial DNA sequences. With this bioinformatics tool I will cluster microbiome data into various LPS-producers: bacteria with LPS with strong inflammatory effects and others with weak- or antagonistic effects. The epidemiological studies will be supported by mice-models of asthma and cell assays of human bronchial epithelial cells, by exposing mice and bronchial cells to chemically synthesized Lipid A (the component that drive the LPS-induced immune responses) of various potency. The goal of BRuSH is to prove a causal relationship between oral microbiome and lung health, and gain knowledge that will enable us to make oral health a feasible target for intervention programs aimed at optimizing lung health and preventing respiratory disease.
Max ERC Funding
1 499 938 €
Duration
Start date: 2019-01-01, End date: 2023-12-31
Project acronym CELLNAIVETY
Project Deciphering the Molecular Foundations and Functional Competence of Alternative Human Naïve Pluripotent Stem Cells
Researcher (PI) Yaqub HANNA
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS7, ERC-2016-COG
Summary An important goal of stem cell therapy is to create “customized” cells that are genetically identical to the patient, which upon transplantation can restore damaged tissues. Such cells can be obtained by in vitro direct reprogramming of somatic cells into embryonic stem (ES)-like cells, termed induced pluripotent stem cells (iPSC). This approach also opens possibilities for modelling human diseases in vitro. However, major hurdles remain that restrain fulfilling conventional human iPSC/ESC potential, as they reside in an advanced primed pluripotent state. Such hurdles include limited differentiation capacity and functional variability. Further, in vitro iPSC based research platforms are simplistic and iPSC based “humanized” chimeric mouse models may be of great benefit.
The recent isolation of distinct and new “mouse-like” naive pluripotent states in humans that correspond to earlier embryonic developmental state(s), constitutes a paradigm shift and may alleviate limitations of conventional primed iPSCs/ESCs. Thus, our proposal aims at dissecting the human naïve pluripotent state(s) and to unveil pathways that facilitate their unique identity and flexible programming.
Specific goals: 1) Transcriptional and Epigenetic Design Principles of Human Naïve Pluripotency 2) Signalling Principles Governing Human Naïve Pluripotency Maintenance and Differentiation 3) Defining Functional Competence and Safety of Human Naïve Pluripotent Stem Cells in vitro 4) Novel human naïve iPSC based cross-species chimeric mice for studying human differentiation and disease modelling in vivo. These aims will be conducted by utilizing engineered human iPSC/ESC models, CRISPR/Cas9 genome-wide screening, advanced microscopy and ex-vivo whole embryo culture methods. Our goals will synergistically lead to the design of strategies that will accelerate the safe medical application of human naive pluripotent stem cells and their use in disease specific modelling and applied stem cell research.
Summary
An important goal of stem cell therapy is to create “customized” cells that are genetically identical to the patient, which upon transplantation can restore damaged tissues. Such cells can be obtained by in vitro direct reprogramming of somatic cells into embryonic stem (ES)-like cells, termed induced pluripotent stem cells (iPSC). This approach also opens possibilities for modelling human diseases in vitro. However, major hurdles remain that restrain fulfilling conventional human iPSC/ESC potential, as they reside in an advanced primed pluripotent state. Such hurdles include limited differentiation capacity and functional variability. Further, in vitro iPSC based research platforms are simplistic and iPSC based “humanized” chimeric mouse models may be of great benefit.
The recent isolation of distinct and new “mouse-like” naive pluripotent states in humans that correspond to earlier embryonic developmental state(s), constitutes a paradigm shift and may alleviate limitations of conventional primed iPSCs/ESCs. Thus, our proposal aims at dissecting the human naïve pluripotent state(s) and to unveil pathways that facilitate their unique identity and flexible programming.
Specific goals: 1) Transcriptional and Epigenetic Design Principles of Human Naïve Pluripotency 2) Signalling Principles Governing Human Naïve Pluripotency Maintenance and Differentiation 3) Defining Functional Competence and Safety of Human Naïve Pluripotent Stem Cells in vitro 4) Novel human naïve iPSC based cross-species chimeric mice for studying human differentiation and disease modelling in vivo. These aims will be conducted by utilizing engineered human iPSC/ESC models, CRISPR/Cas9 genome-wide screening, advanced microscopy and ex-vivo whole embryo culture methods. Our goals will synergistically lead to the design of strategies that will accelerate the safe medical application of human naive pluripotent stem cells and their use in disease specific modelling and applied stem cell research.
Max ERC Funding
2 000 000 €
Duration
Start date: 2017-11-01, End date: 2022-10-31
Project acronym CIRCOMMUNICATION
Project Deciphering molecular pathways of circadian clock communication
Researcher (PI) gad ASHER
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS1, ERC-2017-COG
Summary The overarching objective of this interdisciplinary project is to elucidate mechanisms through which billions of individual clocks in the body communicate with each other and tick in harmony. The mammalian circadian timing system consists of a master clock in the brain and subsidiary oscillators in almost every cell of the body. Since these clocks anticipate environmental changes and function together to orchestrate daily physiology and behavior their temporal synchronization is critical.
Our recent finding that oxygen serves as a resetting cue for circadian clocks points towards the unprecedented involvement of blood gases as time signals. We will apply cutting edge continuous physiological measurements in freely moving animals, alongside biochemical/molecular biology approaches and advanced cell culture setup to determine the molecular role of oxygen, carbon dioxide and pH in circadian clock communication and function.
The intricate nature of the mammalian circadian system demands the presence of communication mechanisms between clocks throughout the body at multiple levels. While previous studies primarily addressed the role of the master clock in resetting peripheral clocks, our knowledge regarding the communication among clocks between and within peripheral organs is rudimentary. We will reconstruct the mammalian circadian system from the bottom up, sequentially restoring clocks in peripheral tissues of a non-rhythmic animal to (i) obtain a system-view of the peripheral circadian communication network; and (ii) study novel tissue-derived circadian communication mechanisms.
This integrative proposal addresses fundamental aspects of circadian biology. It is expected to unravel the circadian communication network and shed light on how billions of clocks in the body function in unison. Its impact extends beyond circadian rhythms and bears great potential for research on communication between cells/tissues in various fields of biology.
Summary
The overarching objective of this interdisciplinary project is to elucidate mechanisms through which billions of individual clocks in the body communicate with each other and tick in harmony. The mammalian circadian timing system consists of a master clock in the brain and subsidiary oscillators in almost every cell of the body. Since these clocks anticipate environmental changes and function together to orchestrate daily physiology and behavior their temporal synchronization is critical.
Our recent finding that oxygen serves as a resetting cue for circadian clocks points towards the unprecedented involvement of blood gases as time signals. We will apply cutting edge continuous physiological measurements in freely moving animals, alongside biochemical/molecular biology approaches and advanced cell culture setup to determine the molecular role of oxygen, carbon dioxide and pH in circadian clock communication and function.
The intricate nature of the mammalian circadian system demands the presence of communication mechanisms between clocks throughout the body at multiple levels. While previous studies primarily addressed the role of the master clock in resetting peripheral clocks, our knowledge regarding the communication among clocks between and within peripheral organs is rudimentary. We will reconstruct the mammalian circadian system from the bottom up, sequentially restoring clocks in peripheral tissues of a non-rhythmic animal to (i) obtain a system-view of the peripheral circadian communication network; and (ii) study novel tissue-derived circadian communication mechanisms.
This integrative proposal addresses fundamental aspects of circadian biology. It is expected to unravel the circadian communication network and shed light on how billions of clocks in the body function in unison. Its impact extends beyond circadian rhythms and bears great potential for research on communication between cells/tissues in various fields of biology.
Max ERC Funding
1 999 945 €
Duration
Start date: 2018-03-01, End date: 2023-02-28
Project acronym CRISS
Project CRISPR Gene Correction for Severe Combined Immunodeficiency Caused by Mutations in Recombination-activating gene 1 and 2 (RAG1 and RAG2)
Researcher (PI) Ayal Hendel
Host Institution (HI) BAR ILAN UNIVERSITY
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary The severe combined immunodeficiencies (SCIDs) are a set of life threatening genetic diseases in which patients are born with mutations in single genes and are unable to develop functional immune systems. While allogeneic bone marrow transplantation can be curative for these diseases, there remain significant limitations to this approach. Gene therapy using viral vectors containing a corrective transgene is being developed for some of these disorders, most successfully for ADA-SCID. However, for other SCID disorders, such as those caused by genetic mutations in RAG1 and RAG2, the transgene needs to be expressed in a precise, developmental and lineage specific manner to achieve functional gene correction and to avoid the risks of cellular transformation. In contrast to using viral vectors to deliver transgenes in an uncontrolled fashion, we are working towards using genome editing by homologous recombination (HR) to correct the disease causing mutation by precisely modifying the genome. We have shown that by using clustered, regularly interspaced, short palindromic repeats (CRISPR) and the CRISPR-associated protein 9 (Cas9) system we can stimulate genome editing by HR at frequencies that should be therapeutically beneficial (>10%) in hematopoietic stem and progenitor cells (HSPCs). The overall focus of the proposal is to translate our basic science studies to use in RAG-SCID patient-derived HSPCs in methodical, careful and pre-clinically relevant fashion. The fundamental approach is to develop a highly active functional genome editing system using CRISPR-Cas9 for RAG-SCIDs and complete pre-clinical efficacy and safety studies to show the approach has a clear path towards future clinical trials. Our goal with this proposal is to develop the next wave of curative therapies for SCIDs and other hematopoietic disorders using genome editing.
Summary
The severe combined immunodeficiencies (SCIDs) are a set of life threatening genetic diseases in which patients are born with mutations in single genes and are unable to develop functional immune systems. While allogeneic bone marrow transplantation can be curative for these diseases, there remain significant limitations to this approach. Gene therapy using viral vectors containing a corrective transgene is being developed for some of these disorders, most successfully for ADA-SCID. However, for other SCID disorders, such as those caused by genetic mutations in RAG1 and RAG2, the transgene needs to be expressed in a precise, developmental and lineage specific manner to achieve functional gene correction and to avoid the risks of cellular transformation. In contrast to using viral vectors to deliver transgenes in an uncontrolled fashion, we are working towards using genome editing by homologous recombination (HR) to correct the disease causing mutation by precisely modifying the genome. We have shown that by using clustered, regularly interspaced, short palindromic repeats (CRISPR) and the CRISPR-associated protein 9 (Cas9) system we can stimulate genome editing by HR at frequencies that should be therapeutically beneficial (>10%) in hematopoietic stem and progenitor cells (HSPCs). The overall focus of the proposal is to translate our basic science studies to use in RAG-SCID patient-derived HSPCs in methodical, careful and pre-clinically relevant fashion. The fundamental approach is to develop a highly active functional genome editing system using CRISPR-Cas9 for RAG-SCIDs and complete pre-clinical efficacy and safety studies to show the approach has a clear path towards future clinical trials. Our goal with this proposal is to develop the next wave of curative therapies for SCIDs and other hematopoietic disorders using genome editing.
Max ERC Funding
1 372 839 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym CT-PROBES
Project Protease Activated X-Ray Contrast Agents for Molecular Imaging of Vulnerable Atherosclerotic Plaques and Cancer Development using Spectral CT
Researcher (PI) Galia Blum
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS7, ERC-2013-StG
Summary The major causes of death in the Western world are cardiovascular diseases and cancer. More accurate detection of these diseases will improve clinical outcomes. Thus, we will develop unique X-ray contrast reagents for use in spectral computerized tomography (CT) that bind active proteases to reveal the exact location and stage of cancer and atherosclerosis.
Activity-based probes (ABPs) are small molecules that covalently bind to active proteases. Based on our success in developing optical ABPs for non-invasive optical detection of cancer and atherosclerosis, we will focus on two novel types of reagents: (1) ABPs conjugated to the various contrast elements that can be visualized by x-rays. (2) “smart probes” conjugated to different contrast reagents on each side of the molecule to overcome clearance limitations. Protease found in diseased tissue will selectively bind and remove a part of the molecule, changing the physical properties of the bound probe. Thus, different signals from bound and unbound probes could be detected by photon counting spectral CT scanners.
Our initial target, cysteine cathepsin proteases, are overexpressed and activated in cancer and arthrosclerosis. The level of active cathepsins correlates with progression of both diseases, thereby serving as a promising biomarker for these pathologies. The “smart probes” are an innovative type of spectral CT agent that will enable high-resolution rapid imaging in humans before probe clearance.
Our probes increase imaging sensitivity since the contrast element remains at the desired site. Moreover, the levels of active cathepsins will reveal critical information regarding disease progression, yielding more accurate diagnoses and improved personalized treatment. For example, these reagents can distinguish between a vulnerable and stable atherosclerotic plaque. Thus, our novel probes will directly reduce cancer and cardiovascular disease mortality by enabling earlier and more accurate disease detection.
Summary
The major causes of death in the Western world are cardiovascular diseases and cancer. More accurate detection of these diseases will improve clinical outcomes. Thus, we will develop unique X-ray contrast reagents for use in spectral computerized tomography (CT) that bind active proteases to reveal the exact location and stage of cancer and atherosclerosis.
Activity-based probes (ABPs) are small molecules that covalently bind to active proteases. Based on our success in developing optical ABPs for non-invasive optical detection of cancer and atherosclerosis, we will focus on two novel types of reagents: (1) ABPs conjugated to the various contrast elements that can be visualized by x-rays. (2) “smart probes” conjugated to different contrast reagents on each side of the molecule to overcome clearance limitations. Protease found in diseased tissue will selectively bind and remove a part of the molecule, changing the physical properties of the bound probe. Thus, different signals from bound and unbound probes could be detected by photon counting spectral CT scanners.
Our initial target, cysteine cathepsin proteases, are overexpressed and activated in cancer and arthrosclerosis. The level of active cathepsins correlates with progression of both diseases, thereby serving as a promising biomarker for these pathologies. The “smart probes” are an innovative type of spectral CT agent that will enable high-resolution rapid imaging in humans before probe clearance.
Our probes increase imaging sensitivity since the contrast element remains at the desired site. Moreover, the levels of active cathepsins will reveal critical information regarding disease progression, yielding more accurate diagnoses and improved personalized treatment. For example, these reagents can distinguish between a vulnerable and stable atherosclerotic plaque. Thus, our novel probes will directly reduce cancer and cardiovascular disease mortality by enabling earlier and more accurate disease detection.
Max ERC Funding
1 499 780 €
Duration
Start date: 2013-12-01, End date: 2018-11-30
Project acronym CuHypMECH
Project New Nuclear Medicine Imaging Radiotracer 64Cu(II) for diagnosing Hypoxia Conditions Based on the Cellular Copper Cycle
Researcher (PI) Sharon RUTHSTEIN
Host Institution (HI) BAR ILAN UNIVERSITY
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary Imaging of hypoxia is important in many disease states in oncology, cardiology, and neurology. Hypoxia is a common condition encountered within the tumour microenvironment that drives proliferation, angiogenesis, and resistance to therapy. Despite on-going efforts to identify hypoxia, until now there is no clinically approved imaging biomarker, due to both low tumour uptake, and a low signal to background (S/B) ratio that affects the imaging quality. Nuclear Medicine is using labelled radio-isotopes for PET/CT and SPECT imaging. These radio-tracers diagnose the metabolic processes in the body. Among these tracers, 18F-FDG is the most routinely used as a marker of glucose metabolism. However, not all tumours consume glucose, and glucose consumption is not specific only for malignant tumours, which limits its application. Copper is a nutritional metal, recently examined as a radiotracer for hypoxia, owing to its to the oxidising environment. Clinical and in-vivo studies on various 64Cu(II)-PET radiotracers resulted in controversial reports on the specificity of the current tracers for hypoxia imaging due to non-selective bio-distribution & low S/B ratio. This multidisciplinary proposal focuses on the discovery of comprehensive signal pathways of the cellular copper cycle using advanced biophysical methods and a proprietary design of 64Cu(II) radiotracer. This radiotracer will be incorporated in the cellular copper cycle, and will enable to selectively target the oxidising environment in tumours. The design of the new radiotracer is based on systematic structural & functional mapping of the copper binding sites to the various copper proteins and the visualisation of the transfer mechanism. This new copper tracer should increase the selectivity of tumour uptake, stability, and improve bio-distribution. This project assimilates cold and hot chemistry and biology, while emphasising the clinical unmet need in metal based radiotracer that form stable complexes.
Summary
Imaging of hypoxia is important in many disease states in oncology, cardiology, and neurology. Hypoxia is a common condition encountered within the tumour microenvironment that drives proliferation, angiogenesis, and resistance to therapy. Despite on-going efforts to identify hypoxia, until now there is no clinically approved imaging biomarker, due to both low tumour uptake, and a low signal to background (S/B) ratio that affects the imaging quality. Nuclear Medicine is using labelled radio-isotopes for PET/CT and SPECT imaging. These radio-tracers diagnose the metabolic processes in the body. Among these tracers, 18F-FDG is the most routinely used as a marker of glucose metabolism. However, not all tumours consume glucose, and glucose consumption is not specific only for malignant tumours, which limits its application. Copper is a nutritional metal, recently examined as a radiotracer for hypoxia, owing to its to the oxidising environment. Clinical and in-vivo studies on various 64Cu(II)-PET radiotracers resulted in controversial reports on the specificity of the current tracers for hypoxia imaging due to non-selective bio-distribution & low S/B ratio. This multidisciplinary proposal focuses on the discovery of comprehensive signal pathways of the cellular copper cycle using advanced biophysical methods and a proprietary design of 64Cu(II) radiotracer. This radiotracer will be incorporated in the cellular copper cycle, and will enable to selectively target the oxidising environment in tumours. The design of the new radiotracer is based on systematic structural & functional mapping of the copper binding sites to the various copper proteins and the visualisation of the transfer mechanism. This new copper tracer should increase the selectivity of tumour uptake, stability, and improve bio-distribution. This project assimilates cold and hot chemistry and biology, while emphasising the clinical unmet need in metal based radiotracer that form stable complexes.
Max ERC Funding
1 499 345 €
Duration
Start date: 2017-09-01, End date: 2022-08-31
Project acronym DIAG-CANCER
Project Diagnosis, Screening and Monitoring of Cancer Diseases via Exhaled Breath Using an Array of Nanosensors
Researcher (PI) Hossam Haick
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS7, ERC-2010-StG_20091118
Summary Cancer is rapidly becoming the greatest health hazard of our days. The most widespread cancers, are lung cancer (LC), breast cancer (BC), colorectal cancer (CC), and prostate cancer (PC). The impact of the various techniques used for diagnosis, screening and monitoring
these cancers is either uncertain and/or inconvenient for the patients. This proposal aims to create a low-cost, easy-to-use and noninvasive screening method for LC, BC, CC, and PC based on breath testing with a novel nanosensors approach. With this in mind, we propose to:
(a) modify an array of nanosensors based on Au nanoparticles for obtaining highly-sensitive detection levels of breath biomarkers of cancer; and
(b) investigate the use of the developed array in a clinical study.
Towards this end, we will collect suitable breath samples from patients and healthy controls in a clinical trial and test the feasibility of the device to detect LC, BC, CC, and PC, also in the presence of other diseases.
We will then investigate possible ways to identify the stage of the disease, monitor the response to cancer
treatment, and to identify cancer subtypes. Further, we propose that the device can be used for monitoring of cancer patients during and after treatment. The chemical nature of the cancer biomarkers will be identified through spectrometry techniques.
The proposed approach would be used outside specialist settings and could considerably lessen the burden on the health budgets, both through the low cost of the proposed all-inclusive cancer test, and through earlier and, hence, more cost-effective cancer treatment.
Summary
Cancer is rapidly becoming the greatest health hazard of our days. The most widespread cancers, are lung cancer (LC), breast cancer (BC), colorectal cancer (CC), and prostate cancer (PC). The impact of the various techniques used for diagnosis, screening and monitoring
these cancers is either uncertain and/or inconvenient for the patients. This proposal aims to create a low-cost, easy-to-use and noninvasive screening method for LC, BC, CC, and PC based on breath testing with a novel nanosensors approach. With this in mind, we propose to:
(a) modify an array of nanosensors based on Au nanoparticles for obtaining highly-sensitive detection levels of breath biomarkers of cancer; and
(b) investigate the use of the developed array in a clinical study.
Towards this end, we will collect suitable breath samples from patients and healthy controls in a clinical trial and test the feasibility of the device to detect LC, BC, CC, and PC, also in the presence of other diseases.
We will then investigate possible ways to identify the stage of the disease, monitor the response to cancer
treatment, and to identify cancer subtypes. Further, we propose that the device can be used for monitoring of cancer patients during and after treatment. The chemical nature of the cancer biomarkers will be identified through spectrometry techniques.
The proposed approach would be used outside specialist settings and could considerably lessen the burden on the health budgets, both through the low cost of the proposed all-inclusive cancer test, and through earlier and, hence, more cost-effective cancer treatment.
Max ERC Funding
1 200 000 €
Duration
Start date: 2011-01-01, End date: 2014-12-31
Project acronym DrugsInPregnancy
Project Effects of Medication Use in Pregnancy on Infant Neurodevelopment
Researcher (PI) Hedvig Marie Egeland Nordeng
Host Institution (HI) UNIVERSITETET I OSLO
Call Details Starting Grant (StG), LS7, ERC-2014-STG
Summary Currently, thousands of pregnant women in the EU and worldwide are being increasingly prescribed medications for which we do not have sufficient information on fetal safety. I hypothesize that our current understanding of safety pharmacology is oversimplified and that medication prescribed during pregnancy may play an unrecognized role in the development of neurodevelopmental disorders.
In this research proposal we have the unique opportunity to use a large population-based birth cohort including over 100 000 mother-child pairs and biological data to study how medications may act on the offspring. This offers novel and innovative pharmaceutical insight into the safety of medications.
By linking several nationwide registries (National Prescription Data Base, Norwegian Patient Registry, Medical Birth Registry) to a population-based birth cohort (n=108 000) we specifically aim to 1) estimate the effect of prenatal exposure to psychotropics and analgesics on neurodevelopment in young children using a range of methodological approaches to strengthen causal inference.
With these data made available, we will 2) determine whether fetal exposure to specific medications results in epigenetic events (i.e. changes in DNA methylation) in the child, and 3) determine whether such changes increase the risks of neurodevelopmental disorders in childhood.
The recent availability of large scale human data, possibility of register linkages and genome-wide mapping of DNA methylation at affordable costs makes this research proposal now possible. The size and richness of data including over hundred thousand pregnancies and existence of biological material makes this project unique. The final outcome will be fundamentally new knowledge about how medications affect the developing unborn child and will open up new horizons and opportunities for future research in a new field of “pharmaco-epigenetics” and enhance our understanding of origins of neurodevelopmental disorders.
Summary
Currently, thousands of pregnant women in the EU and worldwide are being increasingly prescribed medications for which we do not have sufficient information on fetal safety. I hypothesize that our current understanding of safety pharmacology is oversimplified and that medication prescribed during pregnancy may play an unrecognized role in the development of neurodevelopmental disorders.
In this research proposal we have the unique opportunity to use a large population-based birth cohort including over 100 000 mother-child pairs and biological data to study how medications may act on the offspring. This offers novel and innovative pharmaceutical insight into the safety of medications.
By linking several nationwide registries (National Prescription Data Base, Norwegian Patient Registry, Medical Birth Registry) to a population-based birth cohort (n=108 000) we specifically aim to 1) estimate the effect of prenatal exposure to psychotropics and analgesics on neurodevelopment in young children using a range of methodological approaches to strengthen causal inference.
With these data made available, we will 2) determine whether fetal exposure to specific medications results in epigenetic events (i.e. changes in DNA methylation) in the child, and 3) determine whether such changes increase the risks of neurodevelopmental disorders in childhood.
The recent availability of large scale human data, possibility of register linkages and genome-wide mapping of DNA methylation at affordable costs makes this research proposal now possible. The size and richness of data including over hundred thousand pregnancies and existence of biological material makes this project unique. The final outcome will be fundamentally new knowledge about how medications affect the developing unborn child and will open up new horizons and opportunities for future research in a new field of “pharmaco-epigenetics” and enhance our understanding of origins of neurodevelopmental disorders.
Max ERC Funding
1 499 439 €
Duration
Start date: 2015-09-01, End date: 2020-08-31
Project acronym ENGVASC
Project Engineering Vascularized Tissues
Researcher (PI) Shulamit Levenberg
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS7, ERC-2011-StG_20101109
Summary Vascularization, the process in which new blood vessels assemble, is fundamental to tissue vitality. Vessel network assembly within 3D tissues can be induced in-vitro by means of multicellular culturing of endothelial cells (EC), fibroblasts and cells specific to the tissue of interest. This approach supports formation of endothelial vessels and promotes EC and tissue-specific cell interactions. Such EC-dependent tube-like openings may also form the basis for improved media penetration to the inner regions of thick 3D constructs, allowing for enhanced construct survival and for effective engineering of large complex tissues in the lab. Moreover, our own breakthrough results describe the beneficial impact of in vitro prevascularization of engineered muscle tissue on its survival and vascularization upon implantation. These studies have also demonstrated that implanted vascular networks of in vitro engineered constructs, can anastomose with host vasculature and form functional blood vessels in vivo. However, the mechanisms underlying enhanced vascularization of endothelialized engineered constructs and implant-host vessel integration remain unclear. In this proposal, our research objectives are (1) to uncover the mechanisms governing in vitro vessel network formation in engineered 3D tissues and (2) to elucidate the process of graft-host vessel network integration and implant vessel-stimulated promotion of neovascularization in vivo. In addition, the impact of construct prevascularization on implant survival and function will be explored in animal disease models. While there are still many challenges ahead, should we succeed, our research could lay the foundation for significantly enhanced tissue construct vascularization procedures and for their application in regenerative medicine. In addition, it may provide alternative models for studying the vascularization processes in embryogenesis and disease.
Summary
Vascularization, the process in which new blood vessels assemble, is fundamental to tissue vitality. Vessel network assembly within 3D tissues can be induced in-vitro by means of multicellular culturing of endothelial cells (EC), fibroblasts and cells specific to the tissue of interest. This approach supports formation of endothelial vessels and promotes EC and tissue-specific cell interactions. Such EC-dependent tube-like openings may also form the basis for improved media penetration to the inner regions of thick 3D constructs, allowing for enhanced construct survival and for effective engineering of large complex tissues in the lab. Moreover, our own breakthrough results describe the beneficial impact of in vitro prevascularization of engineered muscle tissue on its survival and vascularization upon implantation. These studies have also demonstrated that implanted vascular networks of in vitro engineered constructs, can anastomose with host vasculature and form functional blood vessels in vivo. However, the mechanisms underlying enhanced vascularization of endothelialized engineered constructs and implant-host vessel integration remain unclear. In this proposal, our research objectives are (1) to uncover the mechanisms governing in vitro vessel network formation in engineered 3D tissues and (2) to elucidate the process of graft-host vessel network integration and implant vessel-stimulated promotion of neovascularization in vivo. In addition, the impact of construct prevascularization on implant survival and function will be explored in animal disease models. While there are still many challenges ahead, should we succeed, our research could lay the foundation for significantly enhanced tissue construct vascularization procedures and for their application in regenerative medicine. In addition, it may provide alternative models for studying the vascularization processes in embryogenesis and disease.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym FORCEMAP
Project Intramolecular force mapping of enzymes in action: the role of strain in motor mechanisms
Researcher (PI) András Málnási-Csizmadia
Host Institution (HI) EOTVOS LORAND TUDOMANYEGYETEM
Call Details Starting Grant (StG), LS1, ERC-2007-StG
Summary A fundamental but unexplored problem in biology is whether and how enzymes use mechanical strain during their functioning. It is now evident that the knowledge of atomic structures and chemical interactions is not sufficient to understand the intricate mechanisms underlying enzyme specificity and efficiency. Several lines of evidence suggest that mechanical effects play crucial roles in enzyme activity. Therefore we aim to create detailed force maps that reveal how the intramolecular distribution of mechanical strains changes during the enzyme cycle and how these rearrangements drive the enzyme processes. The applicability of current nanotechniques for the investigation of this problem is limited because they do not allow simultaneous measurement of mechanical and enzymatic parameters. Thus we seek to open new avenues of research by developing site-specific sensors and passive or photoinducible molecular springs to measure force-dependent chemical/structural changes with high spatiotemporal resolution in myosin. Since force perturbations occur very rapidly, we are able to combine experimental studies with quasi-realistic in silico simulations to describe the physical background of enzyme function. We expect that our research will yield fundamental insights into the role of intramolecular strains in enzymes and thus greatly aid the design and control of enzyme processes (specificity, activity, regulation). Our studies may also lead to new paradigms in the understanding of motor systems.
Summary
A fundamental but unexplored problem in biology is whether and how enzymes use mechanical strain during their functioning. It is now evident that the knowledge of atomic structures and chemical interactions is not sufficient to understand the intricate mechanisms underlying enzyme specificity and efficiency. Several lines of evidence suggest that mechanical effects play crucial roles in enzyme activity. Therefore we aim to create detailed force maps that reveal how the intramolecular distribution of mechanical strains changes during the enzyme cycle and how these rearrangements drive the enzyme processes. The applicability of current nanotechniques for the investigation of this problem is limited because they do not allow simultaneous measurement of mechanical and enzymatic parameters. Thus we seek to open new avenues of research by developing site-specific sensors and passive or photoinducible molecular springs to measure force-dependent chemical/structural changes with high spatiotemporal resolution in myosin. Since force perturbations occur very rapidly, we are able to combine experimental studies with quasi-realistic in silico simulations to describe the physical background of enzyme function. We expect that our research will yield fundamental insights into the role of intramolecular strains in enzymes and thus greatly aid the design and control of enzyme processes (specificity, activity, regulation). Our studies may also lead to new paradigms in the understanding of motor systems.
Max ERC Funding
750 000 €
Duration
Start date: 2008-09-01, End date: 2014-08-31
Project acronym GENEXP
Project Gene Expression Explored in Space and Time Using Single Gene and Single Molecule Analysis
Researcher (PI) Yaron Shav-Tal
Host Institution (HI) BAR ILAN UNIVERSITY
Call Details Starting Grant (StG), LS1, ERC-2010-StG_20091118
Summary "Live-cell imaging combined with kinetic analyses has provided new biological insights on the gene expression pathway. However, such studies in mammalian cells typically require use of exogenous over-expressed gene constructs, which often form large tandem gene arrays and usually lack the complete endogenous regulatory sequences. It is therefore imperative to design methodology for analyzing gene expression kinetics of single alleles of endogenous genes. While certain steps have been taken in this direction, there are many experimental obstacles standing in the way of a robust genome-wide system for the in vivo examination of endogenous gene expression within the natural nuclear environment. GENEXP sets out to provide such a system.
It will start with methodology for robust tagging of a multitude of endogenous genes and their transcribed mRNAs in human cells using the ""CD tagging"" approach. Thereby, in vivo mRNA synthesis at the nuclear site of RNA birth will be explored in a unique manner. A high-resolution study of gene expression, in particular mRNA transcription and mRNA export, under endogenous cellular context and using a genome-wide live-cell approach will be performed. GENEXP will specifically focus on the:
i) Transcriptional kinetics of endogenous genes in single cells and cell populations;
ii) Kinetics of mRNA export on the single molecule level;
iii) Examination of the protein composition of endogenous mRNPs;
iv) High throughput scan for drugs that affect gene expression and mRNA export.
Altogether, GENEXP will provide breakthrough capability in kinetically quantifying the gene expression pathway of a large variety of endogenous genes, and the ability to examine the generated molecules on the single-molecule level. This will be done within their normal genomic and biological environment, at the single-allele level."
Summary
"Live-cell imaging combined with kinetic analyses has provided new biological insights on the gene expression pathway. However, such studies in mammalian cells typically require use of exogenous over-expressed gene constructs, which often form large tandem gene arrays and usually lack the complete endogenous regulatory sequences. It is therefore imperative to design methodology for analyzing gene expression kinetics of single alleles of endogenous genes. While certain steps have been taken in this direction, there are many experimental obstacles standing in the way of a robust genome-wide system for the in vivo examination of endogenous gene expression within the natural nuclear environment. GENEXP sets out to provide such a system.
It will start with methodology for robust tagging of a multitude of endogenous genes and their transcribed mRNAs in human cells using the ""CD tagging"" approach. Thereby, in vivo mRNA synthesis at the nuclear site of RNA birth will be explored in a unique manner. A high-resolution study of gene expression, in particular mRNA transcription and mRNA export, under endogenous cellular context and using a genome-wide live-cell approach will be performed. GENEXP will specifically focus on the:
i) Transcriptional kinetics of endogenous genes in single cells and cell populations;
ii) Kinetics of mRNA export on the single molecule level;
iii) Examination of the protein composition of endogenous mRNPs;
iv) High throughput scan for drugs that affect gene expression and mRNA export.
Altogether, GENEXP will provide breakthrough capability in kinetically quantifying the gene expression pathway of a large variety of endogenous genes, and the ability to examine the generated molecules on the single-molecule level. This will be done within their normal genomic and biological environment, at the single-allele level."
Max ERC Funding
1 498 510 €
Duration
Start date: 2010-12-01, End date: 2015-11-30
Project acronym HealthierWomen
Project A woman's reproductive experience: Long-term implications for chronic disease and death
Researcher (PI) Rolv SKJAERVEN
Host Institution (HI) UNIVERSITETET I BERGEN
Call Details Advanced Grant (AdG), LS7, ERC-2018-ADG
Summary Pregnancy complications such as preeclampsia and preterm birth are known to affect infant health, but their influence on mothers’ long-term health is not well understood. Most previous studies are seriously limited by their reliance on information from the first pregnancy. Often they lack the data to study women’s complete reproductive histories. Without a complete reproductive history, the relationship between pregnancy complications and women’s long-term health cannot be reliably studied. The Medical Birth Registry of Norway, covering all births from 1967-, includes information on more than 3 million births and 1.5 million sibships. Linking this to population based death and cancer registries provides a worldwide unique source of population-based data which can be analysed to identify heterogeneities in risk by lifetime parity and the cumulative experience of pregnancy complications. Having worked in this field of research for many years, I see many erroneous conclusions in studies based on insufficient data. For instance, both after preeclampsia and after a stillbirth, the high risk of heart disease observed in one-child mothers is strongly attenuated in women with subsequent pregnancies. I will study different patterns of pregnancy complications that occur alone or in combination across pregnancies, and analyse their associations with cause specific maternal mortality. Using this unique methodology, I will challenge the idea that placental dysfunction is the origin of preeclampsia and test the hypothesis that pregnancy complications may cause direct long-term effects on maternal health. The findings of this research have the potential to advance our understanding of how pregnancy complications affect the long-term maternal health and help to develop more effective chronic disease prevention strategies.
Summary
Pregnancy complications such as preeclampsia and preterm birth are known to affect infant health, but their influence on mothers’ long-term health is not well understood. Most previous studies are seriously limited by their reliance on information from the first pregnancy. Often they lack the data to study women’s complete reproductive histories. Without a complete reproductive history, the relationship between pregnancy complications and women’s long-term health cannot be reliably studied. The Medical Birth Registry of Norway, covering all births from 1967-, includes information on more than 3 million births and 1.5 million sibships. Linking this to population based death and cancer registries provides a worldwide unique source of population-based data which can be analysed to identify heterogeneities in risk by lifetime parity and the cumulative experience of pregnancy complications. Having worked in this field of research for many years, I see many erroneous conclusions in studies based on insufficient data. For instance, both after preeclampsia and after a stillbirth, the high risk of heart disease observed in one-child mothers is strongly attenuated in women with subsequent pregnancies. I will study different patterns of pregnancy complications that occur alone or in combination across pregnancies, and analyse their associations with cause specific maternal mortality. Using this unique methodology, I will challenge the idea that placental dysfunction is the origin of preeclampsia and test the hypothesis that pregnancy complications may cause direct long-term effects on maternal health. The findings of this research have the potential to advance our understanding of how pregnancy complications affect the long-term maternal health and help to develop more effective chronic disease prevention strategies.
Max ERC Funding
2 500 000 €
Duration
Start date: 2019-09-01, End date: 2024-08-31
Project acronym HEALTHMPOWR
Project A New Paradigm for Public Health Surveillance: Unlocking the Potential of Data to Empower Women and Health Systems
Researcher (PI) Jahn Frederik Froen
Host Institution (HI) FOLKEHELSEINSTITUTTET
Call Details Consolidator Grant (CoG), LS7, ERC-2013-CoG
Summary Rationale: State of the art health registries, cornerstones of public health surveillance, have hardly capitalized on the information and communication technology revolution. Many continue to be static, fragmented, and passive data repositories disseminating outdated reports only to a closed loop of public health officials.
Aim: In a radical departure from traditional science, this proposal introduces a new paradigm for public health surveillance: Maximizing the potential of data by disseminating data-driven individualized real-time information directly to women and providers to empower self-care in pregnancy and better healthcare delivery.
Research question: Is routine, data-driven and automated feedback from a reproductive health registry (RHR) effective in improving health behaviour and quality of care?
Plan: Based on the roll-out of a RHR in the Palestinian West Bank, four stepped wedged cluster randomized controlled trials will be undertaken to investigate the comparative effectiveness of a series of feedback modules to women and care providers. Main outcomes include adherence to evidence-based guidelines for providers, and self-care and care seeking among women.
Impact: Coalescing with WHO/NIPH’s dissemination of the harmonized Reproductive Health Registries (hRHR) Initiative, the scientific horizons emerging from this proposal have potential for exceptional impact. Radically transforming public health surveillance by empowering women and health care providers with information can translate into better health care and behaviours thus saving lives.
Summary
Rationale: State of the art health registries, cornerstones of public health surveillance, have hardly capitalized on the information and communication technology revolution. Many continue to be static, fragmented, and passive data repositories disseminating outdated reports only to a closed loop of public health officials.
Aim: In a radical departure from traditional science, this proposal introduces a new paradigm for public health surveillance: Maximizing the potential of data by disseminating data-driven individualized real-time information directly to women and providers to empower self-care in pregnancy and better healthcare delivery.
Research question: Is routine, data-driven and automated feedback from a reproductive health registry (RHR) effective in improving health behaviour and quality of care?
Plan: Based on the roll-out of a RHR in the Palestinian West Bank, four stepped wedged cluster randomized controlled trials will be undertaken to investigate the comparative effectiveness of a series of feedback modules to women and care providers. Main outcomes include adherence to evidence-based guidelines for providers, and self-care and care seeking among women.
Impact: Coalescing with WHO/NIPH’s dissemination of the harmonized Reproductive Health Registries (hRHR) Initiative, the scientific horizons emerging from this proposal have potential for exceptional impact. Radically transforming public health surveillance by empowering women and health care providers with information can translate into better health care and behaviours thus saving lives.
Max ERC Funding
2 212 136 €
Duration
Start date: 2014-09-01, End date: 2019-08-31
Project acronym HoloVision
Project Advanced holographic optical neural stimulation for vision restoration and basic research
Researcher (PI) Shy Shoham
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Consolidator Grant (CoG), LS7, ERC-2014-CoG
Summary When natural sensory input is disrupted, as in outer-retinal degenerative diseases, artificial stimulation of surviving nerve cells offers a potential strategy for bypassing compromised neural circuits and substituting sensory perception. Current neuro-stimulation interfaces that use electrical currents from micro-electrode arrays are already being clinically applied for retinal stimulation, but their performance is ultimately limited by current spread and the requirement for physical contact with an implanted device. Future minimally-invasive systems could use light patterns to photo-induce complex yet precise spatio-temporal activity patterns among surviving retinal neurons, with the ultimate potential of restoring vision to a nearly normal level.
Here, we will advance, optimize and test in vivo a powerful new strategy for cellular-resolution controlled patterned optogenetic excitation, based on computer-generated holographic optical neural-stimulation (HONS). Regular (one-photon) HONS systems can dynamically address large populations of optogenetically-transduced retinal ganglion cells with single-cell resolution, while related multiphoton HONS systems can extend these capabilities to three-dimensional cortical tissue (relevant to many research applications). A series of in vivo experiments will resolve basic questions regarding the efficacy of these approaches by directly examining the retinal and cortical responses to structured holographic photo-stimulation, and test novel strategies for improving it. Finally, as a major step towards clinical translation of this technology, we will design and evaluate (in blind sheep and sighted individuals) a human-scale prototype.
Overall, by combining both basic and translational research, this study will advance novel optical neuro-technologies with potential impact on multiple scientific and clinical applications. Specifically, it will tackle the major engineering requirements and constraints towards the development of a
Summary
When natural sensory input is disrupted, as in outer-retinal degenerative diseases, artificial stimulation of surviving nerve cells offers a potential strategy for bypassing compromised neural circuits and substituting sensory perception. Current neuro-stimulation interfaces that use electrical currents from micro-electrode arrays are already being clinically applied for retinal stimulation, but their performance is ultimately limited by current spread and the requirement for physical contact with an implanted device. Future minimally-invasive systems could use light patterns to photo-induce complex yet precise spatio-temporal activity patterns among surviving retinal neurons, with the ultimate potential of restoring vision to a nearly normal level.
Here, we will advance, optimize and test in vivo a powerful new strategy for cellular-resolution controlled patterned optogenetic excitation, based on computer-generated holographic optical neural-stimulation (HONS). Regular (one-photon) HONS systems can dynamically address large populations of optogenetically-transduced retinal ganglion cells with single-cell resolution, while related multiphoton HONS systems can extend these capabilities to three-dimensional cortical tissue (relevant to many research applications). A series of in vivo experiments will resolve basic questions regarding the efficacy of these approaches by directly examining the retinal and cortical responses to structured holographic photo-stimulation, and test novel strategies for improving it. Finally, as a major step towards clinical translation of this technology, we will design and evaluate (in blind sheep and sighted individuals) a human-scale prototype.
Overall, by combining both basic and translational research, this study will advance novel optical neuro-technologies with potential impact on multiple scientific and clinical applications. Specifically, it will tackle the major engineering requirements and constraints towards the development of a
Max ERC Funding
2 624 517 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym HybridRetina
Project Hybrid Retinal Prosthesis: High-Resolution Electrode Array Integrated with Neurons for Restoration of Sight
Researcher (PI) Yosef Mandel
Host Institution (HI) BAR ILAN UNIVERSITY
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary Vision restoration in patients with outer retinal degenerative diseases, such as Age-related Macular Degeneration and Retinitis Pigmentosa can be achieved by bypassing the degenerated photoreceptors and the electrical stimulation of the relatively well-preserved inner retina through electrode implants. Although current retinal prostheses have been shown to provide useful vision in blind patients, the obtained visual acuity and quality are still relatively low. Several challenges cannot be addressed with the current retinal prosthetic technologies. First, increasing the electrode density for achieving high visual acuity is limited by the distance between the electrodes and the target neurons. Second, electrical stimulation by the current technologies is not selective for specific retinal circuitry (e.g. ON and OFF pathways). Finally, retinal neurons are stimulated by pulsed rather than in a continuously graded potential fashion, which provides the photoreceptors with an unrivalled dynamic range and sensitivity in natural vision.
Here we propose a paradigm shift toward sight restoration with a hybrid retinal prosthesis aimed at overcoming the aforementioned limitations. The hybrid implant is composed of a very high density electrode array (pixel distance of 15µm) coupled with neurons to create a tight neuron-electrode coupling. Following implantation of the hybrid prosthesis, the neurons integrate and synapse with the host retinal circuits. Upon patterned electrical stimulation of the neurons by the electrodes, the host bipolar cells are activated while preserving the natural vision circuits. The ultimate electrode-neurons proximity allows for the significant increase in pixel density, the low charge neural activation, and the continuous graded potential activation. This research can advance our knowledge in the retinal field and in other neural prosthetics and if successful, it will enable future vision restoration with unprecedented resolution.
Summary
Vision restoration in patients with outer retinal degenerative diseases, such as Age-related Macular Degeneration and Retinitis Pigmentosa can be achieved by bypassing the degenerated photoreceptors and the electrical stimulation of the relatively well-preserved inner retina through electrode implants. Although current retinal prostheses have been shown to provide useful vision in blind patients, the obtained visual acuity and quality are still relatively low. Several challenges cannot be addressed with the current retinal prosthetic technologies. First, increasing the electrode density for achieving high visual acuity is limited by the distance between the electrodes and the target neurons. Second, electrical stimulation by the current technologies is not selective for specific retinal circuitry (e.g. ON and OFF pathways). Finally, retinal neurons are stimulated by pulsed rather than in a continuously graded potential fashion, which provides the photoreceptors with an unrivalled dynamic range and sensitivity in natural vision.
Here we propose a paradigm shift toward sight restoration with a hybrid retinal prosthesis aimed at overcoming the aforementioned limitations. The hybrid implant is composed of a very high density electrode array (pixel distance of 15µm) coupled with neurons to create a tight neuron-electrode coupling. Following implantation of the hybrid prosthesis, the neurons integrate and synapse with the host retinal circuits. Upon patterned electrical stimulation of the neurons by the electrodes, the host bipolar cells are activated while preserving the natural vision circuits. The ultimate electrode-neurons proximity allows for the significant increase in pixel density, the low charge neural activation, and the continuous graded potential activation. This research can advance our knowledge in the retinal field and in other neural prosthetics and if successful, it will enable future vision restoration with unprecedented resolution.
Max ERC Funding
1 499 582 €
Duration
Start date: 2018-05-01, End date: 2023-04-30
Project acronym Hyperpolarized MRI
Project Citicoline and deoxyglucose as new molecular imaging probes of DNP hyperpolarized MRI for cancer and neuroimaging
Researcher (PI) Rachel Katz-Brull
Host Institution (HI) HADASSAH MEDICAL ORGANIZATION
Call Details Starting Grant (StG), LS7, ERC-2013-StG
Summary Radioactively labeled deoxyglucose and choline are the leading molecular imaging probes for positron emission tomography (PET). The clinical applications for this imaging modality include brain function, cardiac imaging, and inflammation, along with oncological applications which are taking the lead. The radiation exposure associated with these examinations is limiting the use of this powerful technology in repeated examinations, in specific populations (pregnant women and children), as a screening tool for the wide population, and as a clinical research tool.
Hyperpolarized magnetic resonance imaging (MRI) is an evolving pre-clinical and clinical imaging modality which is non-invasive and non-radioactive. As in PET, the molecular imaging probe used is at the heart of this examination. Originally developed for the purpose of distinguishing the metabolic products of the injected molecular probe, our group, in collaborations with researchers abroad, is a pioneer in showing that direct imaging of specific molecular probes (stable isotope labeled choline and glucose analogs) with hyperpolarized MRI is capable of showing specific tissue uptake, a pre-requisite for diagnostic imaging.
The purpose of the current proposal is to establish hyperpolarized MRI capabilities in our own lab and reach two general goals: 1) to use various physiological and pharmacological models to further establish and characterize the conditions in which non-radioactive choline and glucose analogs and derivatives can be useful as imaging probes; and 2) to investigate further the molecular probe that is best suitable for these imaging applications in terms of pharmacokinetics, metabolism, and imaging efficiency. Our focus will be on 1) the actual chemical entity of the probes - where citicoline and deoxyglucose are promising candidates; and 2) the stable isotope labeling strategy. The overriding goal is to aid in translation of this pre-clinical imaging approach to clinical use.
Summary
Radioactively labeled deoxyglucose and choline are the leading molecular imaging probes for positron emission tomography (PET). The clinical applications for this imaging modality include brain function, cardiac imaging, and inflammation, along with oncological applications which are taking the lead. The radiation exposure associated with these examinations is limiting the use of this powerful technology in repeated examinations, in specific populations (pregnant women and children), as a screening tool for the wide population, and as a clinical research tool.
Hyperpolarized magnetic resonance imaging (MRI) is an evolving pre-clinical and clinical imaging modality which is non-invasive and non-radioactive. As in PET, the molecular imaging probe used is at the heart of this examination. Originally developed for the purpose of distinguishing the metabolic products of the injected molecular probe, our group, in collaborations with researchers abroad, is a pioneer in showing that direct imaging of specific molecular probes (stable isotope labeled choline and glucose analogs) with hyperpolarized MRI is capable of showing specific tissue uptake, a pre-requisite for diagnostic imaging.
The purpose of the current proposal is to establish hyperpolarized MRI capabilities in our own lab and reach two general goals: 1) to use various physiological and pharmacological models to further establish and characterize the conditions in which non-radioactive choline and glucose analogs and derivatives can be useful as imaging probes; and 2) to investigate further the molecular probe that is best suitable for these imaging applications in terms of pharmacokinetics, metabolism, and imaging efficiency. Our focus will be on 1) the actual chemical entity of the probes - where citicoline and deoxyglucose are promising candidates; and 2) the stable isotope labeling strategy. The overriding goal is to aid in translation of this pre-clinical imaging approach to clinical use.
Max ERC Funding
1 650 000 €
Duration
Start date: 2013-11-01, End date: 2018-10-31
Project acronym IMAGO
Project Imaging regulatory pathways of angiogenesis
Researcher (PI) Michal Neeman
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS7, ERC-2008-AdG
Summary Homeostasis of multicellular tissues relies on accurate match of vascular supply and drain to the needs of the tissue. Multiple pathways are involved in detection, signalling and execution of the required steps involved in organization of blood and lymphatic vessels during embryonic development. Similar mechanisms are utilized for overcoming changes in tissue requirements also in adult tissues and in pathological processes. The goal of this work is to reveal the dynamic forces that shape the blood vessels during angiogenesis. In particular, we would like to explore the impact of interstitial convective flow in dynamic imprinting of growth factor signalling, thereby regulating vascular patterning. Angiogenesis is explored here as an example for a possible general role for interstitial convection of growth factors in determination of the fine spatial patterning of tissue morphogenesis in vertebrates. To achieve this goal, we will develop multi-modality tools for imaging the regulation of vascular patterning. In vivo imaging will then be utilized for mapping vascular patterning in pathological and physiological angiogenesis including tumours, wound repair, the preovulatory ovarian follicle and foetal implantation sites. Whole body optical, CT, ultrasound and MRI will be applied for non-invasive imaging of deep organs. Microscopic morphometric and molecular information will be derived from the macroscopic imaging data, using selective molecular imaging approaches and functional imaging tools with specific pharmacological models that will be developed to account for interstitial convective flow. Intravital two photon microscopy and fluorescence endoscopy will be used for high resolution evaluation of vascular patterning. The evaluation of novel mechanisms for spatial regulation of intercellular growth factor signalling, will allow us to define new potential targets for intervention, and to develop new tools for preclinical and clinical imaging of angiogenesis.
Summary
Homeostasis of multicellular tissues relies on accurate match of vascular supply and drain to the needs of the tissue. Multiple pathways are involved in detection, signalling and execution of the required steps involved in organization of blood and lymphatic vessels during embryonic development. Similar mechanisms are utilized for overcoming changes in tissue requirements also in adult tissues and in pathological processes. The goal of this work is to reveal the dynamic forces that shape the blood vessels during angiogenesis. In particular, we would like to explore the impact of interstitial convective flow in dynamic imprinting of growth factor signalling, thereby regulating vascular patterning. Angiogenesis is explored here as an example for a possible general role for interstitial convection of growth factors in determination of the fine spatial patterning of tissue morphogenesis in vertebrates. To achieve this goal, we will develop multi-modality tools for imaging the regulation of vascular patterning. In vivo imaging will then be utilized for mapping vascular patterning in pathological and physiological angiogenesis including tumours, wound repair, the preovulatory ovarian follicle and foetal implantation sites. Whole body optical, CT, ultrasound and MRI will be applied for non-invasive imaging of deep organs. Microscopic morphometric and molecular information will be derived from the macroscopic imaging data, using selective molecular imaging approaches and functional imaging tools with specific pharmacological models that will be developed to account for interstitial convective flow. Intravital two photon microscopy and fluorescence endoscopy will be used for high resolution evaluation of vascular patterning. The evaluation of novel mechanisms for spatial regulation of intercellular growth factor signalling, will allow us to define new potential targets for intervention, and to develop new tools for preclinical and clinical imaging of angiogenesis.
Max ERC Funding
2 278 344 €
Duration
Start date: 2009-01-01, End date: 2013-12-31
Project acronym ImPRESS
Project Imaging Perfusion Restrictions from Extracellular Solid Stress
Researcher (PI) Kyrre Eeg Emblem
Host Institution (HI) OSLO UNIVERSITETSSYKEHUS HF
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary Even the perfect cancer drug must reach its target to have an effect. The ImPRESS project main objective is to develop a novel imaging paradigm coined Restricted Perfusion Imaging (RPI) to reveal - for the first time in humans - vascular restrictions in solid cancers caused by mechanical solid stress, and use RPI to demonstrate that alleviating this force will repair the cancerous microenvironment and improve therapeutic response. Delivery of anti-cancer drugs to the tumor is critically dependent on a functional vascular bed. Developing biomarkers that can measure how mechanical forces in a solid tumor impair perfusion and promotes therapy resistance is essential for treatment of disease.
The ImPRESS project is based on the following observations; (I) pre-clinical work suggests that therapies targeting the tumor microenvironment and extracellular matrix may enhance drug delivery by decompressing tumor vessels; (II) results from animal models may not be transferable because compressive forces in human tumors in vivo can be many times higher; and (III) there are no available imaging technologies for medical diagnostics of solid stress in human cancers. Using RPI, ImPRESS will conduct a comprehensive series of innovative studies in brain cancer patients to answer three key questions: (Q1) Can we image vascular restrictions in human cancers and map how the vasculature changes with tumor growth or treatment? (Q2) Can we use medical engineering to image solid stress in vivo? (Q3) Can RPI show that matrix-depleting drugs improve patient response to conventional chemo- and radiation therapy as well as new targeted therapies?
The ImPRESS project holds a unique position to answer these questions by our unrivaled experience with advanced imaging of cancer patients. With successful delivery, ImPRESS will have a direct impact on patient treatment and establish an imaging paradigm that will pave the way for new scientific knowledge on how to revitalize cancer therapies.
Summary
Even the perfect cancer drug must reach its target to have an effect. The ImPRESS project main objective is to develop a novel imaging paradigm coined Restricted Perfusion Imaging (RPI) to reveal - for the first time in humans - vascular restrictions in solid cancers caused by mechanical solid stress, and use RPI to demonstrate that alleviating this force will repair the cancerous microenvironment and improve therapeutic response. Delivery of anti-cancer drugs to the tumor is critically dependent on a functional vascular bed. Developing biomarkers that can measure how mechanical forces in a solid tumor impair perfusion and promotes therapy resistance is essential for treatment of disease.
The ImPRESS project is based on the following observations; (I) pre-clinical work suggests that therapies targeting the tumor microenvironment and extracellular matrix may enhance drug delivery by decompressing tumor vessels; (II) results from animal models may not be transferable because compressive forces in human tumors in vivo can be many times higher; and (III) there are no available imaging technologies for medical diagnostics of solid stress in human cancers. Using RPI, ImPRESS will conduct a comprehensive series of innovative studies in brain cancer patients to answer three key questions: (Q1) Can we image vascular restrictions in human cancers and map how the vasculature changes with tumor growth or treatment? (Q2) Can we use medical engineering to image solid stress in vivo? (Q3) Can RPI show that matrix-depleting drugs improve patient response to conventional chemo- and radiation therapy as well as new targeted therapies?
The ImPRESS project holds a unique position to answer these questions by our unrivaled experience with advanced imaging of cancer patients. With successful delivery, ImPRESS will have a direct impact on patient treatment and establish an imaging paradigm that will pave the way for new scientific knowledge on how to revitalize cancer therapies.
Max ERC Funding
1 499 638 €
Duration
Start date: 2018-01-01, End date: 2022-12-31
Project acronym LeukoTheranostics
Project Harnessing Targeted Nanotheranostics to Reprogram Activated Leukocytes in Inflammatory Bowel Disease
Researcher (PI) Dan Peer
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Consolidator Grant (CoG), LS7, ERC-2014-CoG
Summary "Inflammatory bowel diseases (IBD) is a group of chronic inflammatory conditions of the gastrointestinal tract, including Crohn’s disease (CD) and Ulcerative Colitis (UC) that can impact both the large and small bowel. IBD affects approximately 3.7 million Europeans and its peak onset is in persons of 15 to 30 years of age. IBD imposes a significant burden on Europe with over €3B in annual health care costs and over €3B in indirect cost. The prevalence of IBD is expected to increase by more than 40% over the next decade in many European countries. Therefore, to meet the needs of IBD patients in the European community, we must prepare for the evolving landscape of IBD care in the near future. Although its etiology remains unknown, unregulated immune cells are implicated in the pathogenesis of IBD. As many IBD patients are refractory to conventional medical treatments, there is an urgent need to develop novel therapeutic modalities in combination with real-time imaging in order to manage the disease. I will achieve this goal by generating a ""Trojan horse"" strategy of targeting activated leukocytes that home to the gut in IBD rodent models and reprogram their fate using RNA interference (RNAi) combined with molecular imaging.
The primary objective of this proposal is to reprogram in vivo activated leukocytes involved in gut inflammation using advanced RNAi-based therapeutics combined with molecular imaging strategies as the first theranostic modality utilizing leukocytes. The following specific aims include: (i) To develop and characterize unique integrin-targeted nanoparticles (I-tsNPs) targeting a high-affinity (HA) conformation of a4b7 integrin expressed on gut leukocytes; (ii) To study I-tsNPs 3-dimensional (3-D) delivery in colitis models using microPET/CT imaging; (iii) To investigate efficacy and safety profiles using the HA I-tsNPs platform for IBD therapeutics and disease management that will lay the foundation for future clinical trials.
"
Summary
"Inflammatory bowel diseases (IBD) is a group of chronic inflammatory conditions of the gastrointestinal tract, including Crohn’s disease (CD) and Ulcerative Colitis (UC) that can impact both the large and small bowel. IBD affects approximately 3.7 million Europeans and its peak onset is in persons of 15 to 30 years of age. IBD imposes a significant burden on Europe with over €3B in annual health care costs and over €3B in indirect cost. The prevalence of IBD is expected to increase by more than 40% over the next decade in many European countries. Therefore, to meet the needs of IBD patients in the European community, we must prepare for the evolving landscape of IBD care in the near future. Although its etiology remains unknown, unregulated immune cells are implicated in the pathogenesis of IBD. As many IBD patients are refractory to conventional medical treatments, there is an urgent need to develop novel therapeutic modalities in combination with real-time imaging in order to manage the disease. I will achieve this goal by generating a ""Trojan horse"" strategy of targeting activated leukocytes that home to the gut in IBD rodent models and reprogram their fate using RNA interference (RNAi) combined with molecular imaging.
The primary objective of this proposal is to reprogram in vivo activated leukocytes involved in gut inflammation using advanced RNAi-based therapeutics combined with molecular imaging strategies as the first theranostic modality utilizing leukocytes. The following specific aims include: (i) To develop and characterize unique integrin-targeted nanoparticles (I-tsNPs) targeting a high-affinity (HA) conformation of a4b7 integrin expressed on gut leukocytes; (ii) To study I-tsNPs 3-dimensional (3-D) delivery in colitis models using microPET/CT imaging; (iii) To investigate efficacy and safety profiles using the HA I-tsNPs platform for IBD therapeutics and disease management that will lay the foundation for future clinical trials.
"
Max ERC Funding
2 703 125 €
Duration
Start date: 2015-11-01, End date: 2020-10-31
Project acronym METACYCLES
Project Uncovering metabolic cycles in mammals and dissecting their interplay with circadian clocks
Researcher (PI) Gad Asher
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS1, ERC-2012-StG_20111109
Summary The physiology and behavior of mammals are subject to daily oscillations that are driven by an endogenous circadian clock. The mammalian circadian timing system is composed of a central pacemaker in the brain that is entrained by daily light-dark cycles and in turn synchronizes subsidiary oscillators in virtually all cells of the body. The core clock molecular circuitry is based on interlocked negative transcription-translation feedback loops that generate daily oscillations of gene expression in cultured cells and living animals.
Circadian clocks play a major role in orchestrating daily metabolism and their disruption can lead to metabolic diseases such as diabetes and obesity. Concomitantly, circadian clocks are tightly coupled to cellular metabolism and respond to feeding cycles. The molecular mechanisms through which metabolism regulates clocks’ function are just starting to emerge. Recent work of ours and others revealed that NAD+/NADH are implicated in the function of circadian clocks, yet the molecular mechanisms involved are largely unknown. We propose to intensively study the role of NAD+/NADH in the function of circadian clocks and to reveal the underlying mechanisms.
The functional interplay between circadian clocks and metabolism raises the question whether there are daily cycles in cellular metabolism and intracellular metabolites. Hitherto, direct measurements of daily changes in cellular metabolism and intracellular metabolite levels are still in their infancy. Our overarching goal is to identify metabolic cycles in mammals and mechanistically address their interplay with circadian clocks. We will monitor metabolic outputs in intact cells and living animals and systemically measure daily changes in intracellular metabolites. Our findings are expected to push forward a paradigm shift in the circadian field from the current “transcriptional-translational clocks” to “metabolic clocks”.
Summary
The physiology and behavior of mammals are subject to daily oscillations that are driven by an endogenous circadian clock. The mammalian circadian timing system is composed of a central pacemaker in the brain that is entrained by daily light-dark cycles and in turn synchronizes subsidiary oscillators in virtually all cells of the body. The core clock molecular circuitry is based on interlocked negative transcription-translation feedback loops that generate daily oscillations of gene expression in cultured cells and living animals.
Circadian clocks play a major role in orchestrating daily metabolism and their disruption can lead to metabolic diseases such as diabetes and obesity. Concomitantly, circadian clocks are tightly coupled to cellular metabolism and respond to feeding cycles. The molecular mechanisms through which metabolism regulates clocks’ function are just starting to emerge. Recent work of ours and others revealed that NAD+/NADH are implicated in the function of circadian clocks, yet the molecular mechanisms involved are largely unknown. We propose to intensively study the role of NAD+/NADH in the function of circadian clocks and to reveal the underlying mechanisms.
The functional interplay between circadian clocks and metabolism raises the question whether there are daily cycles in cellular metabolism and intracellular metabolites. Hitherto, direct measurements of daily changes in cellular metabolism and intracellular metabolite levels are still in their infancy. Our overarching goal is to identify metabolic cycles in mammals and mechanistically address their interplay with circadian clocks. We will monitor metabolic outputs in intact cells and living animals and systemically measure daily changes in intracellular metabolites. Our findings are expected to push forward a paradigm shift in the circadian field from the current “transcriptional-translational clocks” to “metabolic clocks”.
Max ERC Funding
1 499 980 €
Duration
Start date: 2012-11-01, End date: 2017-10-31
Project acronym METSTEM
Project DNA methylation in stem cells
Researcher (PI) Howard Cedar
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS1, ERC-2010-AdG_20100317
Summary Embryonic and adult stem cells constitute an important component of biology by providing a pool of pluri- and multi-potent cells that supply a variety of different cell lineages. Little is known about the mechanisms involved in establishing and maintaining cell ¿stemness,¿ but it is most likely controlled by epigenetic signals such as DNA methylation. This proposal aims to understand these mechanisms and decipher the molecular logic used to program this plasticity.
We have developed a new strategy for studying the ¿DNA methylation potential¿ of any cell type throughout normal development. This utilizes a unique set of transgenic vectors programmed to detect both de novo methylation as well as the ability to protect CpG islands, and will, for the first time, allow one to evaluate the role of demethylation in normal stem cells and during reprogramming. This will be done using a new technique called ¿reverse epigenetics¿.
Preliminary studies indicate that embryonic stem cells differentiated in vitro undergo extensive aberrant methylation that does not reflect the normal pattern of methylation found in vivo. This artifact may be responsible for our inability to attain efficient differentiation in culture and may generate cells that are unhealthy and prone to cancer. We will characterize the causes of this phenomenon and decipher its underlying mechanism. This research should lead to the development of improved methods for tissue generation in vitro.
One of the most basic properties of adult stem cells is their ability to undergo asymmetric cell division that is often associated with unequal segregation of DNA. This mechanism is one of the most elemental, yet mysterious, aspects of stem cell biology. We have developed a completely new molecular model for this process that is based on the idea that non-symmetric DNA methylation serves as a strand-specific marker, and it is very likely that this will enable us to finally decipher this basic aspect of stem cells.
Summary
Embryonic and adult stem cells constitute an important component of biology by providing a pool of pluri- and multi-potent cells that supply a variety of different cell lineages. Little is known about the mechanisms involved in establishing and maintaining cell ¿stemness,¿ but it is most likely controlled by epigenetic signals such as DNA methylation. This proposal aims to understand these mechanisms and decipher the molecular logic used to program this plasticity.
We have developed a new strategy for studying the ¿DNA methylation potential¿ of any cell type throughout normal development. This utilizes a unique set of transgenic vectors programmed to detect both de novo methylation as well as the ability to protect CpG islands, and will, for the first time, allow one to evaluate the role of demethylation in normal stem cells and during reprogramming. This will be done using a new technique called ¿reverse epigenetics¿.
Preliminary studies indicate that embryonic stem cells differentiated in vitro undergo extensive aberrant methylation that does not reflect the normal pattern of methylation found in vivo. This artifact may be responsible for our inability to attain efficient differentiation in culture and may generate cells that are unhealthy and prone to cancer. We will characterize the causes of this phenomenon and decipher its underlying mechanism. This research should lead to the development of improved methods for tissue generation in vitro.
One of the most basic properties of adult stem cells is their ability to undergo asymmetric cell division that is often associated with unequal segregation of DNA. This mechanism is one of the most elemental, yet mysterious, aspects of stem cell biology. We have developed a completely new molecular model for this process that is based on the idea that non-symmetric DNA methylation serves as a strand-specific marker, and it is very likely that this will enable us to finally decipher this basic aspect of stem cells.
Max ERC Funding
1 941 930 €
Duration
Start date: 2011-04-01, End date: 2016-03-31
Project acronym MRECB
Project Molecular Robots Exhibiting Collective Behaviours
Researcher (PI) Ido Bachelet
Host Institution (HI) AUGMANITY NANO LTD
Call Details Starting Grant (StG), LS7, ERC-2013-StG
Summary Cells carry out biochemical and physiological functions in the body by intricately controlling the position and activity of matter at the nanoscale. In contrast, our ability to exert equivalent spatiotemporal control over therapeutic molecules is extremely inadequate. That is the reason why although we have very effective therapeutics, their use is often limited by excess toxicity and adverse effects.
We have recently described an autonomous, logic-guided DNA origami nanorobot, capable of transporting molecular payloads between selected points of origin and target, and demonstrated its selectivity and efficacy in inducing tumor cell apoptosis.
Here, we propose to create nanorobots that can be programmed to exhibit collective behaviours. This will provide unprecedented, nearly absolute spatiotemporal control over many therapeutic molecules simultaneously. Using the molecular equivalents of computer programs, which we describe here, these nanorobots will be able to communicate with each other, coordinate timing of activity, form meta-structures, exhibit quorum-sensing capabilities and emulate logical operations using biological cues as bits.
This proposal describes nanorobot design and control, and shows how the nanorobots can be modified to exhibit improved pharmacokinetic performance with negligible immunogenicity and toxicity. This technology could revolutionize therapeutic paradigms and procedures, along with other aspects of current medicine.
Summary
Cells carry out biochemical and physiological functions in the body by intricately controlling the position and activity of matter at the nanoscale. In contrast, our ability to exert equivalent spatiotemporal control over therapeutic molecules is extremely inadequate. That is the reason why although we have very effective therapeutics, their use is often limited by excess toxicity and adverse effects.
We have recently described an autonomous, logic-guided DNA origami nanorobot, capable of transporting molecular payloads between selected points of origin and target, and demonstrated its selectivity and efficacy in inducing tumor cell apoptosis.
Here, we propose to create nanorobots that can be programmed to exhibit collective behaviours. This will provide unprecedented, nearly absolute spatiotemporal control over many therapeutic molecules simultaneously. Using the molecular equivalents of computer programs, which we describe here, these nanorobots will be able to communicate with each other, coordinate timing of activity, form meta-structures, exhibit quorum-sensing capabilities and emulate logical operations using biological cues as bits.
This proposal describes nanorobot design and control, and shows how the nanorobots can be modified to exhibit improved pharmacokinetic performance with negligible immunogenicity and toxicity. This technology could revolutionize therapeutic paradigms and procedures, along with other aspects of current medicine.
Max ERC Funding
1 496 063 €
Duration
Start date: 2013-10-01, End date: 2018-09-30
Project acronym MTrix
Project Mechanical Targeting as an Integrative Approach for Personalized Nanomedicine
Researcher (PI) Ofra BENNY
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary The ability to direct drug delivery to specific tissues is a central challenge in treating diseases as it determines the balance between drug selectivity and toxicity. Clinical drug failures, commonly due to safety issues or poor efficacy, are extremely costly to the pharmaceutical industry. In light of this, there is a global effort to develop Targeted Drug Delivery Systems (DDS) and Nanomedicine-based drugs to increase the therapeutic efficacy of a drug while substantially reducing its off-target exposure. In oncology, this issue is critical since chemotherapies have poor selectivity, thus causing severe side effects due to undesired systemic exposure. However, the enormous heterogeneity and dynamic nature of tumors makes it extremely challenging to identify universal target molecules. In this ERC I introduce a novel concept according to which the specificity of DDS can be dramatically enhanced by tuning the physical parameters of DDS based on mechanical cues of target and non-target cells. In many cancers, it is well-established that the flexibility and deformability of cells are correlated with their metastatic potential. This leads to our hypothesis that the enhanced deformability of cancer cells allows them to engulf and uptake particles whose internalization requires massive shape change, unlike the stiffer and normal cells. The rationale of the proposed study is that by considering physical parameters of cells, the mechanical properties of DDS can be tuned to achieve selective uptake. We thus propose to develop tools for rational design of DDS for personalized nanomedicine that will use simple tests performed on a patient’s own cells. This is the basis of our visionary Mechanical Targeting (MT) scheme, a crosstalk between experimental and computational models, for drug specificity. Accordingly, this ERC is expected to yield breakthroughs, both conceptual and technical.
Summary
The ability to direct drug delivery to specific tissues is a central challenge in treating diseases as it determines the balance between drug selectivity and toxicity. Clinical drug failures, commonly due to safety issues or poor efficacy, are extremely costly to the pharmaceutical industry. In light of this, there is a global effort to develop Targeted Drug Delivery Systems (DDS) and Nanomedicine-based drugs to increase the therapeutic efficacy of a drug while substantially reducing its off-target exposure. In oncology, this issue is critical since chemotherapies have poor selectivity, thus causing severe side effects due to undesired systemic exposure. However, the enormous heterogeneity and dynamic nature of tumors makes it extremely challenging to identify universal target molecules. In this ERC I introduce a novel concept according to which the specificity of DDS can be dramatically enhanced by tuning the physical parameters of DDS based on mechanical cues of target and non-target cells. In many cancers, it is well-established that the flexibility and deformability of cells are correlated with their metastatic potential. This leads to our hypothesis that the enhanced deformability of cancer cells allows them to engulf and uptake particles whose internalization requires massive shape change, unlike the stiffer and normal cells. The rationale of the proposed study is that by considering physical parameters of cells, the mechanical properties of DDS can be tuned to achieve selective uptake. We thus propose to develop tools for rational design of DDS for personalized nanomedicine that will use simple tests performed on a patient’s own cells. This is the basis of our visionary Mechanical Targeting (MT) scheme, a crosstalk between experimental and computational models, for drug specificity. Accordingly, this ERC is expected to yield breakthroughs, both conceptual and technical.
Max ERC Funding
1 499 875 €
Duration
Start date: 2017-12-01, End date: 2022-11-30
Project acronym NeuroInCellNMR
Project In-cell NMR monitoring of alpha-Synuclein aggregation in neuronal cells
Researcher (PI) Philipp Selenko
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS1, ERC-2014-CoG
Summary Intracellular aggregation of the human amyloid protein alpha-synuclein is causally involved in Parkinson’s disease, a debilitating neurodegenerative disorder. The goal of this project is to combine low-resolution, fluorescence-imaging methods with high-resolution in-cell NMR and EPR spectroscopy techniques to derive macroscopic and microscopic insights into alpha-synuclein aggregate structures directly in neuronal cells.
To achieve this goal, we will employ different sets of cultured neurons and investigate intracellular alpha-synuclein aggregation under defined conditions of mitochondrial dysfunction and cellular oxidative stress, two of the most common denominators of the disease. Importantly, we will also establish a human stem cell model for studying alpha-synuclein aggregation with high-resolution in-cell NMR and EPR methods, by using induced pluripotent stem cell (iPSC) derived dopaminergic neurons from Parkinson’s disease patients and control individuals.
Results from this study will provide novel insights into the native mechanisms of intracellular aggregate formation and ultimately enable novel pharmacological approaches for therapeutic intervention.
Summary
Intracellular aggregation of the human amyloid protein alpha-synuclein is causally involved in Parkinson’s disease, a debilitating neurodegenerative disorder. The goal of this project is to combine low-resolution, fluorescence-imaging methods with high-resolution in-cell NMR and EPR spectroscopy techniques to derive macroscopic and microscopic insights into alpha-synuclein aggregate structures directly in neuronal cells.
To achieve this goal, we will employ different sets of cultured neurons and investigate intracellular alpha-synuclein aggregation under defined conditions of mitochondrial dysfunction and cellular oxidative stress, two of the most common denominators of the disease. Importantly, we will also establish a human stem cell model for studying alpha-synuclein aggregation with high-resolution in-cell NMR and EPR methods, by using induced pluripotent stem cell (iPSC) derived dopaminergic neurons from Parkinson’s disease patients and control individuals.
Results from this study will provide novel insights into the native mechanisms of intracellular aggregate formation and ultimately enable novel pharmacological approaches for therapeutic intervention.
Max ERC Funding
1 996 500 €
Duration
Start date: 2015-11-01, End date: 2021-02-28
Project acronym NIPD
Project A Novel Non-Invasive Prenatal Diagnosis for Genetic Disorders
Researcher (PI) Philippos Patsalis
Host Institution (HI) NIPD GENETICS PUBLIC COMPANY LIMITED
Call Details Advanced Grant (AdG), LS7, ERC-2012-ADG_20120314
Summary Non-Invasive Prenatal Diagnosis (NIPD) has been one of the most fascinating research fields during the last decade. The identification of small amounts of fetal DNA in maternal circulation has opened new possibilities for NIPD. Up until today, two methods have achieved accurate and validated NIPD methods for trisomy 21. The first NIPD for trisomy 21 was based on next generation sequencing and the second was developed by our group and is based on a MeDIP real time qPCR. However, nothing has been achieved for the NIPD of other genomic disorders caused by pathogenic copy number changes or mutations. The primary goal of this proposal is to develop, validate and provide to clinical practice a novel NIPD method, which will enable fast, sensitive, accurate, robust and cost effective NIPD of the great majority of genetic disorders caused by either pathogenic copy number changes of genomic segments or single and small size mutations. Initially, biomarkers with differential methylation between fetal and maternal DNA located within the entire human exome will be identified using methylation DNA immunoprecipitation and whole-exome massive parallel sequencing. Then a novel MeDIP exome NGS NIPD method for the great majority (~85%) of genetic disorders will be developed and validated. The method will undergo a blind evaluation study using 300 normal and abnormal maternal peripheral blood samples of pregnant women at 10-12 week of gestation. The intellectual property which may arise will be protected by filing internationally PCT patent(s) followed by dissemination of the results of the project. The new method will not only provide a greater number of highly accurate prenatal diagnoses of genetic disorders, but will do so without any risk for the fetus. Thus, the provision of such prenatal diagnoses may be provided to all pregnant women. The proposed proposal goes beyond the current state of the art and provides multiple medical, social and economic benefits.
Summary
Non-Invasive Prenatal Diagnosis (NIPD) has been one of the most fascinating research fields during the last decade. The identification of small amounts of fetal DNA in maternal circulation has opened new possibilities for NIPD. Up until today, two methods have achieved accurate and validated NIPD methods for trisomy 21. The first NIPD for trisomy 21 was based on next generation sequencing and the second was developed by our group and is based on a MeDIP real time qPCR. However, nothing has been achieved for the NIPD of other genomic disorders caused by pathogenic copy number changes or mutations. The primary goal of this proposal is to develop, validate and provide to clinical practice a novel NIPD method, which will enable fast, sensitive, accurate, robust and cost effective NIPD of the great majority of genetic disorders caused by either pathogenic copy number changes of genomic segments or single and small size mutations. Initially, biomarkers with differential methylation between fetal and maternal DNA located within the entire human exome will be identified using methylation DNA immunoprecipitation and whole-exome massive parallel sequencing. Then a novel MeDIP exome NGS NIPD method for the great majority (~85%) of genetic disorders will be developed and validated. The method will undergo a blind evaluation study using 300 normal and abnormal maternal peripheral blood samples of pregnant women at 10-12 week of gestation. The intellectual property which may arise will be protected by filing internationally PCT patent(s) followed by dissemination of the results of the project. The new method will not only provide a greater number of highly accurate prenatal diagnoses of genetic disorders, but will do so without any risk for the fetus. Thus, the provision of such prenatal diagnoses may be provided to all pregnant women. The proposed proposal goes beyond the current state of the art and provides multiple medical, social and economic benefits.
Max ERC Funding
2 500 000 €
Duration
Start date: 2013-05-01, End date: 2019-04-30
Project acronym NMR-DisAgg
Project The Dynamic Composition of the Protein Chaperone Network: Unraveling Human Protein Disaggregation via NMR Spectroscopy
Researcher (PI) Rina ROSENZWEIG
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS1, ERC-2018-STG
Summary Molecular chaperones are a diverse group of proteins critical to maintaining cellular homeostasis. Aside from protein refolding, it has recently been discovered that certain combinations of human chaperones can break apart toxic protein aggregates and even amyloids that have been linked to a host of neurodegenerative diseases. The first chaperones in this disaggregation reaction that are responsible for recognizing and performing initial remodeling of aggregates, are members of the Hsp40 (DnaJ) and small heat shock protein (sHSP) families. Very little, though, is known regarding how these chaperones perform their functions, and characterization of sHsp- and DnaJ-substrate complexes by most structural techniques has proven extremely challenging, as most chaperones are dynamic in nature and typically operate through a series of transient interactions with both their clients and other chaperones.
The advanced NMR techniques used in our lab, however, are ideally suited for the study of these exact types of dynamic systems, and include recently developed experiments (CEST, CPMG) that allow us to monitor the transient and low populated protein states typical of chaperone-chaperone and chaperone-client interactions, as well as to study the structure of these potentially very large protein complexes (methyl-TROSY).
By exploiting these NMR methodologies and additional, novel labeling schemes, we will characterize, for the first time, the recognition and substrate remodeling performed by the many members of the DnaJ and sHsp chaperone families on their clients. We will then take these approaches one step further and develop real time NMR experiments to observe the client remodeling performed over the course of the disaggregation reaction itself.
By combining advanced NMR with biophysical and functional assays, we ultimately aim to identify the specific sets of chaperones that, with the Hsp70 system, protect our cells by dissolving disease-linked aggregates and amyloid fibers.
Summary
Molecular chaperones are a diverse group of proteins critical to maintaining cellular homeostasis. Aside from protein refolding, it has recently been discovered that certain combinations of human chaperones can break apart toxic protein aggregates and even amyloids that have been linked to a host of neurodegenerative diseases. The first chaperones in this disaggregation reaction that are responsible for recognizing and performing initial remodeling of aggregates, are members of the Hsp40 (DnaJ) and small heat shock protein (sHSP) families. Very little, though, is known regarding how these chaperones perform their functions, and characterization of sHsp- and DnaJ-substrate complexes by most structural techniques has proven extremely challenging, as most chaperones are dynamic in nature and typically operate through a series of transient interactions with both their clients and other chaperones.
The advanced NMR techniques used in our lab, however, are ideally suited for the study of these exact types of dynamic systems, and include recently developed experiments (CEST, CPMG) that allow us to monitor the transient and low populated protein states typical of chaperone-chaperone and chaperone-client interactions, as well as to study the structure of these potentially very large protein complexes (methyl-TROSY).
By exploiting these NMR methodologies and additional, novel labeling schemes, we will characterize, for the first time, the recognition and substrate remodeling performed by the many members of the DnaJ and sHsp chaperone families on their clients. We will then take these approaches one step further and develop real time NMR experiments to observe the client remodeling performed over the course of the disaggregation reaction itself.
By combining advanced NMR with biophysical and functional assays, we ultimately aim to identify the specific sets of chaperones that, with the Hsp70 system, protect our cells by dissolving disease-linked aggregates and amyloid fibers.
Max ERC Funding
1 499 956 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym NO-DISEASE
Project Developing novel therapies for systemic disorders by regulating Nitric Oxide (NO) substrates' availability
Researcher (PI) Ayelet Erez
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS1, ERC-2013-CoG
Summary Nitric oxide (NO) is an essential signalling molecule for diverse physiological and disease processes. The current paradigm of how NO production is regulated focuses at the level of nitric oxide synthase (NOS), with respect to substrate and co-factor availability and the precise spatial and temporal arrangement of protein complexes. However, the respective unique or combined genetic deficiencies of the NOS isoforms exhibit relatively modest phenotypes in mice. Moreover, approaches targeted at modulating NOS activities have not successfully translated into different disease applications. All NOS isoforms are dependent on arginine as their sole substrate and interestingly, only one enzyme in mammals argininosuccinic lyase (ASL), can generate endogenous arginine. We propose that global regulation of NO production occurs earlier at the step of regulating arginine substrate availability within the cell. Until now, regulation at the level of arginine availability has been under-appreciated since arginine is readily available from external and dietary sources, irrespective of endogenous cellular production. However, extracellular arginine levels can affect NO production even though the intracellular levels of arginine are saturated.
In humans, the natural history of argininosuccinic aciduria caused by deficiency of ASL shows systemic and chronic features that reflect in part global dysregulation of NO homeostasis. This led us to discover that ASL is required for the channelling of both endogenously synthesized arginine and exogenous arginine to NOS. By challenging the existing paradigm, I hypothesize that regulating ASL would allow us to characterize the cellular and molecular mechanisms underlying NO flux regulation at normal and pathological conditions, for therapeutic applications. This proposal is hence novel both in its concept but also in its approach that is based on targeting therapy for systemic disorders through regulating cellular metabolism.
Summary
Nitric oxide (NO) is an essential signalling molecule for diverse physiological and disease processes. The current paradigm of how NO production is regulated focuses at the level of nitric oxide synthase (NOS), with respect to substrate and co-factor availability and the precise spatial and temporal arrangement of protein complexes. However, the respective unique or combined genetic deficiencies of the NOS isoforms exhibit relatively modest phenotypes in mice. Moreover, approaches targeted at modulating NOS activities have not successfully translated into different disease applications. All NOS isoforms are dependent on arginine as their sole substrate and interestingly, only one enzyme in mammals argininosuccinic lyase (ASL), can generate endogenous arginine. We propose that global regulation of NO production occurs earlier at the step of regulating arginine substrate availability within the cell. Until now, regulation at the level of arginine availability has been under-appreciated since arginine is readily available from external and dietary sources, irrespective of endogenous cellular production. However, extracellular arginine levels can affect NO production even though the intracellular levels of arginine are saturated.
In humans, the natural history of argininosuccinic aciduria caused by deficiency of ASL shows systemic and chronic features that reflect in part global dysregulation of NO homeostasis. This led us to discover that ASL is required for the channelling of both endogenously synthesized arginine and exogenous arginine to NOS. By challenging the existing paradigm, I hypothesize that regulating ASL would allow us to characterize the cellular and molecular mechanisms underlying NO flux regulation at normal and pathological conditions, for therapeutic applications. This proposal is hence novel both in its concept but also in its approach that is based on targeting therapy for systemic disorders through regulating cellular metabolism.
Max ERC Funding
1 915 555 €
Duration
Start date: 2014-03-01, End date: 2019-02-28
Project acronym NOVRIB
Project Novel Insights into Multi-drug Resistance to Antibiotics and the Primordial Ribosome
Researcher (PI) Ada Yonath
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS1, ERC-2012-ADG_20120314
Summary Multi-drug resistant phenotype formation creates global severe clinical threat among the most important challenges facing medicine today, dictating an urgent need for novel approaches. We aim to reveal the scope and mechanisms of resistance in pathogens, by studies that have not been pursued so far worldwide. In parallel we initiated innovative research towards understanding the ribosome origin, aiming at illuminating the transition from the primordial RNA world to the contemporary coded translation era, alongside exploring new targets and providing useful clues for antibiotics design. We base our interdisciplinary objectives on our discoveries originating from the ribosomes high resolution structures, the results of our pioneering efforts and subsequent perseverance.
By revealing unique properties of genuine pathogens that facilitate their exclusive resistance pathways,instead of depending solely on benign eubacterial models, we expect to gain matchless new insights. As no crystals of ribosomes from pathogens are available, we have initiated crystallographic studies, and present here preliminary results on two pathogenic life threatening bacteria, Staphylococcus aureus (associated with MRSA resistance) and Mycobacterium tuberculosis via Mycobacterium smegmatis that serve as its medical diagnostic tool. We also aim at experimentally defining the intra-ribosome region suggested by us to be a vestige of a prebiotic apparatus (proto-ribosome) by designing autonomous molecular entities with catalytic capabilities. Constructs the bind substrates have already been obtained. The expected enhancement in understanding peptide bond formation should lead to novel insights into this universal essential process. Our studies are designed to provide unprecedentedly powerful new tools for minimizing pathogens resistance thus should be of immense therapeutic relevance & will open up new horizons for researchers seeking response to challenges of the increasing antibiotic resistance.
Summary
Multi-drug resistant phenotype formation creates global severe clinical threat among the most important challenges facing medicine today, dictating an urgent need for novel approaches. We aim to reveal the scope and mechanisms of resistance in pathogens, by studies that have not been pursued so far worldwide. In parallel we initiated innovative research towards understanding the ribosome origin, aiming at illuminating the transition from the primordial RNA world to the contemporary coded translation era, alongside exploring new targets and providing useful clues for antibiotics design. We base our interdisciplinary objectives on our discoveries originating from the ribosomes high resolution structures, the results of our pioneering efforts and subsequent perseverance.
By revealing unique properties of genuine pathogens that facilitate their exclusive resistance pathways,instead of depending solely on benign eubacterial models, we expect to gain matchless new insights. As no crystals of ribosomes from pathogens are available, we have initiated crystallographic studies, and present here preliminary results on two pathogenic life threatening bacteria, Staphylococcus aureus (associated with MRSA resistance) and Mycobacterium tuberculosis via Mycobacterium smegmatis that serve as its medical diagnostic tool. We also aim at experimentally defining the intra-ribosome region suggested by us to be a vestige of a prebiotic apparatus (proto-ribosome) by designing autonomous molecular entities with catalytic capabilities. Constructs the bind substrates have already been obtained. The expected enhancement in understanding peptide bond formation should lead to novel insights into this universal essential process. Our studies are designed to provide unprecedentedly powerful new tools for minimizing pathogens resistance thus should be of immense therapeutic relevance & will open up new horizons for researchers seeking response to challenges of the increasing antibiotic resistance.
Max ERC Funding
2 487 989 €
Duration
Start date: 2013-02-01, End date: 2018-01-31
Project acronym NterAct
Project Discovery and functional significance of post-translational N-terminal acetylation
Researcher (PI) Thomas ARNESEN
Host Institution (HI) UNIVERSITETET I BERGEN
Call Details Consolidator Grant (CoG), LS1, ERC-2017-COG
Summary In mammalian cells, N-terminal (Nt) acetylation is one of the most abundant protein modifications. It is catalysed by N-terminal acetyltransferases (NATs) and mostly occurs co-translationally. However, in contrast to the defined co-translational NATs, post-translational NATs which have crucial regulatory roles are mostly unexplored. Distinct peptide hormones regulate appetite, metabolism, sexual behaviour and pain, and their biological activity is critically modulated by post-translational Nt-acetylation. However, the identity of the NAT responsible for this modification, ‘HormNat’, is unknown, thus the molecular and physiological ramifications of this regulatory circuit remain elusive. Another example is actin, a key regulator of cell motility and cell division. The actin N-terminus is crucial for actin function and in mammals actin is modified by an unknown post-translational NAT, ‘ActNat’. Hence, the objectives of this project are to identify these human NATs acting post-translationally, and to investigate their molecular mechanisms, regulation and impact.
We will identify the novel NATs by a combination of classical and newly developed in-house tools like in vitro acetylation assays, unique bisubstrate analogues, Nt-acetylation specific antibodies, and targeted mass spectrometry. Interestingly, Nt-acetylation is considered irreversible, but there is reason to believe that specific substrates are Nt-deacetylated. Elucidation of post-translational NATs and the reversible nature of Nt-acetylation would represent a new era in the field of protein and peptide regulation and identify key cellular and organismal switches.
Summary
In mammalian cells, N-terminal (Nt) acetylation is one of the most abundant protein modifications. It is catalysed by N-terminal acetyltransferases (NATs) and mostly occurs co-translationally. However, in contrast to the defined co-translational NATs, post-translational NATs which have crucial regulatory roles are mostly unexplored. Distinct peptide hormones regulate appetite, metabolism, sexual behaviour and pain, and their biological activity is critically modulated by post-translational Nt-acetylation. However, the identity of the NAT responsible for this modification, ‘HormNat’, is unknown, thus the molecular and physiological ramifications of this regulatory circuit remain elusive. Another example is actin, a key regulator of cell motility and cell division. The actin N-terminus is crucial for actin function and in mammals actin is modified by an unknown post-translational NAT, ‘ActNat’. Hence, the objectives of this project are to identify these human NATs acting post-translationally, and to investigate their molecular mechanisms, regulation and impact.
We will identify the novel NATs by a combination of classical and newly developed in-house tools like in vitro acetylation assays, unique bisubstrate analogues, Nt-acetylation specific antibodies, and targeted mass spectrometry. Interestingly, Nt-acetylation is considered irreversible, but there is reason to believe that specific substrates are Nt-deacetylated. Elucidation of post-translational NATs and the reversible nature of Nt-acetylation would represent a new era in the field of protein and peptide regulation and identify key cellular and organismal switches.
Max ERC Funding
1 999 273 €
Duration
Start date: 2018-06-01, End date: 2023-05-31
Project acronym OCLD
Project Tracking the Dynamics of Human Metabolism using Spectroscopy-Integrated Liver-on-Chip Microdevices
Researcher (PI) Yaakov Nahmias
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Consolidator Grant (CoG), LS7, ERC-2015-CoG
Summary The liver is the main organ responsible for the systemic regulation of human metabolism, responding to hormonal stimulation, nutritional challenges, and circadian rhythms using fast enzymatic processes and slow transcriptional mechanisms. This regulatory complexity limits our ability to create efficient pharmaceutical interventions for metabolic diseases such as fatty liver disease and diabetes. In addition, circadian changes in drug metabolism can impact pharmacokinetics and pharmacodynamics affecting our ability to optimize drug dosage or properly assess chronic liver toxicity.
The challenge in rationally designing efficient drug interventions stems from current reliance on end-point assays and animal models that provide intermittent information with limited human relevance. Therefore, there is a need to develop systems capable of tracking transcriptional and metabolic dynamics of human tissue with high-resolution preferably in real time. Over the past 5 years, we established state-of-the-art models of human hepatocytes; oxygen nanosensors; and cutting-edge liver-on-chip devices, making us uniquely suited to address this challenge.
We aim to develop a platform capable of tracking the metabolism of tissue engineered livers in real time, enabling an accurate assessment of chronic liver toxicity (e.g. repeated dose response) and the deconstruction of complex metabolic regulation during nutritional events. Our approach is to integrate liver-on-chip devices, with real time measurements of oxygen uptake, infrared microspectroscopy, and continuous MS/MS analysis. This innovative endeavour capitalizes on advances in nanotechnology and chemical characterization offering the ability to non-invasively monitor the metabolic state of the cells (e.g. steatosis) while tracking minute changes in metabolic pathways. This project has the short-term potential to replace animal models in toxicity studies and long-term potential to elucidate critical aspects in metabolic homeostasis.
Summary
The liver is the main organ responsible for the systemic regulation of human metabolism, responding to hormonal stimulation, nutritional challenges, and circadian rhythms using fast enzymatic processes and slow transcriptional mechanisms. This regulatory complexity limits our ability to create efficient pharmaceutical interventions for metabolic diseases such as fatty liver disease and diabetes. In addition, circadian changes in drug metabolism can impact pharmacokinetics and pharmacodynamics affecting our ability to optimize drug dosage or properly assess chronic liver toxicity.
The challenge in rationally designing efficient drug interventions stems from current reliance on end-point assays and animal models that provide intermittent information with limited human relevance. Therefore, there is a need to develop systems capable of tracking transcriptional and metabolic dynamics of human tissue with high-resolution preferably in real time. Over the past 5 years, we established state-of-the-art models of human hepatocytes; oxygen nanosensors; and cutting-edge liver-on-chip devices, making us uniquely suited to address this challenge.
We aim to develop a platform capable of tracking the metabolism of tissue engineered livers in real time, enabling an accurate assessment of chronic liver toxicity (e.g. repeated dose response) and the deconstruction of complex metabolic regulation during nutritional events. Our approach is to integrate liver-on-chip devices, with real time measurements of oxygen uptake, infrared microspectroscopy, and continuous MS/MS analysis. This innovative endeavour capitalizes on advances in nanotechnology and chemical characterization offering the ability to non-invasively monitor the metabolic state of the cells (e.g. steatosis) while tracking minute changes in metabolic pathways. This project has the short-term potential to replace animal models in toxicity studies and long-term potential to elucidate critical aspects in metabolic homeostasis.
Max ERC Funding
2 118 175 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym onCOMBINE
Project Towards evidence-based combinations of approved and novel cancer drugs
Researcher (PI) Yosef YARDEN
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS7, ERC-2016-ADG
Summary Background: Molecular targeted therapy (TT; e.g., monoclonal antibodies, mAbs, and protein kinase inhibitors, PKIs) intercepts oncogene and other addictions of tumours. However, unlike chemotherapy, which employs cocktails of drugs, only rarely does TT harness poly-pharmacology. Because lung cancer is the major cause of oncology related fatalities and many driver mutations are known, this disease offers opportunities for establishing and generalizing novel TT combinations and their interface with the immune system.
Working hypothesis: High granularity maps of compensatory loops evoked by TT, along with deeper understanding of mechanisms underlying drug action, resistance and interactions with lymphoid/myeloid cells, will conceptualize drug combinations able to persistently inhibit tumours, while inducing only limited toxicities.
Goal and specific aims: Addressing resistance to TT, potential synergies and the immune system, we will employ lung cancer models driven by mutant EGFR, HER2, MET or AXL. Phosphoproteomics, transcriptomics and RNA interference, will enable mapping adaptations evoked by specific drugs. Once identified, we will test combinations of interceptors able to inhibit the primary target as well as the emerging, resistance-conferring route(s). Next, we will determine the mechanisms of action of selected interceptors (e.g., apoptosis, immunological cytotoxicity and senescence) as bases for optimising effective combinations. Homo-combinations of antibodies (i.e., antibodies recognising distinct epitopes of a receptor), hetero-combinations targeting distinct signalling and immune receptors, and combinations with PKIs will be examined in animal models.
Significance: More than 30 PKIs and >25 mAbs are approved in oncology, but most are used as monotherapies. Detailed knowledge of adaptation-driven resistance, mechanisms of drug action and immune effectors, will guide the long awaited application of TT combinations in oncology, including lung cancer.
Summary
Background: Molecular targeted therapy (TT; e.g., monoclonal antibodies, mAbs, and protein kinase inhibitors, PKIs) intercepts oncogene and other addictions of tumours. However, unlike chemotherapy, which employs cocktails of drugs, only rarely does TT harness poly-pharmacology. Because lung cancer is the major cause of oncology related fatalities and many driver mutations are known, this disease offers opportunities for establishing and generalizing novel TT combinations and their interface with the immune system.
Working hypothesis: High granularity maps of compensatory loops evoked by TT, along with deeper understanding of mechanisms underlying drug action, resistance and interactions with lymphoid/myeloid cells, will conceptualize drug combinations able to persistently inhibit tumours, while inducing only limited toxicities.
Goal and specific aims: Addressing resistance to TT, potential synergies and the immune system, we will employ lung cancer models driven by mutant EGFR, HER2, MET or AXL. Phosphoproteomics, transcriptomics and RNA interference, will enable mapping adaptations evoked by specific drugs. Once identified, we will test combinations of interceptors able to inhibit the primary target as well as the emerging, resistance-conferring route(s). Next, we will determine the mechanisms of action of selected interceptors (e.g., apoptosis, immunological cytotoxicity and senescence) as bases for optimising effective combinations. Homo-combinations of antibodies (i.e., antibodies recognising distinct epitopes of a receptor), hetero-combinations targeting distinct signalling and immune receptors, and combinations with PKIs will be examined in animal models.
Significance: More than 30 PKIs and >25 mAbs are approved in oncology, but most are used as monotherapies. Detailed knowledge of adaptation-driven resistance, mechanisms of drug action and immune effectors, will guide the long awaited application of TT combinations in oncology, including lung cancer.
Max ERC Funding
2 488 306 €
Duration
Start date: 2017-10-01, End date: 2022-09-30
Project acronym PhenoSwitch
Project Phenotype switching: plasticity and/or differentiation of stromal cells and their progenitors within the tumour microenvironment regulate tumour fate.
Researcher (PI) YUVAL YECHIEL SHAKED
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Consolidator Grant (CoG), LS7, ERC-2017-COG
Summary The limited success of cancer therapy especially in advanced metastatic disease warrants a reassessment, especially given our limited understanding of the nature of cancer cells and the factors that allow them to proliferate and metastasise. Stromal cells of the tumour microenvironment, including fibroblasts, endothelial, immune, adipose and mesenchymal cells, significantly affect cancer cell characteristics and tumour fate; however, their sometimes dichotomous function in high- and low-aggressive tumours has not been thoroughly investigated. Here, we propose to elucidate the largely unknown role of haematopoietic stem and/or progenitor cells (HSCs) on tumour growth and metastases. We found that such cells reside in the tumour niche predominantly in non-aggressive tumours. We hypothesise that cancer cells trigger the differentiation of HSCs into haematopoietic tumour-supporting stromal cells, thereby inducing a phenotypic and functional switch that skews them towards a tumour-promoting phenotype, hence promoting tumour cell aggressiveness and metastases.
To test our hypothesis, we will use high-throughput technologies to track the lineage, differentiation and commitment of HSCs during tumour progression. Our specific aims are therefore:
(a) To systematically analyse tumour-promoting and tumour-restricting stromal phenotypes at the cellular and molecular levels.
(b) To characterise stromal cell plasticity and the contribution of tumour cells to the phenotype switch.
(c) To determine whether differentiated stromal cells and HSCs in cancer patients can predict clinical outcome.
(d) To screen for molecules that inhibit the tumour-promoting stromal switch.
Blocking the tumour-promoting phenotypic switch and maintaining a pre-mature tumour-restricting stromal microenvironment represent a novel strategy in the fight against cancer. This study will lead to the development of new tools to predict prognosis and pharmacological strategies to restrict tumour growth.
Summary
The limited success of cancer therapy especially in advanced metastatic disease warrants a reassessment, especially given our limited understanding of the nature of cancer cells and the factors that allow them to proliferate and metastasise. Stromal cells of the tumour microenvironment, including fibroblasts, endothelial, immune, adipose and mesenchymal cells, significantly affect cancer cell characteristics and tumour fate; however, their sometimes dichotomous function in high- and low-aggressive tumours has not been thoroughly investigated. Here, we propose to elucidate the largely unknown role of haematopoietic stem and/or progenitor cells (HSCs) on tumour growth and metastases. We found that such cells reside in the tumour niche predominantly in non-aggressive tumours. We hypothesise that cancer cells trigger the differentiation of HSCs into haematopoietic tumour-supporting stromal cells, thereby inducing a phenotypic and functional switch that skews them towards a tumour-promoting phenotype, hence promoting tumour cell aggressiveness and metastases.
To test our hypothesis, we will use high-throughput technologies to track the lineage, differentiation and commitment of HSCs during tumour progression. Our specific aims are therefore:
(a) To systematically analyse tumour-promoting and tumour-restricting stromal phenotypes at the cellular and molecular levels.
(b) To characterise stromal cell plasticity and the contribution of tumour cells to the phenotype switch.
(c) To determine whether differentiated stromal cells and HSCs in cancer patients can predict clinical outcome.
(d) To screen for molecules that inhibit the tumour-promoting stromal switch.
Blocking the tumour-promoting phenotypic switch and maintaining a pre-mature tumour-restricting stromal microenvironment represent a novel strategy in the fight against cancer. This study will lead to the development of new tools to predict prognosis and pharmacological strategies to restrict tumour growth.
Max ERC Funding
1 906 250 €
Duration
Start date: 2018-04-01, End date: 2023-03-31
Project acronym PolyDorm
Project Uncovering the molecular and cellular mechanism of tumor dormancy for the rational design of theranostic nanomedicines
Researcher (PI) Ronit Satchi-Fainaro
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Consolidator Grant (CoG), LS7, ERC-2013-CoG
Summary Tumor progression is dependent on a number of sequential steps, including initial tumor-vascular interactions and recruitment of blood vessels, as well as an established interaction of tumor cells with their surrounding microenvironment. Failure of a microscopic tumor, either primary, recurrent or metastatic, to complete one or more of these early stages may lead to delayed clinical manifestation of the cancer and a state of stable non-progressing disease. Micrometastasis, dormant tumors, and residual tumor cells contribute to the occurrence of relapse, and constitute fundamental clinical manifestations of tumor dormancy that together are responsible for the vast majority of cancer deaths. However, although the tumor dormancy phenomenon has critical implications for early detection and treatment of cancer, its biology and genetic characteristics are poorly understood. We now propose to investigate the molecular and cellular changes in tumor-host interactions that govern tumor dormancy, which may lead to the discovery of novel tumor dormancy targets and provide tools for dormancy-dependent tumor therapy strategies. In order to achieve this goal, we will integrate the following basic and translational approaches: (i) Establishment of mouse models of dormant and fast-growing tumor pairs; (ii) Functional and molecular characterization of dormant versus fast-growing tumors, (iii) Design of dormancy-promoting tailor-made polymer therapeutics delivering a combination of microRNAs with chemotherapies; (iv) Polymer conjugation to a prodrug designed to be activated by specific enzymes overexpressed in tumors, Turning-ON a near infra-red (NIR) fluorescence signal.
When completed, this proposal will shed light on this fundamental cancer biology phenomenon. A better understanding of tumor dormancy and the availability of markers and therapeutic targets will most likely change our perception of tumor progression and, consequently, the way we diagnose and treat the disease.
Summary
Tumor progression is dependent on a number of sequential steps, including initial tumor-vascular interactions and recruitment of blood vessels, as well as an established interaction of tumor cells with their surrounding microenvironment. Failure of a microscopic tumor, either primary, recurrent or metastatic, to complete one or more of these early stages may lead to delayed clinical manifestation of the cancer and a state of stable non-progressing disease. Micrometastasis, dormant tumors, and residual tumor cells contribute to the occurrence of relapse, and constitute fundamental clinical manifestations of tumor dormancy that together are responsible for the vast majority of cancer deaths. However, although the tumor dormancy phenomenon has critical implications for early detection and treatment of cancer, its biology and genetic characteristics are poorly understood. We now propose to investigate the molecular and cellular changes in tumor-host interactions that govern tumor dormancy, which may lead to the discovery of novel tumor dormancy targets and provide tools for dormancy-dependent tumor therapy strategies. In order to achieve this goal, we will integrate the following basic and translational approaches: (i) Establishment of mouse models of dormant and fast-growing tumor pairs; (ii) Functional and molecular characterization of dormant versus fast-growing tumors, (iii) Design of dormancy-promoting tailor-made polymer therapeutics delivering a combination of microRNAs with chemotherapies; (iv) Polymer conjugation to a prodrug designed to be activated by specific enzymes overexpressed in tumors, Turning-ON a near infra-red (NIR) fluorescence signal.
When completed, this proposal will shed light on this fundamental cancer biology phenomenon. A better understanding of tumor dormancy and the availability of markers and therapeutic targets will most likely change our perception of tumor progression and, consequently, the way we diagnose and treat the disease.
Max ERC Funding
2 255 920 €
Duration
Start date: 2014-04-01, End date: 2019-03-31
Project acronym ProstOmics
Project 'Tissue is the issue': a multi-omics approach to improve prostate cancer diagnosis
Researcher (PI) May-Britt Tessem
Host Institution (HI) NORGES TEKNISK-NATURVITENSKAPELIGE UNIVERSITET NTNU
Call Details Starting Grant (StG), LS7, ERC-2017-STG
Summary Overtreatment in prostate cancer (PCa) is a burden for health care economy and for quality of life. Correct diagnosis of early stage PCa is challenging given the limitations of the currently available clinical tools and the biological understanding of PCa. In this inter-disciplinary project, I propose an innovative approach enabling several cutting-edge ‘omics’ technologies (spatial metabolomics, genomics, transcriptomics) as well as histopathology to be performed on the same tissue sample. My goal is to reveal the molecular mechanisms of novel, but also promising metabolite biomarkers (citrate, polyamines, succinate and zinc) and their connection to recurrence, tissue heterogeneity and immune responses in complex human tissues. Such markers can personalize PCa diagnosis, open up new treatment strategies and fundamentally change the view of how to analyze tissue samples in the future. Furthermore, I want to demonstrate that citrate and polyamines are reliable prognostic markers that can be analyzed both in tissue and in patients in vivo by MR spectroscopic imaging. This work is made possible by the availability of high-quality fresh frozen tissue biobanks of prostatectomy biopsies with 5-10 years of follow-up data (N=1000)/slices (N=1000) and targeted in vivo snap-shot biopsies from clinical MR guided procedures (N=100). Among other techniques, I will implement high speed MALDI imaging (RapifleX MALDI TissueTyper) to the multi-omics protocol to study the spatial distribution and provide high resolution metabolic maps for each cell type, and which can be matched to both histopathology and MR Imaging. Multi-disciplinary platforms on large cohorts are needed to explore the clinical potential of the suggested molecular mechanisms. I expect that this ambitious proposal will address the diagnostic challenges of PCa and will further inspire the clinic and scientific community to follow the multi-omics approach within diagnosis and cancer research.
Summary
Overtreatment in prostate cancer (PCa) is a burden for health care economy and for quality of life. Correct diagnosis of early stage PCa is challenging given the limitations of the currently available clinical tools and the biological understanding of PCa. In this inter-disciplinary project, I propose an innovative approach enabling several cutting-edge ‘omics’ technologies (spatial metabolomics, genomics, transcriptomics) as well as histopathology to be performed on the same tissue sample. My goal is to reveal the molecular mechanisms of novel, but also promising metabolite biomarkers (citrate, polyamines, succinate and zinc) and their connection to recurrence, tissue heterogeneity and immune responses in complex human tissues. Such markers can personalize PCa diagnosis, open up new treatment strategies and fundamentally change the view of how to analyze tissue samples in the future. Furthermore, I want to demonstrate that citrate and polyamines are reliable prognostic markers that can be analyzed both in tissue and in patients in vivo by MR spectroscopic imaging. This work is made possible by the availability of high-quality fresh frozen tissue biobanks of prostatectomy biopsies with 5-10 years of follow-up data (N=1000)/slices (N=1000) and targeted in vivo snap-shot biopsies from clinical MR guided procedures (N=100). Among other techniques, I will implement high speed MALDI imaging (RapifleX MALDI TissueTyper) to the multi-omics protocol to study the spatial distribution and provide high resolution metabolic maps for each cell type, and which can be matched to both histopathology and MR Imaging. Multi-disciplinary platforms on large cohorts are needed to explore the clinical potential of the suggested molecular mechanisms. I expect that this ambitious proposal will address the diagnostic challenges of PCa and will further inspire the clinic and scientific community to follow the multi-omics approach within diagnosis and cancer research.
Max ERC Funding
1 500 000 €
Duration
Start date: 2018-02-01, End date: 2023-01-31
Project acronym QSOX1BIOFUNC
Project Frontiers of Oxidative Protein Folding and Assembly: Catalysis of Disulfide Formation Downstream of the Endoplasmic Reticulum
Researcher (PI) Deborah Fass
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS1, ERC-2012-StG_20111109
Summary Quiescin sulfhydryl oxidase (QSOX), which catalyzes disulfide cross-linking in proteins, is up-regulated in many tumor types. QSOX is the only disulfide catalyst to undergo regulated secretion from cells, but the biological role of the enzyme, its substrates, and its mechanistic link to cancer are obscure. In addition to determining the first X-ray crystal structures of QSOXs, we recently discovered that QSOX is required for incorporation of laminins into basement membrane extracellular matrix (ECM). Fibroblasts depleted of QSOX make defective ECM that fails to support adherence and migration of tumor-derived cells in co-culture, but ECM composition and cell migration are restored by supplying recombinant QSOX exogenously. Our observations suggest a role for QSOX in building a microenvironment favorable for tumor cell survival and migration. We are motivated by a number of observations to expand our studies of QSOX enzymes. First, QSOX is ideally suited for single-molecule fluorescence resonance energy transfer experiments, which will shed light on hitherto invisible steps in the QSOX reaction cycle. Second, we have evidence for additional functions of QSOX with intriguing links to lipid metabolism and lipid storage diseases, and we are compelled to discover the pathways involved. Third, the modulation of tumor/stromal interactions is a promising direction for anti-metastatic cancer therapy, so the QSOX inhibitors we are developing may find use in the clinic as well as in the laboratory. Fourth, the complex QSOX expression patterns in developing and adult mammals prompted us to generate a conditional QSOX knockout mouse with which to explore QSOX function in vivo. In consolidating my laboratory, I will continue to rely heavily on my strengths as a structural biologist while pushing forward the frontier of our understanding of oxidative protein folding and assembly in cell and matrix biology, a fascinating subject that spans the Angstrom to the organism.
Summary
Quiescin sulfhydryl oxidase (QSOX), which catalyzes disulfide cross-linking in proteins, is up-regulated in many tumor types. QSOX is the only disulfide catalyst to undergo regulated secretion from cells, but the biological role of the enzyme, its substrates, and its mechanistic link to cancer are obscure. In addition to determining the first X-ray crystal structures of QSOXs, we recently discovered that QSOX is required for incorporation of laminins into basement membrane extracellular matrix (ECM). Fibroblasts depleted of QSOX make defective ECM that fails to support adherence and migration of tumor-derived cells in co-culture, but ECM composition and cell migration are restored by supplying recombinant QSOX exogenously. Our observations suggest a role for QSOX in building a microenvironment favorable for tumor cell survival and migration. We are motivated by a number of observations to expand our studies of QSOX enzymes. First, QSOX is ideally suited for single-molecule fluorescence resonance energy transfer experiments, which will shed light on hitherto invisible steps in the QSOX reaction cycle. Second, we have evidence for additional functions of QSOX with intriguing links to lipid metabolism and lipid storage diseases, and we are compelled to discover the pathways involved. Third, the modulation of tumor/stromal interactions is a promising direction for anti-metastatic cancer therapy, so the QSOX inhibitors we are developing may find use in the clinic as well as in the laboratory. Fourth, the complex QSOX expression patterns in developing and adult mammals prompted us to generate a conditional QSOX knockout mouse with which to explore QSOX function in vivo. In consolidating my laboratory, I will continue to rely heavily on my strengths as a structural biologist while pushing forward the frontier of our understanding of oxidative protein folding and assembly in cell and matrix biology, a fascinating subject that spans the Angstrom to the organism.
Max ERC Funding
1 498 083 €
Duration
Start date: 2012-12-01, End date: 2017-11-30
Project acronym RespMicroFlows
Project Unravelling respiratory microflows in silico and in vitro: novel paths for targeted pulmonary delivery in infants and young children
Researcher (PI) Josue Sznitman
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS7, ERC-2015-STG
Summary Fundamental research on respiratory transport phenomena, quantifying momentum and mass transfer in the lung depths, is overwhelmingly focused on adults. Yet, children are not just miniature adults; their distinct lung structures and heterogeneous ventilation patterns set them aside from their parents. In RespMicroFlows, we will break this cycle and unravel the complex microflows characterizing alveolar airflows in the developing pulmonary acini. Our discoveries will foster ground-breaking transport strategies to tackle two urgent clinical needs that burden infants and young children. The first challenge relates to radically enhancing the delivery and deposition of therapeutics using inhalation aerosols; the second involves targeting liquid bolus installations in deep airways for surfactant replacement therapy.
By developing advanced in silico numerical simulations together with microfluidic in vitro platforms mimicking the pulmonary acinar environment, our efforts will not only deliver a gateway to reliably assess the outcomes of inhaling aerosols and predict deposition patterns in young populations, we will furthermore unravel the fundamentals of liquid bolus transport to achieve optimal surfactant delivery strategies in premature neonates. By recreating cellular alveolar environments that capture underlying physiological functions, our advanced acinus-on-chips will deliver both at true scale and in real time the first robust and reliable in vitro screening platforms of exogenous therapeutic materials in the context of inhaled aerosols and surfactant-laden installations. Combining advanced engineering-driven flow visualization solutions with strong foundations in transport phenomena, fluid dynamics and respiratory physiology, RespMicroFlows will pave the way to a new and unprecedented level in our understanding and quantitative mapping of respiratory flow phenomena and act as catalyst for novel targeted pulmonary drug delivery strategies in young children.
Summary
Fundamental research on respiratory transport phenomena, quantifying momentum and mass transfer in the lung depths, is overwhelmingly focused on adults. Yet, children are not just miniature adults; their distinct lung structures and heterogeneous ventilation patterns set them aside from their parents. In RespMicroFlows, we will break this cycle and unravel the complex microflows characterizing alveolar airflows in the developing pulmonary acini. Our discoveries will foster ground-breaking transport strategies to tackle two urgent clinical needs that burden infants and young children. The first challenge relates to radically enhancing the delivery and deposition of therapeutics using inhalation aerosols; the second involves targeting liquid bolus installations in deep airways for surfactant replacement therapy.
By developing advanced in silico numerical simulations together with microfluidic in vitro platforms mimicking the pulmonary acinar environment, our efforts will not only deliver a gateway to reliably assess the outcomes of inhaling aerosols and predict deposition patterns in young populations, we will furthermore unravel the fundamentals of liquid bolus transport to achieve optimal surfactant delivery strategies in premature neonates. By recreating cellular alveolar environments that capture underlying physiological functions, our advanced acinus-on-chips will deliver both at true scale and in real time the first robust and reliable in vitro screening platforms of exogenous therapeutic materials in the context of inhaled aerosols and surfactant-laden installations. Combining advanced engineering-driven flow visualization solutions with strong foundations in transport phenomena, fluid dynamics and respiratory physiology, RespMicroFlows will pave the way to a new and unprecedented level in our understanding and quantitative mapping of respiratory flow phenomena and act as catalyst for novel targeted pulmonary drug delivery strategies in young children.
Max ERC Funding
1 937 500 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
Project acronym RxmiRcanceR
Project Tumor suppressive microRNAs for cancer therapy
Researcher (PI) Eithan Galun
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS7, ERC-2017-ADG
Summary The challenge for cancer therapy involves hampering the mechanisms by which the normal gene expression machinery is taken over to allow the aberrant appearance of cancer driving genes. I propose exploiting the therapeutic potential of a special class of tumor microRNAs (miRs) that function as natural post-transcriptional tumor suppressive regulators of many genes in key pathways. These anti-cancer effectors represent an inherent organismal property that I propose to augment and thereby translate into a form of systemic anti-cancer therapy. First, focusing on hepatocellular carcinoma (HCC), I shall perform high throughput screening to identify preferred HCC tumor suppressive miRs. Second, I shall search for small molecules capable of elevating the level of those relevant miRs in tumor cells and tissues. Increasing miR expression will potentially also enhance their secretion into the circulation in exosomes thereby suppressing gene expression at remote tissue sites as well. Third, I shall test the potential of these miRs to better target and inhibit the growth of tumor cells both, in culture and in vivo. This unprecedented conceptual strategy should stimulate the organism’s self-healing potential by enhancing inherent anti-tumor mechanisms. This project is built on robust preliminary findings that show promiscuous anti-cancer effects and predictably fewer side effects due to its completely host-based nature, with the administered miR inducer being the only foreign element. Additionally, due to the fact that each miR simultaneously targets a number of molecular pathways as well as multiple steps within a given pathway, it could help to overcome the major problem of cancer therapy – resistance. This strategy of harnessing these efficient and robust miRs and exosomes for delivery of anti-cancer therapeutics may overcome the high-risk challenge involved and enable high gain value.
Summary
The challenge for cancer therapy involves hampering the mechanisms by which the normal gene expression machinery is taken over to allow the aberrant appearance of cancer driving genes. I propose exploiting the therapeutic potential of a special class of tumor microRNAs (miRs) that function as natural post-transcriptional tumor suppressive regulators of many genes in key pathways. These anti-cancer effectors represent an inherent organismal property that I propose to augment and thereby translate into a form of systemic anti-cancer therapy. First, focusing on hepatocellular carcinoma (HCC), I shall perform high throughput screening to identify preferred HCC tumor suppressive miRs. Second, I shall search for small molecules capable of elevating the level of those relevant miRs in tumor cells and tissues. Increasing miR expression will potentially also enhance their secretion into the circulation in exosomes thereby suppressing gene expression at remote tissue sites as well. Third, I shall test the potential of these miRs to better target and inhibit the growth of tumor cells both, in culture and in vivo. This unprecedented conceptual strategy should stimulate the organism’s self-healing potential by enhancing inherent anti-tumor mechanisms. This project is built on robust preliminary findings that show promiscuous anti-cancer effects and predictably fewer side effects due to its completely host-based nature, with the administered miR inducer being the only foreign element. Additionally, due to the fact that each miR simultaneously targets a number of molecular pathways as well as multiple steps within a given pathway, it could help to overcome the major problem of cancer therapy – resistance. This strategy of harnessing these efficient and robust miRs and exosomes for delivery of anti-cancer therapeutics may overcome the high-risk challenge involved and enable high gain value.
Max ERC Funding
2 840 729 €
Duration
Start date: 2018-09-01, End date: 2023-08-31
Project acronym SELECTIONPREDISPOSED
Project Novel Tools for Early Childhood Predisposition to Obesity
Researcher (PI) Pål Rasmus Njølstad
Host Institution (HI) UNIVERSITETET I BERGEN
Call Details Advanced Grant (AdG), LS7, ERC-2011-ADG_20110310
Summary An estimated 22 million children worldwide under age five are overweight. In these children, obesity is a primary indicator for development of type 2 diabetes and possibly cancer. I present a research program, SELECTionPREDISPOSED, to identify novel obesity-risk genes as tools for detection of early childhood obesity making possible a selective prevention program in predisposed children. I will use records and blood samples from children and their parents in the Mother-Child Cohort of Norway, the Health Survey of Nord-Trøndelag and the Norwegian Birth Registry and cross-correlate the databases for genetic research. I hypothesize that children large at birth with enhanced infantile growth may be predisposed to obesity by genetic factors. Obesity-linked genes are likely to include a mix of variants associated with glucose, insulin and fat metabolism and may be identifiable in population studies using biobanks and end-point registries. The state-of-the-art approach is to identify diabetes- or obesity-associated genes in subjects with disease. My approach is to investigate subsets of children with high and low birth weights and BMIs at age six. Using cutting-edge genetic techniques like GWAS, copy-number variation and massive parallel exome and epigenome sequencing I will correlate the genetic information with clinical data in large national end-point registries by a case-control design subsequent replication in large data sets and control for environmental confounders by cross-correlation to the national birth registry. I want to change the field by working with predisposed children in order to influence the ratio between those that may and may not develop obesity and diabetes. In this way my team will develop contextual tools of a groundbreaking nature. This “tool-kit” may make it possible to identify and implement in predisposed children, an early low-cost prevention program to slow down and reverse the development of obesity and prevent diabetes and possibly cancer.
Summary
An estimated 22 million children worldwide under age five are overweight. In these children, obesity is a primary indicator for development of type 2 diabetes and possibly cancer. I present a research program, SELECTionPREDISPOSED, to identify novel obesity-risk genes as tools for detection of early childhood obesity making possible a selective prevention program in predisposed children. I will use records and blood samples from children and their parents in the Mother-Child Cohort of Norway, the Health Survey of Nord-Trøndelag and the Norwegian Birth Registry and cross-correlate the databases for genetic research. I hypothesize that children large at birth with enhanced infantile growth may be predisposed to obesity by genetic factors. Obesity-linked genes are likely to include a mix of variants associated with glucose, insulin and fat metabolism and may be identifiable in population studies using biobanks and end-point registries. The state-of-the-art approach is to identify diabetes- or obesity-associated genes in subjects with disease. My approach is to investigate subsets of children with high and low birth weights and BMIs at age six. Using cutting-edge genetic techniques like GWAS, copy-number variation and massive parallel exome and epigenome sequencing I will correlate the genetic information with clinical data in large national end-point registries by a case-control design subsequent replication in large data sets and control for environmental confounders by cross-correlation to the national birth registry. I want to change the field by working with predisposed children in order to influence the ratio between those that may and may not develop obesity and diabetes. In this way my team will develop contextual tools of a groundbreaking nature. This “tool-kit” may make it possible to identify and implement in predisposed children, an early low-cost prevention program to slow down and reverse the development of obesity and prevent diabetes and possibly cancer.
Max ERC Funding
2 299 549 €
Duration
Start date: 2012-09-01, End date: 2018-08-31
Project acronym SmartCardiacPatch
Project Next-Generation Cardiac Tissue Engineering: Smart Self-Regulating Patches
Researcher (PI) Tal Dvir
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS7, ERC-2014-STG
Summary Ischemic heart disease is a major cause of death in the Western world. There is no sustainable regenerative therapy available at the moment, with cardiac transplantation being the only therapy. However, tissue engineering is envisioned as a true regenerative therapeutic alternative. Despite the incremental improvements no technology is currently available that can provide on-line monitoring and reporting of the engineered tissue performance, and if needed, automatically activate regenerative processes. As one initial step in that direction, we have recently shown on a non-implantable chip-supported level that a sensory system can be integrated with engineered tissues, providing report on cardiac electrical activity.
In this proposal, I plan to expand far beyond the state-of-the-art and develop a conceptually new approach to engineer the next generation of smart implantable cardiac patches. These patches will integrate complex electronics with engineered cardiac tissues to enable on-line monitoring and at the same time self-regulation of the tissue function. Since cardiac performance will be recorded over time, physicians could follow heart regeneration in real-time, providing new means for the disease management.
To achieve this goal I will first develop new porous, stretchable and biocompatible microelectronics enabling electrical activity recording and stimulation. The electronics will interact with an efficient electroactive controlled release system enabling on-demand release of biomolecules. The system will be integrated with a 3D biomaterial scaffold and cardiac cells to compose the microelectronic cardiac patch (microECP). Development of feedback loop software will ensure efficient regulation of the patch’s function over time. Next, we will elucidate the interplay between the electronics, scaffold and cells, and provide a proof-of-principle for the microECP in vitro. Finally, we will investigate the regenerative potential of the system following infarction.
Summary
Ischemic heart disease is a major cause of death in the Western world. There is no sustainable regenerative therapy available at the moment, with cardiac transplantation being the only therapy. However, tissue engineering is envisioned as a true regenerative therapeutic alternative. Despite the incremental improvements no technology is currently available that can provide on-line monitoring and reporting of the engineered tissue performance, and if needed, automatically activate regenerative processes. As one initial step in that direction, we have recently shown on a non-implantable chip-supported level that a sensory system can be integrated with engineered tissues, providing report on cardiac electrical activity.
In this proposal, I plan to expand far beyond the state-of-the-art and develop a conceptually new approach to engineer the next generation of smart implantable cardiac patches. These patches will integrate complex electronics with engineered cardiac tissues to enable on-line monitoring and at the same time self-regulation of the tissue function. Since cardiac performance will be recorded over time, physicians could follow heart regeneration in real-time, providing new means for the disease management.
To achieve this goal I will first develop new porous, stretchable and biocompatible microelectronics enabling electrical activity recording and stimulation. The electronics will interact with an efficient electroactive controlled release system enabling on-demand release of biomolecules. The system will be integrated with a 3D biomaterial scaffold and cardiac cells to compose the microelectronic cardiac patch (microECP). Development of feedback loop software will ensure efficient regulation of the patch’s function over time. Next, we will elucidate the interplay between the electronics, scaffold and cells, and provide a proof-of-principle for the microECP in vitro. Finally, we will investigate the regenerative potential of the system following infarction.
Max ERC Funding
1 499 500 €
Duration
Start date: 2015-07-01, End date: 2021-06-30
Project acronym StopAutoimmunity
Project Recurrent disease in the liver transplant: window to identify and stop gut signals driving autoimmunity
Researcher (PI) Johannes Espolin Roksund HOV
Host Institution (HI) UNIVERSITETET I OSLO
Call Details Starting Grant (StG), LS7, ERC-2018-STG
Summary Autoimmune disease is an increasing health concern. These diseases are strongly associated with altered gut microbiome. When immunosuppression fails there is little to offer in terms of therapy. In this project, I hypothesize that gut signals (microbial factors from the intestine) unaffected by immunosuppression are key drivers of autoimmune diseases. I propose to use recurrent autoimmune disease after organ transplantation as a human disease model to identify and stop these gut signals, providing a novel approach to close the gap between basic microbiome research and patient care in autoimmune diseases.
To identify autoimmunity-related gut signals, I will use patients with primary sclerosing cholangitis (PSC), an inflammatory disease of the bile ducts. PSC is a common indication for liver transplantation, but after transplantation there is high risk of recurrent PSC (rPSC). I recently showed that the PSC gut microbiome has low diversity and identified microbial metabolites associated with severe PSC. Preliminary data show that the post-transplant gut is even less diverse, suggesting that microbial factors drive autoimmunity.
In this project I will identify gut signals by in-depth investigation of gut bacterial genes and circulating metabolites in the blood. The outcome will be diagnostic and prognostic markers overlapping in PSC and rPSC, defined by changes in gut bacterial genes and concentrations of bacterial metabolites in the blood. Next, I will investigate if common drugs or interventions influence the identified autoimmunity-related gut signals. By generating a library of interventions influencing the gut microbiome it will be possible to select promising candidates for pilot treatment trials after liver transplantation.
The outcome of StopAutoimmunity will be gut signals useful as novel biomarkers and treatment targets. These may directly translate into improved patient care but also provide a foundation for understanding the mechanisms of autoimmunity.
Summary
Autoimmune disease is an increasing health concern. These diseases are strongly associated with altered gut microbiome. When immunosuppression fails there is little to offer in terms of therapy. In this project, I hypothesize that gut signals (microbial factors from the intestine) unaffected by immunosuppression are key drivers of autoimmune diseases. I propose to use recurrent autoimmune disease after organ transplantation as a human disease model to identify and stop these gut signals, providing a novel approach to close the gap between basic microbiome research and patient care in autoimmune diseases.
To identify autoimmunity-related gut signals, I will use patients with primary sclerosing cholangitis (PSC), an inflammatory disease of the bile ducts. PSC is a common indication for liver transplantation, but after transplantation there is high risk of recurrent PSC (rPSC). I recently showed that the PSC gut microbiome has low diversity and identified microbial metabolites associated with severe PSC. Preliminary data show that the post-transplant gut is even less diverse, suggesting that microbial factors drive autoimmunity.
In this project I will identify gut signals by in-depth investigation of gut bacterial genes and circulating metabolites in the blood. The outcome will be diagnostic and prognostic markers overlapping in PSC and rPSC, defined by changes in gut bacterial genes and concentrations of bacterial metabolites in the blood. Next, I will investigate if common drugs or interventions influence the identified autoimmunity-related gut signals. By generating a library of interventions influencing the gut microbiome it will be possible to select promising candidates for pilot treatment trials after liver transplantation.
The outcome of StopAutoimmunity will be gut signals useful as novel biomarkers and treatment targets. These may directly translate into improved patient care but also provide a foundation for understanding the mechanisms of autoimmunity.
Max ERC Funding
1 500 000 €
Duration
Start date: 2019-04-01, End date: 2024-03-31
Project acronym SULTENG
Project Protein engineering for the study of detoxification enzymes and hub proteins
Researcher (PI) Amir Aharoni
Host Institution (HI) BEN-GURION UNIVERSITY OF THE NEGEV
Call Details Starting Grant (StG), LS7, ERC-2007-StG
Summary Proteins that exhibit broad specificity play important roles in different biological processes. These proteins include enzymes that catalyse the chemical transformation of many different substrates and proteins that bind to multiple protein partners. We propose to develop and apply novel directed evolution and chemical genetic methodologies for the study of proteins that exhibit broad specificity, with focus on cytosolic sulfotransferases (SULTs), which detoxify a broad range of xeno- and endobiotics, and proliferating cellular nuclear antigen (PCNA), which binds to multiple protein partners to play a central role in DNA replication and repair. SULTs belong to a large family of detoxification enzymes that exhibit broad specificity and relatively poor catalytic efficiency. It is not clear how SULTs can detoxify a variety of different compounds and what constitutes the molecular basis for their broad specificity. Application of directed evolution methodologies will allow us to identify and isolate SULT mutants with improved catalytic efficiency and novel specificity. These mutants will be thoroughly characterised by applying a variety of biochemical and structural methodologies to provide new insights into the broad specificity, catalytic activity and biological functions of SULTs. In parallel, we propose to develop and apply directed evolution methodologies for the study of PCNA. PCNA is a homotrimeric hub protein that forms a DNA sliding clamp to mediate DNA replication and repair by recruitment of a variety of essential proteins to the DNA template. Very little is known about how these multiple binding choices are regulated or about the importance of the different PCNA-protein interactions at different stages of replication. We propose to generate PCNA mutants with new binding activity and novel specificity, followed by thorough in-vitro and in-vivo characterisation, to study the roles of PCNA-protein interactions in DNA replication and repair.
Summary
Proteins that exhibit broad specificity play important roles in different biological processes. These proteins include enzymes that catalyse the chemical transformation of many different substrates and proteins that bind to multiple protein partners. We propose to develop and apply novel directed evolution and chemical genetic methodologies for the study of proteins that exhibit broad specificity, with focus on cytosolic sulfotransferases (SULTs), which detoxify a broad range of xeno- and endobiotics, and proliferating cellular nuclear antigen (PCNA), which binds to multiple protein partners to play a central role in DNA replication and repair. SULTs belong to a large family of detoxification enzymes that exhibit broad specificity and relatively poor catalytic efficiency. It is not clear how SULTs can detoxify a variety of different compounds and what constitutes the molecular basis for their broad specificity. Application of directed evolution methodologies will allow us to identify and isolate SULT mutants with improved catalytic efficiency and novel specificity. These mutants will be thoroughly characterised by applying a variety of biochemical and structural methodologies to provide new insights into the broad specificity, catalytic activity and biological functions of SULTs. In parallel, we propose to develop and apply directed evolution methodologies for the study of PCNA. PCNA is a homotrimeric hub protein that forms a DNA sliding clamp to mediate DNA replication and repair by recruitment of a variety of essential proteins to the DNA template. Very little is known about how these multiple binding choices are regulated or about the importance of the different PCNA-protein interactions at different stages of replication. We propose to generate PCNA mutants with new binding activity and novel specificity, followed by thorough in-vitro and in-vivo characterisation, to study the roles of PCNA-protein interactions in DNA replication and repair.
Max ERC Funding
1 000 000 €
Duration
Start date: 2008-07-01, End date: 2013-06-30
Project acronym SweetAim
Project Selective glycoimmuno-targeting for cancer therapy
Researcher (PI) VERED KARAVANI
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS7, ERC-2016-STG
Summary Immunotherapy recently became an important alternative to conventional treatment regimes, yet cancer remains a universal leading cause of death. Thus, novel cancer theranostic approaches are still much desired. Although altered cell surface glycosylation is one of the hallmarks of cancer, targeting this ‘sweet aim’ for cancer therapy has been elusive, largely due to carbohydrates poor immunogenicity and the low affinity of antibodies against them. A red meat-derived carbohydrate antigen is a novel immunogenic moiety providing a key to unlock the theranostic potential of tumor-associated carbohydrate antigens. This foreign non-human sugar can be acquired only through the diet and subsequently appears on diverse cell surface glycoconjugates as ‘self’, accumulating mostly on carcinomas, and resulting in a polyclonal xeno-autoantibodies response. I have shown that such antibodies have both diagnostic and therapeutic potential, although basic understanding of their specificity and potency is scarce. The primary objective of this proposal is to design a novel personalized cancer therapeutic approach based on xeno-autoantibodies against the dietary sugar antigen. We propose an innovative interdisciplinary approach crossing the boundaries of cancer research, glycosciences, immunology and nanotechnology, with cutting-edge technologies, to design, engineer, screen and fully investigate potent targeting of ‘SweetAim’ moieties. Our discovery line is based on a two-arms platform to generate optimized antibodies for passive/active therapy, together with refined tumor cells through glyco-engineering/reprogramming for unveiling novel theranostics, finally evaluated both in vitro and in vivo. I expect our groundbreaking achievements will lead to promising new clinical tools, particularly for cancer, but also for other chronic inflammation-mediated diseases. Importantly, it will establish fundamental new concepts regarding carbohydrate recognition and response by the immune system.
Summary
Immunotherapy recently became an important alternative to conventional treatment regimes, yet cancer remains a universal leading cause of death. Thus, novel cancer theranostic approaches are still much desired. Although altered cell surface glycosylation is one of the hallmarks of cancer, targeting this ‘sweet aim’ for cancer therapy has been elusive, largely due to carbohydrates poor immunogenicity and the low affinity of antibodies against them. A red meat-derived carbohydrate antigen is a novel immunogenic moiety providing a key to unlock the theranostic potential of tumor-associated carbohydrate antigens. This foreign non-human sugar can be acquired only through the diet and subsequently appears on diverse cell surface glycoconjugates as ‘self’, accumulating mostly on carcinomas, and resulting in a polyclonal xeno-autoantibodies response. I have shown that such antibodies have both diagnostic and therapeutic potential, although basic understanding of their specificity and potency is scarce. The primary objective of this proposal is to design a novel personalized cancer therapeutic approach based on xeno-autoantibodies against the dietary sugar antigen. We propose an innovative interdisciplinary approach crossing the boundaries of cancer research, glycosciences, immunology and nanotechnology, with cutting-edge technologies, to design, engineer, screen and fully investigate potent targeting of ‘SweetAim’ moieties. Our discovery line is based on a two-arms platform to generate optimized antibodies for passive/active therapy, together with refined tumor cells through glyco-engineering/reprogramming for unveiling novel theranostics, finally evaluated both in vitro and in vivo. I expect our groundbreaking achievements will lead to promising new clinical tools, particularly for cancer, but also for other chronic inflammation-mediated diseases. Importantly, it will establish fundamental new concepts regarding carbohydrate recognition and response by the immune system.
Max ERC Funding
1 479 995 €
Duration
Start date: 2016-12-01, End date: 2021-11-30
Project acronym TARGETING_CANCER
Project Eradication of tumors by targeting dsRNA selectively to cancer cells and recruitment of the innate immune system
Researcher (PI) Alexander Levitzki
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS7, ERC-2009-AdG
Summary We have recently shown that EGFR over-expressing tumors can be eradicated by an EGFR homing chemical vector, carrying dsRNA. The vector is PolyInosine/Cytosine (PolyIC) bound to Polyethleneimine-Polyethyleneglycol-EGF (PEI-PEG-EGF, PPE). We have shown that even tumors in which up to 50% of cells do not express EGFR are eradicated, due to the strong tumor-localized bystander effects, which involve the innate immune system. Using this EGFR homing vector we have been able to eradicate EGFR overexpressing tumors by either local or systemic application. Since the success of this strategy seems to be due to the strong bystander effects induced by the internalized PolyIC it is likely that heterogeneous tumors, in which only a portion of the cells harbor the targeted receptor, will be eradicated too, as shown in our preliminary studies (PloS Med, 2006). This strategy actually targets the innate immune system to the tumor. We propose to establish tumors in which decreasing portions of cells over-express EGFR and determine the lowest number of EGFR over-expressing cells that can yield tumor eradication by the lowest dose of PolyIC/PPE. The principle behind the success of the Trojan horse approach is that the targeting moiety, EGF, is tethered to the other components of the vector in such a way that it retains its native EGFR binding properties and its ability to internalize with the receptor. The composition of the vector is such that the ligand EGF can be replaced by any other ligand, if the appropriate coupling conditions are used, retaining the ability of the ligand to bind to the target protein and internalize with it. We propose to replace EGF by a number of other ligands, such PSMA binding ligand (targeting prostate cancer) and Her-2 affibodies. Although only a fraction of women who over-express Her-2 respond to Herceptin, it is likely that they will respond to PolyIC/PP-Her-2 affibody.
Summary
We have recently shown that EGFR over-expressing tumors can be eradicated by an EGFR homing chemical vector, carrying dsRNA. The vector is PolyInosine/Cytosine (PolyIC) bound to Polyethleneimine-Polyethyleneglycol-EGF (PEI-PEG-EGF, PPE). We have shown that even tumors in which up to 50% of cells do not express EGFR are eradicated, due to the strong tumor-localized bystander effects, which involve the innate immune system. Using this EGFR homing vector we have been able to eradicate EGFR overexpressing tumors by either local or systemic application. Since the success of this strategy seems to be due to the strong bystander effects induced by the internalized PolyIC it is likely that heterogeneous tumors, in which only a portion of the cells harbor the targeted receptor, will be eradicated too, as shown in our preliminary studies (PloS Med, 2006). This strategy actually targets the innate immune system to the tumor. We propose to establish tumors in which decreasing portions of cells over-express EGFR and determine the lowest number of EGFR over-expressing cells that can yield tumor eradication by the lowest dose of PolyIC/PPE. The principle behind the success of the Trojan horse approach is that the targeting moiety, EGF, is tethered to the other components of the vector in such a way that it retains its native EGFR binding properties and its ability to internalize with the receptor. The composition of the vector is such that the ligand EGF can be replaced by any other ligand, if the appropriate coupling conditions are used, retaining the ability of the ligand to bind to the target protein and internalize with it. We propose to replace EGF by a number of other ligands, such PSMA binding ligand (targeting prostate cancer) and Her-2 affibodies. Although only a fraction of women who over-express Her-2 respond to Herceptin, it is likely that they will respond to PolyIC/PP-Her-2 affibody.
Max ERC Funding
2 054 340 €
Duration
Start date: 2010-02-01, End date: 2015-01-31
Project acronym TICE
Project TRANSCRIPTOMICS IN CANCER EPIDEMIOLOGY
Researcher (PI) Eiliv Lund
Host Institution (HI) UNIVERSITETET I TROMSOE - NORGES ARKTISKE UNIVERSITET
Call Details Advanced Grant (AdG), LS7, ERC-2008-AdG
Summary NOWAC is the first prospective study with a globolomic design. This is an extension of the current cohort study with its questionnaire information and biological material for analysis of biomarkers, proteomics and single nucleotide polymorphisms (SNPs). The design of NOWAC adds biological material for analysis of the transcriptome in prospectively collected buffered peripheral blood samples, the postgenome biobank. Further, both peripheral blood and tumor tissue are collected from breast cancer patients diagnosed within the cohort together with matched controls. The latter biological material gives a new multidimensional design with a unique biological material at the end-point. The transcriptomic analysis will include both mRNA and miRNA as new technology (microarray and massive parallel sequencing) allows large scale studies. miRNAs could be promising markers for pathways analysis related to the carcinogenic process and for diagnosis and screening tests of breast cancer. These high-troughput technologies have analyses challenges both in bioinformatics and biostatistics therefore success depends on the development of new analytical strategies.This novel design is the observational counterpart to systems biology, or systems epidemiology. Systems epidemiology will seek to understand biological processes by integrating observational derived pathways information into the current prospective design. A true interdisciplinary approach has been implemented. The upside is the potential for an improved understanding of causality in epidemiology by opening up for quantification of traditional criteria of biological plausibility in a more complete biological model. The postgenome biobank with 50 000 participants out of the 172 000 participants in NOWAC and its unique national design and richness of biological material makes it a very strong case for interdisciplinary collaboration based on a population-based study representative of the real and complex lifestyle environment.
Summary
NOWAC is the first prospective study with a globolomic design. This is an extension of the current cohort study with its questionnaire information and biological material for analysis of biomarkers, proteomics and single nucleotide polymorphisms (SNPs). The design of NOWAC adds biological material for analysis of the transcriptome in prospectively collected buffered peripheral blood samples, the postgenome biobank. Further, both peripheral blood and tumor tissue are collected from breast cancer patients diagnosed within the cohort together with matched controls. The latter biological material gives a new multidimensional design with a unique biological material at the end-point. The transcriptomic analysis will include both mRNA and miRNA as new technology (microarray and massive parallel sequencing) allows large scale studies. miRNAs could be promising markers for pathways analysis related to the carcinogenic process and for diagnosis and screening tests of breast cancer. These high-troughput technologies have analyses challenges both in bioinformatics and biostatistics therefore success depends on the development of new analytical strategies.This novel design is the observational counterpart to systems biology, or systems epidemiology. Systems epidemiology will seek to understand biological processes by integrating observational derived pathways information into the current prospective design. A true interdisciplinary approach has been implemented. The upside is the potential for an improved understanding of causality in epidemiology by opening up for quantification of traditional criteria of biological plausibility in a more complete biological model. The postgenome biobank with 50 000 participants out of the 172 000 participants in NOWAC and its unique national design and richness of biological material makes it a very strong case for interdisciplinary collaboration based on a population-based study representative of the real and complex lifestyle environment.
Max ERC Funding
2 300 000 €
Duration
Start date: 2009-01-01, End date: 2014-06-30