Project acronym 3Ps
Project 3Ps
Plastic-Antibodies, Plasmonics and Photovoltaic-Cells: on-site screening of cancer biomarkers made possible
Researcher (PI) Maria Goreti Ferreira Sales
Host Institution (HI) INSTITUTO SUPERIOR DE ENGENHARIA DO PORTO
Call Details Starting Grant (StG), LS7, ERC-2012-StG_20111109
Summary This project presents a new concept for the detection, diagnosis and monitoring of cancer biomarker patterns in point-of-care. The device under development will make use of the selectivity of the plastic antibodies as sensing materials and the interference they will play on the normal operation of a photovoltaic cell.
Plastic antibodies will be designed by surface imprinting procedures. Self-assembled monolayer and molecular imprinting techniques will be merged in this process because they allow the self-assembly of nanostructured materials on a “bottom-up” nanofabrication approach. A dye-sensitized solar cell will be used as photovoltaic cell. It includes a liquid interface in the cell circuit, which allows the introduction of the sample (also in liquid phase) without disturbing the normal cell operation. Furthermore, it works well with rather low cost materials and requires mild and easy processing conditions. The cell will be equipped with plasmonic structures to enhance light absorption and cell efficiency.
The device under development will be easily operated by any clinician or patient. It will require ambient light and a regular multimeter. Eye detection will be also tried out.
Summary
This project presents a new concept for the detection, diagnosis and monitoring of cancer biomarker patterns in point-of-care. The device under development will make use of the selectivity of the plastic antibodies as sensing materials and the interference they will play on the normal operation of a photovoltaic cell.
Plastic antibodies will be designed by surface imprinting procedures. Self-assembled monolayer and molecular imprinting techniques will be merged in this process because they allow the self-assembly of nanostructured materials on a “bottom-up” nanofabrication approach. A dye-sensitized solar cell will be used as photovoltaic cell. It includes a liquid interface in the cell circuit, which allows the introduction of the sample (also in liquid phase) without disturbing the normal cell operation. Furthermore, it works well with rather low cost materials and requires mild and easy processing conditions. The cell will be equipped with plasmonic structures to enhance light absorption and cell efficiency.
The device under development will be easily operated by any clinician or patient. It will require ambient light and a regular multimeter. Eye detection will be also tried out.
Max ERC Funding
998 584 €
Duration
Start date: 2013-02-01, End date: 2018-01-31
Project acronym AcetyLys
Project Unravelling the role of lysine acetylation in the regulation of glycolysis in cancer cells through the development of synthetic biology-based tools
Researcher (PI) Eyal Arbely
Host Institution (HI) BEN-GURION UNIVERSITY OF THE NEGEV
Call Details Starting Grant (StG), LS9, ERC-2015-STG
Summary Synthetic biology is an emerging discipline that offers powerful tools to control and manipulate fundamental processes in living matter. We propose to develop and apply such tools to modify the genetic code of cultured mammalian cells and bacteria with the aim to study the role of lysine acetylation in the regulation of metabolism and in cancer development. Thousands of lysine acetylation sites were recently discovered on non-histone proteins, suggesting that acetylation is a widespread and evolutionarily conserved post translational modification, similar in scope to phosphorylation and ubiquitination. Specifically, it has been found that most of the enzymes of metabolic processes—including glycolysis—are acetylated, implying that acetylation is key regulator of cellular metabolism in general and in glycolysis in particular. The regulation of metabolic pathways is of particular importance to cancer research, as misregulation of metabolic pathways, especially upregulation of glycolysis, is common to most transformed cells and is now considered a new hallmark of cancer. These data raise an immediate question: what is the role of acetylation in the regulation of glycolysis and in the metabolic reprogramming of cancer cells? While current methods rely on mutational analyses, we will genetically encode the incorporation of acetylated lysine and directly measure the functional role of each acetylation site in cancerous and non-cancerous cell lines. Using this methodology, we will study the structural and functional implications of all the acetylation sites in glycolytic enzymes. We will also decipher the mechanism by which acetylation is regulated by deacetylases and answer a long standing question – how 18 deacetylases recognise their substrates among thousands of acetylated proteins? The developed methodologies can be applied to a wide range of protein families known to be acetylated, thereby making this study relevant to diverse research fields.
Summary
Synthetic biology is an emerging discipline that offers powerful tools to control and manipulate fundamental processes in living matter. We propose to develop and apply such tools to modify the genetic code of cultured mammalian cells and bacteria with the aim to study the role of lysine acetylation in the regulation of metabolism and in cancer development. Thousands of lysine acetylation sites were recently discovered on non-histone proteins, suggesting that acetylation is a widespread and evolutionarily conserved post translational modification, similar in scope to phosphorylation and ubiquitination. Specifically, it has been found that most of the enzymes of metabolic processes—including glycolysis—are acetylated, implying that acetylation is key regulator of cellular metabolism in general and in glycolysis in particular. The regulation of metabolic pathways is of particular importance to cancer research, as misregulation of metabolic pathways, especially upregulation of glycolysis, is common to most transformed cells and is now considered a new hallmark of cancer. These data raise an immediate question: what is the role of acetylation in the regulation of glycolysis and in the metabolic reprogramming of cancer cells? While current methods rely on mutational analyses, we will genetically encode the incorporation of acetylated lysine and directly measure the functional role of each acetylation site in cancerous and non-cancerous cell lines. Using this methodology, we will study the structural and functional implications of all the acetylation sites in glycolytic enzymes. We will also decipher the mechanism by which acetylation is regulated by deacetylases and answer a long standing question – how 18 deacetylases recognise their substrates among thousands of acetylated proteins? The developed methodologies can be applied to a wide range of protein families known to be acetylated, thereby making this study relevant to diverse research fields.
Max ERC Funding
1 499 375 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym AXIAL.EC
Project PRINCIPLES OF AXIAL POLARITY-DRIVEN VASCULAR PATTERNING
Researcher (PI) Claudio Franco
Host Institution (HI) INSTITUTO DE MEDICINA MOLECULAR JOAO LOBO ANTUNES
Call Details Starting Grant (StG), LS4, ERC-2015-STG
Summary The formation of a functional patterned vascular network is essential for development, tissue growth and organ physiology. Several human vascular disorders arise from the mis-patterning of blood vessels, such as arteriovenous malformations, aneurysms and diabetic retinopathy. Although blood flow is recognised as a stimulus for vascular patterning, very little is known about the molecular mechanisms that regulate endothelial cell behaviour in response to flow and promote vascular patterning.
Recently, we uncovered that endothelial cells migrate extensively in the immature vascular network, and that endothelial cells polarise against the blood flow direction. Here, we put forward the hypothesis that vascular patterning is dependent on the polarisation and migration of endothelial cells against the flow direction, in a continuous flux of cells going from low-shear stress to high-shear stress regions. We will establish new reporter mouse lines to observe and manipulate endothelial polarity in vivo in order to investigate how polarisation and coordination of endothelial cells movements are orchestrated to generate vascular patterning. We will manipulate cell polarity using mouse models to understand the importance of cell polarisation in vascular patterning. Also, using a unique zebrafish line allowing analysis of endothelial cell polarity, we will perform a screen to identify novel regulators of vascular patterning. Finally, we will explore the hypothesis that defective flow-dependent endothelial polarisation underlies arteriovenous malformations using two genetic models.
This integrative approach, based on high-resolution imaging and unique experimental models, will provide a unifying model defining the cellular and molecular principles involved in vascular patterning. Given the physiological relevance of vascular patterning in health and disease, this research plan will set the basis for the development of novel clinical therapies targeting vascular disorders.
Summary
The formation of a functional patterned vascular network is essential for development, tissue growth and organ physiology. Several human vascular disorders arise from the mis-patterning of blood vessels, such as arteriovenous malformations, aneurysms and diabetic retinopathy. Although blood flow is recognised as a stimulus for vascular patterning, very little is known about the molecular mechanisms that regulate endothelial cell behaviour in response to flow and promote vascular patterning.
Recently, we uncovered that endothelial cells migrate extensively in the immature vascular network, and that endothelial cells polarise against the blood flow direction. Here, we put forward the hypothesis that vascular patterning is dependent on the polarisation and migration of endothelial cells against the flow direction, in a continuous flux of cells going from low-shear stress to high-shear stress regions. We will establish new reporter mouse lines to observe and manipulate endothelial polarity in vivo in order to investigate how polarisation and coordination of endothelial cells movements are orchestrated to generate vascular patterning. We will manipulate cell polarity using mouse models to understand the importance of cell polarisation in vascular patterning. Also, using a unique zebrafish line allowing analysis of endothelial cell polarity, we will perform a screen to identify novel regulators of vascular patterning. Finally, we will explore the hypothesis that defective flow-dependent endothelial polarisation underlies arteriovenous malformations using two genetic models.
This integrative approach, based on high-resolution imaging and unique experimental models, will provide a unifying model defining the cellular and molecular principles involved in vascular patterning. Given the physiological relevance of vascular patterning in health and disease, this research plan will set the basis for the development of novel clinical therapies targeting vascular disorders.
Max ERC Funding
1 618 750 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym BACNK
Project Recognition of bacteria by NK cells
Researcher (PI) Ofer Mandelboim
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS6, ERC-2012-ADG_20120314
Summary NK cells that are well known by their ability to recognize and eliminate virus infected and tumor cells were also implicated in the defence against bacteria. However, the recognition of bacteria by NK cells is only poorly understood. we do not know how bacteria are recognized and the functional consequences of such recognition are also weakly understood. In the current proposal we aimed at determining the “NK cell receptor-bacterial interactome”. We will examine the hypothesis that NK inhibitory and activating receptors are directly involved in bacterial recognition. This ground breaking hypothesis is based on our preliminary results in which we show that several NK cell receptors directly recognize various bacterial strains as well as on a few other publications. We will generate various mice knockouts for NCR1 (a major NK killer receptor) and determine their microbiota to understand the physiological function of NCR1 and whether certain bacterial strains affects its activity. We will use different human and mouse NK killer and inhibitory receptors fused to IgG1 to pull-down bacteria from saliva and fecal samples and then use 16S rRNA analysis and next generation sequencing to determine the nature of the bacteria species isolated. We will identify the bacterial ligands that are recognized by the relevant NK cell receptors, using bacterial random transposon insertion mutagenesis approach. We will end this research with functional assays. In the wake of the emerging threat of bacterial drug resistance and the involvement of bacteria in the pathogenesis of many different chronic diseases and in shaping the immune response, the completion of this study will open a new field of research; the direct recognition of bacteria by NK cell receptors.
Summary
NK cells that are well known by their ability to recognize and eliminate virus infected and tumor cells were also implicated in the defence against bacteria. However, the recognition of bacteria by NK cells is only poorly understood. we do not know how bacteria are recognized and the functional consequences of such recognition are also weakly understood. In the current proposal we aimed at determining the “NK cell receptor-bacterial interactome”. We will examine the hypothesis that NK inhibitory and activating receptors are directly involved in bacterial recognition. This ground breaking hypothesis is based on our preliminary results in which we show that several NK cell receptors directly recognize various bacterial strains as well as on a few other publications. We will generate various mice knockouts for NCR1 (a major NK killer receptor) and determine their microbiota to understand the physiological function of NCR1 and whether certain bacterial strains affects its activity. We will use different human and mouse NK killer and inhibitory receptors fused to IgG1 to pull-down bacteria from saliva and fecal samples and then use 16S rRNA analysis and next generation sequencing to determine the nature of the bacteria species isolated. We will identify the bacterial ligands that are recognized by the relevant NK cell receptors, using bacterial random transposon insertion mutagenesis approach. We will end this research with functional assays. In the wake of the emerging threat of bacterial drug resistance and the involvement of bacteria in the pathogenesis of many different chronic diseases and in shaping the immune response, the completion of this study will open a new field of research; the direct recognition of bacteria by NK cell receptors.
Max ERC Funding
2 499 800 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
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 BISON
Project Bio-Inspired Self-Assembled Supramolecular Organic Nanostructures
Researcher (PI) Ehud Gazit
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Advanced Grant (AdG), LS9, ERC-2015-AdG
Summary Peptide building blocks serve as very attractive bio-inspired elements in nanotechnology owing to their controlled self-assembly, inherent biocompatibility, chemical versatility, biological recognition abilities and facile synthesis. We have demonstrated the ability of remarkably simple aromatic peptides to form well-ordered nanostructures of exceptional physical properties. By taking inspiration from the minimal recognition modules used by nature to mediate coordinated processes of self-assembly, we have developed building blocks that form well-ordered nanostructures. The compact design of the building blocks, and therefore, the unique structural organization, resulted in metallic-like Young's modulus, blue luminescence due to quantum confinement, and notable piezoelectric properties. The goal of this proposal is to develop two new fronts for bio-inspired building block repertoire along with co-assembly to provide new avenues for organic nanotechnology. This will combine our vast experience in the assembly of aromatic peptides together with additional structural modules from nature. The new entities will be developed by exploiting the design principles of small aromatic building blocks to arrive at the smallest possible module that form super helical assembly based on the coiled coil motifs and establishing peptide nucleic acids based systems to combine the worlds of peptide and DNA nanotechnologies. The proposed research will combine extensive design and synthesis effort to provide a very diverse collection of novel buildings blocks and determination of their self-assembly process, followed by broad chemical, physical, and biological characterization of the nanostructures. Furthermore, effort will be made to establish supramolecular co-polymer systems to extend the morphological control of the assembly process. The result of the project will be a large and defined collection of novel chemical entities that will help reshape the field of bioorganic nanotechnology.
Summary
Peptide building blocks serve as very attractive bio-inspired elements in nanotechnology owing to their controlled self-assembly, inherent biocompatibility, chemical versatility, biological recognition abilities and facile synthesis. We have demonstrated the ability of remarkably simple aromatic peptides to form well-ordered nanostructures of exceptional physical properties. By taking inspiration from the minimal recognition modules used by nature to mediate coordinated processes of self-assembly, we have developed building blocks that form well-ordered nanostructures. The compact design of the building blocks, and therefore, the unique structural organization, resulted in metallic-like Young's modulus, blue luminescence due to quantum confinement, and notable piezoelectric properties. The goal of this proposal is to develop two new fronts for bio-inspired building block repertoire along with co-assembly to provide new avenues for organic nanotechnology. This will combine our vast experience in the assembly of aromatic peptides together with additional structural modules from nature. The new entities will be developed by exploiting the design principles of small aromatic building blocks to arrive at the smallest possible module that form super helical assembly based on the coiled coil motifs and establishing peptide nucleic acids based systems to combine the worlds of peptide and DNA nanotechnologies. The proposed research will combine extensive design and synthesis effort to provide a very diverse collection of novel buildings blocks and determination of their self-assembly process, followed by broad chemical, physical, and biological characterization of the nanostructures. Furthermore, effort will be made to establish supramolecular co-polymer systems to extend the morphological control of the assembly process. The result of the project will be a large and defined collection of novel chemical entities that will help reshape the field of bioorganic nanotechnology.
Max ERC Funding
3 003 125 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
Project acronym BRAINVISIONREHAB
Project ‘Seeing’ with the ears, hands and bionic eyes: from theories about brain organization to visual rehabilitation
Researcher (PI) Amir Amedi
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS5, ERC-2012-StG_20111109
Summary My lab's work ranges from basic science, querying brain plasticity and sensory integration, to technological developments, allowing the blind to be more independent and even “see” using sounds and touch similar to bats and dolphins (a.k.a. Sensory Substitution Devices, SSDs), and back to applying these devices in research. We propose that, with proper training, any brain area or network can change the type of sensory input it uses to retrieve behaviorally task-relevant information within a matter of days. If this is true, it can have far reaching implications also for clinical rehabilitation. To achieve this, we are developing several innovative SSDs which encode the most crucial aspects of vision and increase their accessibility the blind, along with targeted, structured training protocols both in virtual environments and in real life. For instance, the “EyeMusic”, encodes colored complex images using pleasant musical scales and instruments, and the “EyeCane”, a palm-size cane, which encodes distance and depth in several directions accurately and efficiently. We provide preliminary but compelling evidence that following such training, SSDs can enable almost blind to recognize daily objects, colors, faces and facial expressions, read street signs, and aiding mobility and navigation. SSDs can also be used in conjunction with (any) invasive approach for visual rehabilitation. We are developing a novel hybrid Visual Rehabilitation Device which combines SSD and bionic eyes. In this set up, the SSDs is used in training the brain to “see” prior to surgery, in providing explanatory signal after surgery and in augmenting the capabilities of the bionic-eyes using information arriving from the same image. We will chart the dynamics of the plastic changes in the brain by performing unprecedented longitudinal Neuroimaging, Electrophysiological and Neurodisruptive approaches while individuals learn to ‘see’ using each of the visual rehabilitation approaches suggested here.
Summary
My lab's work ranges from basic science, querying brain plasticity and sensory integration, to technological developments, allowing the blind to be more independent and even “see” using sounds and touch similar to bats and dolphins (a.k.a. Sensory Substitution Devices, SSDs), and back to applying these devices in research. We propose that, with proper training, any brain area or network can change the type of sensory input it uses to retrieve behaviorally task-relevant information within a matter of days. If this is true, it can have far reaching implications also for clinical rehabilitation. To achieve this, we are developing several innovative SSDs which encode the most crucial aspects of vision and increase their accessibility the blind, along with targeted, structured training protocols both in virtual environments and in real life. For instance, the “EyeMusic”, encodes colored complex images using pleasant musical scales and instruments, and the “EyeCane”, a palm-size cane, which encodes distance and depth in several directions accurately and efficiently. We provide preliminary but compelling evidence that following such training, SSDs can enable almost blind to recognize daily objects, colors, faces and facial expressions, read street signs, and aiding mobility and navigation. SSDs can also be used in conjunction with (any) invasive approach for visual rehabilitation. We are developing a novel hybrid Visual Rehabilitation Device which combines SSD and bionic eyes. In this set up, the SSDs is used in training the brain to “see” prior to surgery, in providing explanatory signal after surgery and in augmenting the capabilities of the bionic-eyes using information arriving from the same image. We will chart the dynamics of the plastic changes in the brain by performing unprecedented longitudinal Neuroimaging, Electrophysiological and Neurodisruptive approaches while individuals learn to ‘see’ using each of the visual rehabilitation approaches suggested here.
Max ERC Funding
1 499 900 €
Duration
Start date: 2013-09-01, End date: 2018-08-31
Project acronym CaNANObinoids
Project From Peripheralized to Cell- and Organelle-Targeted Medicine: The 3rd Generation of Cannabinoid-1 Receptor Antagonists for the Treatment of Chronic Kidney Disease
Researcher (PI) Yossef Tam
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS4, ERC-2015-STG
Summary Clinical experience with globally-acting cannabinoid-1 receptor (CB1R) antagonists revealed the benefits of blocking CB1Rs for the treatment of obesity and diabetes. However, their use is hampered by increased CNS-mediated side effects. Recently, I have demonstrated that peripherally-restricted CB1R antagonists have the potential to treat the metabolic syndrome without eliciting these adverse effects. While these results are promising and are currently being developed into the clinic, our ability to rationally design CB1R blockers that would target a diseased organ is limited.
The current proposal aims to develop and test cell- and organelle-specific CB1R antagonists. To establish this paradigm, I will focus our interest on the kidney, since chronic kidney disease (CKD) is the leading cause of increased morbidity and mortality of patients with diabetes. Our first goal will be to characterize the obligatory role of the renal proximal tubular CB1R in the pathogenesis of diabetic renal complications. Next, we will attempt to link renal proximal CB1R with diabetic mitochondrial dysfunction. Finally, we will develop proximal tubular (cell-specific) and mitochondrial (organelle-specific) CB1R blockers and test their effectiveness in treating CKD. To that end, we will encapsulate CB1R blockers into biocompatible polymeric nanoparticles that will serve as targeted drug delivery systems, via their conjugation to targeting ligands.
The implications of this work are far reaching as they will (i) point to renal proximal tubule CB1R as a novel target for CKD; (ii) identify mitochondrial CB1R as a new player in the regulation of proximal tubular cell function, and (iii) eventually become the drug-of-choice in treating diabetic CKD and its comorbidities. Moreover, this work will lead to the development of a novel organ-specific drug delivery system for CB1R blockers, which could be then exploited in other tissues affected by obesity, diabetes and the metabolic syndrome.
Summary
Clinical experience with globally-acting cannabinoid-1 receptor (CB1R) antagonists revealed the benefits of blocking CB1Rs for the treatment of obesity and diabetes. However, their use is hampered by increased CNS-mediated side effects. Recently, I have demonstrated that peripherally-restricted CB1R antagonists have the potential to treat the metabolic syndrome without eliciting these adverse effects. While these results are promising and are currently being developed into the clinic, our ability to rationally design CB1R blockers that would target a diseased organ is limited.
The current proposal aims to develop and test cell- and organelle-specific CB1R antagonists. To establish this paradigm, I will focus our interest on the kidney, since chronic kidney disease (CKD) is the leading cause of increased morbidity and mortality of patients with diabetes. Our first goal will be to characterize the obligatory role of the renal proximal tubular CB1R in the pathogenesis of diabetic renal complications. Next, we will attempt to link renal proximal CB1R with diabetic mitochondrial dysfunction. Finally, we will develop proximal tubular (cell-specific) and mitochondrial (organelle-specific) CB1R blockers and test their effectiveness in treating CKD. To that end, we will encapsulate CB1R blockers into biocompatible polymeric nanoparticles that will serve as targeted drug delivery systems, via their conjugation to targeting ligands.
The implications of this work are far reaching as they will (i) point to renal proximal tubule CB1R as a novel target for CKD; (ii) identify mitochondrial CB1R as a new player in the regulation of proximal tubular cell function, and (iii) eventually become the drug-of-choice in treating diabetic CKD and its comorbidities. Moreover, this work will lead to the development of a novel organ-specific drug delivery system for CB1R blockers, which could be then exploited in other tissues affected by obesity, diabetes and the metabolic syndrome.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-04-01, End date: 2021-03-31
Project acronym CentrioleBirthDeath
Project Mechanism of centriole inheritance and maintenance
Researcher (PI) Monica BETTENCOURT CARVALHO DIAS
Host Institution (HI) FUNDACAO CALOUSTE GULBENKIAN
Call Details Consolidator Grant (CoG), LS3, ERC-2015-CoG
Summary Centrioles assemble centrosomes and cilia/flagella, critical structures for cell division, polarity, motility and signalling, which are often deregulated in human disease. Centriole inheritance, in particular the preservation of their copy number and position in the cell is critical in many eukaryotes. I propose to investigate, in an integrative and quantitative way, how centrioles are formed in the right numbers at the right time and place, and how they are maintained to ensure their function and inheritance. We first ask how centrioles guide their own assembly position and centriole copy number. Our recent work highlighted several properties of the system, including positive and negative feedbacks and spatial cues. We explore critical hypotheses through a combination of biochemistry, quantitative live cell microscopy and computational modelling. We then ask how the centrosome and the cell cycle are both coordinated. We recently identified the triggering event in centriole biogenesis and how its regulation is akin to cell cycle control of DNA replication and centromere assembly. We will explore new hypotheses to understand how assembly time is coupled to the cell cycle. Lastly, we ask how centriole maintenance is regulated. By studying centriole disappearance in the female germline we uncovered that centrioles need to be actively maintained by their surrounding matrix. We propose to investigate how that matrix provides stability to the centrioles, whether this is differently regulated in different cell types and the possible consequences of its misregulation for the organism (infertility and ciliopathy-like symptoms). We will take advantage of several experimental systems (in silico, ex-vivo, flies and human cells), tailoring the assay to the question and allowing for comparisons across experimental systems to provide a deeper understanding of the process and its regulation.
Summary
Centrioles assemble centrosomes and cilia/flagella, critical structures for cell division, polarity, motility and signalling, which are often deregulated in human disease. Centriole inheritance, in particular the preservation of their copy number and position in the cell is critical in many eukaryotes. I propose to investigate, in an integrative and quantitative way, how centrioles are formed in the right numbers at the right time and place, and how they are maintained to ensure their function and inheritance. We first ask how centrioles guide their own assembly position and centriole copy number. Our recent work highlighted several properties of the system, including positive and negative feedbacks and spatial cues. We explore critical hypotheses through a combination of biochemistry, quantitative live cell microscopy and computational modelling. We then ask how the centrosome and the cell cycle are both coordinated. We recently identified the triggering event in centriole biogenesis and how its regulation is akin to cell cycle control of DNA replication and centromere assembly. We will explore new hypotheses to understand how assembly time is coupled to the cell cycle. Lastly, we ask how centriole maintenance is regulated. By studying centriole disappearance in the female germline we uncovered that centrioles need to be actively maintained by their surrounding matrix. We propose to investigate how that matrix provides stability to the centrioles, whether this is differently regulated in different cell types and the possible consequences of its misregulation for the organism (infertility and ciliopathy-like symptoms). We will take advantage of several experimental systems (in silico, ex-vivo, flies and human cells), tailoring the assay to the question and allowing for comparisons across experimental systems to provide a deeper understanding of the process and its regulation.
Max ERC Funding
2 000 000 €
Duration
Start date: 2017-01-01, End date: 2021-12-31
Project acronym CHOLINOMIRS
Project CholinomiRs: MicroRNA Regulators of Cholinergic Signalling in the Neuro-Immune Interface
Researcher (PI) Hermona Soreq
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS5, ERC-2012-ADG_20120314
Summary "Communication between the nervous and the immune system is pivotal for maintaining homeostasis and ensuring rapid and efficient reaction to stress and infection insults. The emergence of microRNAs (miRs) as regulators of gene expression and of acetylcholine (ACh) signalling as regulator of anxiety and inflammation provides a model for studying this interaction. My hypothesis is that 1) a specific sub-group of miRs, designated ""CholinomiRs"", may silence multiple target genes in the neuro-immune interface; 2) these miRs compete with each other on the interaction with their targets, and 3) mutations interfering with miR binding lead to inherited susceptibility to anxiety and inflammation disorders by modifying these interactions. Our preliminary findings have shown that by targeting acetylcholinesterase (AChE), CholinomiR-132 can intensify acute stress, resolve intestinal inflammation and change post-ischemic stroke responses. Further, we have identified clustered single nucleotide polymorphisms (SNPs) interfering with AChE silencing by several miRs which associate with elevated trait anxiety, blood pressure and inflammation. To further study miR regulators of ACh signalling, I plan to: (1) Identify anxiety and inflammation-induced changes in CholinomiRs and their targets in challenged brain and immune cells. (2) Establish the roles of these targets for one selected CholinomiR by tissue-specific manipulations. (3) Study primate-specific CholinomiRs by continued human DNA screens to identify SNPs and in ""humanized"" mice with knocked-in human AChE and transgenic CholinomiR-608. (4) Test if therapeutic modulation of aberrant CholinomiR expression can restore homeostasis. This research will clarify how miRs interact with each other in health and disease, introduce the dimension of complexity of multi-target competition and miR interactions and make a conceptual change in miRs research while enhancing the ability to intervene with diseases involving impaired ACh signalling."
Summary
"Communication between the nervous and the immune system is pivotal for maintaining homeostasis and ensuring rapid and efficient reaction to stress and infection insults. The emergence of microRNAs (miRs) as regulators of gene expression and of acetylcholine (ACh) signalling as regulator of anxiety and inflammation provides a model for studying this interaction. My hypothesis is that 1) a specific sub-group of miRs, designated ""CholinomiRs"", may silence multiple target genes in the neuro-immune interface; 2) these miRs compete with each other on the interaction with their targets, and 3) mutations interfering with miR binding lead to inherited susceptibility to anxiety and inflammation disorders by modifying these interactions. Our preliminary findings have shown that by targeting acetylcholinesterase (AChE), CholinomiR-132 can intensify acute stress, resolve intestinal inflammation and change post-ischemic stroke responses. Further, we have identified clustered single nucleotide polymorphisms (SNPs) interfering with AChE silencing by several miRs which associate with elevated trait anxiety, blood pressure and inflammation. To further study miR regulators of ACh signalling, I plan to: (1) Identify anxiety and inflammation-induced changes in CholinomiRs and their targets in challenged brain and immune cells. (2) Establish the roles of these targets for one selected CholinomiR by tissue-specific manipulations. (3) Study primate-specific CholinomiRs by continued human DNA screens to identify SNPs and in ""humanized"" mice with knocked-in human AChE and transgenic CholinomiR-608. (4) Test if therapeutic modulation of aberrant CholinomiR expression can restore homeostasis. This research will clarify how miRs interact with each other in health and disease, introduce the dimension of complexity of multi-target competition and miR interactions and make a conceptual change in miRs research while enhancing the ability to intervene with diseases involving impaired ACh signalling."
Max ERC Funding
2 375 600 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym CLUE-BGD
Project Closing the Loop between Understanding and Effective Treatment of the Basal Ganglia and their Disorders
Researcher (PI) Hagai Bergman
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS5, ERC-2012-ADG_20120314
Summary In this project, the basal ganglia are defined as actor-critic reinforcement learning networks that aim at an optimal tradeoff between the maximization of future cumulative rewards and the minimization of the cost (the reinforcement driven multi objective optimization RDMOO model).
This computational model will be tested by multiple neuron recordings in the major basal ganglia structures of monkeys engaged in a similar behavioral task. We will further validate the RMDOO computational model of the basal ganglia by extending our previous studies of neural activity in the MPTP primate model of Parkinson's disease to a primate model of central serotonin depletion and emotional dysregulation disorders. The findings in the primate model of emotional dysregulation will then be compared to electrophysiological recordings carried out in human patients with treatment-resistant major depression and obsessive compulsive disorder during deep brain stimulation (DBS) procedures. I aim to find neural signatures (e.g., synchronous gamma oscillations in the actor part of the basal ganglia as predicted by the RMDOO model) characterizing these emotional disorders and to use them as triggers for closed loop adaptive DBS. Our working hypothesis holds that, as for the MPTP model of Parkinson's disease, closed loop DBS will lead to greater amelioration of the emotional deficits in serotonin depleted monkeys.
This project incorporates extensive collaborations with a team of neurosurgeons, neurologists, psychiatrists, and computer science/ neural network researchers. If successful, the findings will provide a firm understanding of the computational physiology of the basal ganglia networks and their disorders. Importantly, they will pave the way to better treatment of human patients with severe mental disorders.
Summary
In this project, the basal ganglia are defined as actor-critic reinforcement learning networks that aim at an optimal tradeoff between the maximization of future cumulative rewards and the minimization of the cost (the reinforcement driven multi objective optimization RDMOO model).
This computational model will be tested by multiple neuron recordings in the major basal ganglia structures of monkeys engaged in a similar behavioral task. We will further validate the RMDOO computational model of the basal ganglia by extending our previous studies of neural activity in the MPTP primate model of Parkinson's disease to a primate model of central serotonin depletion and emotional dysregulation disorders. The findings in the primate model of emotional dysregulation will then be compared to electrophysiological recordings carried out in human patients with treatment-resistant major depression and obsessive compulsive disorder during deep brain stimulation (DBS) procedures. I aim to find neural signatures (e.g., synchronous gamma oscillations in the actor part of the basal ganglia as predicted by the RMDOO model) characterizing these emotional disorders and to use them as triggers for closed loop adaptive DBS. Our working hypothesis holds that, as for the MPTP model of Parkinson's disease, closed loop DBS will lead to greater amelioration of the emotional deficits in serotonin depleted monkeys.
This project incorporates extensive collaborations with a team of neurosurgeons, neurologists, psychiatrists, and computer science/ neural network researchers. If successful, the findings will provide a firm understanding of the computational physiology of the basal ganglia networks and their disorders. Importantly, they will pave the way to better treatment of human patients with severe mental disorders.
Max ERC Funding
2 476 922 €
Duration
Start date: 2013-12-01, End date: 2018-11-30
Project acronym CODECHECK
Project CRACKING THE CODE BEHIND MITOTIC FIDELITY: the roles of tubulin post-translational modifications and a chromosome separation checkpoint
Researcher (PI) Helder Jose Martins Maiato
Host Institution (HI) INSTITUTO DE BIOLOGIA MOLECULAR E CELULAR-IBMC
Call Details Consolidator Grant (CoG), LS3, ERC-2015-CoG
Summary During the human lifetime 10000 trillion cell divisions take place to ensure tissue homeostasis and several vital functions in the organism. Mitosis is the process that ensures that dividing cells preserve the chromosome number of their progenitors, while deviation from this, a condition known as aneuploidy, represents the most common feature in human cancers. Here we will test two original concepts with strong implications for chromosome segregation fidelity. The first concept is based on the “tubulin code” hypothesis, which predicts that molecular motors “read” tubulin post-translational modifications on spindle microtubules. Our proof-of-concept experiments demonstrate that tubulin detyrosination works as a navigation system that guides chromosomes towards the cell equator. Thus, in addition to regulating the motors required for chromosome motion, the cell might regulate the tracks in which they move on. We will combine proteomic, super-resolution and live-cell microscopy, with in vitro reconstitutions, to perform a comprehensive survey of the tubulin code and the respective implications for motors involved in chromosome motion, mitotic spindle assembly and correction of kinetochore-microtubule attachments. The second concept is centered on the recently uncovered chromosome separation checkpoint mediated by a midzone-associated Aurora B gradient, which delays nuclear envelope reformation in response to incompletely separated chromosomes. We aim to identify Aurora B targets involved in the spatiotemporal regulation of the anaphase-telophase transition. We will establish powerful live-cell microscopy assays and a novel mammalian model system to dissect how this checkpoint allows the detection and correction of lagging/long chromosomes and DNA bridges that would otherwise contribute to genomic instability. Overall, this work will establish a paradigm shift in our understanding of how spatial information is conveyed to faithfully segregate chromosomes during mitosis.
Summary
During the human lifetime 10000 trillion cell divisions take place to ensure tissue homeostasis and several vital functions in the organism. Mitosis is the process that ensures that dividing cells preserve the chromosome number of their progenitors, while deviation from this, a condition known as aneuploidy, represents the most common feature in human cancers. Here we will test two original concepts with strong implications for chromosome segregation fidelity. The first concept is based on the “tubulin code” hypothesis, which predicts that molecular motors “read” tubulin post-translational modifications on spindle microtubules. Our proof-of-concept experiments demonstrate that tubulin detyrosination works as a navigation system that guides chromosomes towards the cell equator. Thus, in addition to regulating the motors required for chromosome motion, the cell might regulate the tracks in which they move on. We will combine proteomic, super-resolution and live-cell microscopy, with in vitro reconstitutions, to perform a comprehensive survey of the tubulin code and the respective implications for motors involved in chromosome motion, mitotic spindle assembly and correction of kinetochore-microtubule attachments. The second concept is centered on the recently uncovered chromosome separation checkpoint mediated by a midzone-associated Aurora B gradient, which delays nuclear envelope reformation in response to incompletely separated chromosomes. We aim to identify Aurora B targets involved in the spatiotemporal regulation of the anaphase-telophase transition. We will establish powerful live-cell microscopy assays and a novel mammalian model system to dissect how this checkpoint allows the detection and correction of lagging/long chromosomes and DNA bridges that would otherwise contribute to genomic instability. Overall, this work will establish a paradigm shift in our understanding of how spatial information is conveyed to faithfully segregate chromosomes during mitosis.
Max ERC Funding
2 323 468 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym DEATHSWITCHING
Project Identifying genes and pathways that drive molecular switches and back-up mechanisms between apoptosis and autophagy
Researcher (PI) Adi Kimchi
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Advanced Grant (AdG), LS3, ERC-2012-ADG_20120314
Summary A cell’s decision to die is governed by multiple input signals received from a complex network of programmed cell death (PCD) pathways, including apoptosis and programmed necrosis. Additionally, under some conditions, autophagy, whose function is mainly pro-survival, may act as a back-up death pathway. We propose to apply new approaches to study the molecular basis of two important questions that await resolution in the field: a) how the cell switches from a pro-survival autophagic response to an apoptotic response and b) whether and how pro-survival autophagy is converted to a death mechanism when apoptosis is blocked. To address the first issue, we will screen for direct physical interactions between autophagic and apoptotic proteins, using the protein fragment complementation assay. Validated pairs will be studied in depth to identify built-in molecular switches that activate apoptosis when autophagy fails to restore homeostasis. As a pilot case to address the concept of molecular ‘sensors’ and ‘switches’, we will focus on the previously identified Atg12/Bcl-2 interaction. In the second line of research we will categorize autophagy-dependent cell death triggers into those that directly result from autophagy-dependent degradation, either by excessive self-digestion or by selective protein degradation, and those that utilize the autophagy machinery to activate programmed necrosis. We will identify the genes regulating these scenarios by whole genome RNAi screens for increased cell survival. In parallel, we will use a cell library of annotated fluorescent-tagged proteins for measuring selective protein degradation. These will be the starting point for identification of the molecular pathways that convert survival autophagy to a death program. Finally, we will explore the physiological relevance of back-up death mechanisms and the newly identified molecular mechanisms to developmental PCD during the cavitation process in early stages of embryogenesis.
Summary
A cell’s decision to die is governed by multiple input signals received from a complex network of programmed cell death (PCD) pathways, including apoptosis and programmed necrosis. Additionally, under some conditions, autophagy, whose function is mainly pro-survival, may act as a back-up death pathway. We propose to apply new approaches to study the molecular basis of two important questions that await resolution in the field: a) how the cell switches from a pro-survival autophagic response to an apoptotic response and b) whether and how pro-survival autophagy is converted to a death mechanism when apoptosis is blocked. To address the first issue, we will screen for direct physical interactions between autophagic and apoptotic proteins, using the protein fragment complementation assay. Validated pairs will be studied in depth to identify built-in molecular switches that activate apoptosis when autophagy fails to restore homeostasis. As a pilot case to address the concept of molecular ‘sensors’ and ‘switches’, we will focus on the previously identified Atg12/Bcl-2 interaction. In the second line of research we will categorize autophagy-dependent cell death triggers into those that directly result from autophagy-dependent degradation, either by excessive self-digestion or by selective protein degradation, and those that utilize the autophagy machinery to activate programmed necrosis. We will identify the genes regulating these scenarios by whole genome RNAi screens for increased cell survival. In parallel, we will use a cell library of annotated fluorescent-tagged proteins for measuring selective protein degradation. These will be the starting point for identification of the molecular pathways that convert survival autophagy to a death program. Finally, we will explore the physiological relevance of back-up death mechanisms and the newly identified molecular mechanisms to developmental PCD during the cavitation process in early stages of embryogenesis.
Max ERC Funding
2 500 000 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym DMR-CODE
Project Decoding the Mammalian transcriptional Regulatory code in development and stimulatory responses
Researcher (PI) Ido Amit
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2012-StG_20111109
Summary Transcription factors (TF) regulate genome function by controlling gene expression. Comprehensive characterization of the in vivo binding of TF to the DNA in relevant primary models is a critical step towards a global understanding of the human genome. Recent advances in high-throughput genomic technologies provide an extraordinary opportunity to develop and apply systematic approaches to learn the underline principles and mechanisms of mammalian transcriptional networks. The premise of this proposal is that a tractable set of rules govern how cells commit to a specific cell type or respond to the environment, and that these rules are coded in regulatory elements in the genome. Currently our understanding of the mammalian regulatory code is hampered by the difficulty of directly measuring in vivo binding of large numbers of TFs to DNA across multiple primary cell types and their natural response to physiological stimuli.
Here, we overcome this bottleneck by systematically exploring the genomic binding network of 1. All relevant TFs of key hematopoietic cells in both steady state and under relevant stimuli. 2. Follow the changes in TF networks as cells differentiate 3. Use these models to engineer cell states and responses. To achieve these goals, we developed a new method for automated high throughput ChIP coupled to sequencing (HT-ChIP-Seq). We used this method to measure binding of 40 TFs in 4 time points following stimulation of dendritic cells with pathogen components. We find that TFs vary substantially in their binding dynamics, genomic localization, number of binding events, and degree of interaction with other TFs. The analysis of this data suggests that the TF network is hierarchically organized, and composed of different types of TFs, cell differentiation factors, factors that prime for gene induction, and factors that bind more specifically and dynamically. This proposal revisits and challenges the current understanding of the mammalian regulatory code.
Summary
Transcription factors (TF) regulate genome function by controlling gene expression. Comprehensive characterization of the in vivo binding of TF to the DNA in relevant primary models is a critical step towards a global understanding of the human genome. Recent advances in high-throughput genomic technologies provide an extraordinary opportunity to develop and apply systematic approaches to learn the underline principles and mechanisms of mammalian transcriptional networks. The premise of this proposal is that a tractable set of rules govern how cells commit to a specific cell type or respond to the environment, and that these rules are coded in regulatory elements in the genome. Currently our understanding of the mammalian regulatory code is hampered by the difficulty of directly measuring in vivo binding of large numbers of TFs to DNA across multiple primary cell types and their natural response to physiological stimuli.
Here, we overcome this bottleneck by systematically exploring the genomic binding network of 1. All relevant TFs of key hematopoietic cells in both steady state and under relevant stimuli. 2. Follow the changes in TF networks as cells differentiate 3. Use these models to engineer cell states and responses. To achieve these goals, we developed a new method for automated high throughput ChIP coupled to sequencing (HT-ChIP-Seq). We used this method to measure binding of 40 TFs in 4 time points following stimulation of dendritic cells with pathogen components. We find that TFs vary substantially in their binding dynamics, genomic localization, number of binding events, and degree of interaction with other TFs. The analysis of this data suggests that the TF network is hierarchically organized, and composed of different types of TFs, cell differentiation factors, factors that prime for gene induction, and factors that bind more specifically and dynamically. This proposal revisits and challenges the current understanding of the mammalian regulatory code.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym ELIMINATESENESCENT
Project The Role of Elimination of Senescent Cells in Cancer Development
Researcher (PI) Valery Krizhanovsky
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS4, ERC-2012-StG_20111109
Summary Cellular senescence, which is a terminal cell cycle arrest, is a potent tumor suppressor mechanism that limits cancer initiation and progression; it also limits tissue damage response. While senescence is protective in the cell autonomous manner, senescent cells secrete a variety of factors that lead to inflammation, tissue destruction and promote tumorigenesis and metastasis in the sites of their presence. Here we propose a unique approach – to eliminate senescent cells from tissues in order to prevent the deleterious cell non-autonomous effects of these cells. We will use our understanding in immune surveillance of senescent cells, and in cell-intrinsic molecular pathways regulating cell viability, to identify the molecular “Achilles’ heal” of senescent cells. We will identify the mechanisms of interaction of senescent cells with NK cells and other immune cells, and harness these mechanisms for elimination of senescent cells. The impact of components of the main pathways regulating cell viability, apoptosis and autophagy, will then be evaluated for their specific contribution to the viability of senescent cells.
The molecular players identified by all these approaches will be readily implemented for the elimination of senescent cells in vivo. We will consequently be able to evaluate the impact of the elimination of senescent cells on tumor progression, in mouse models, where these cells are present during initial stages of tumorigenesis. Additionally, we will develop a novel mouse model that will allow identification of senescent cells in vivo in real time. This model is particularly challenging and valuable due to absence of single molecular marker for senescent cells.
The ability to eliminate senescent cells will lead to the understanding of the role of presence of senescent cells in tissues and the mechanisms regulating their viability. This might suggest novel ways of cancer prevention and treatment.
Summary
Cellular senescence, which is a terminal cell cycle arrest, is a potent tumor suppressor mechanism that limits cancer initiation and progression; it also limits tissue damage response. While senescence is protective in the cell autonomous manner, senescent cells secrete a variety of factors that lead to inflammation, tissue destruction and promote tumorigenesis and metastasis in the sites of their presence. Here we propose a unique approach – to eliminate senescent cells from tissues in order to prevent the deleterious cell non-autonomous effects of these cells. We will use our understanding in immune surveillance of senescent cells, and in cell-intrinsic molecular pathways regulating cell viability, to identify the molecular “Achilles’ heal” of senescent cells. We will identify the mechanisms of interaction of senescent cells with NK cells and other immune cells, and harness these mechanisms for elimination of senescent cells. The impact of components of the main pathways regulating cell viability, apoptosis and autophagy, will then be evaluated for their specific contribution to the viability of senescent cells.
The molecular players identified by all these approaches will be readily implemented for the elimination of senescent cells in vivo. We will consequently be able to evaluate the impact of the elimination of senescent cells on tumor progression, in mouse models, where these cells are present during initial stages of tumorigenesis. Additionally, we will develop a novel mouse model that will allow identification of senescent cells in vivo in real time. This model is particularly challenging and valuable due to absence of single molecular marker for senescent cells.
The ability to eliminate senescent cells will lead to the understanding of the role of presence of senescent cells in tissues and the mechanisms regulating their viability. This might suggest novel ways of cancer prevention and treatment.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-11-01, End date: 2017-10-31
Project acronym EVODEVOPATHS
Project Evolution of Developmental Gene Pathways
Researcher (PI) Itai Yanai
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS8, ERC-2012-StG_20111109
Summary The staggering diversity of the living world is a testament to the amount of variation available to the agency of natural selection. While it has been assumed that variation is entirely uniform and unbiased, recent work has challenged this notion. Evolutionary developmental biology seeks to understand the biases on variation imposed by developmental processes and their distinction from selective constraints. Metazoan development is best described by developmental gene pathways which are composed of transcription factors, signaling molecules, and terminal differentiation genes. A systematic comparison of such pathways across species would reveal the patterns of conservation and divergence; however this has not yet been achieved. In the EvoDevoPaths project we will develop a new approach to unravel pathways using both single-cell and tissue-specific transcriptomics. Our aim is to elucidate the evolution of developmental gene pathways using intricate embryology in the nematode phylum, a single-cell transcriptomic method we have developed, and sophisticated computational approaches for pathway comparisons. We will ask how variation is distributed across the specification and differentiation modules of a pathway using the nematode endoderm pathway as a model system. We further propose that the evolutionary change in the tissue specification pathways of early cell lineages is constrained by the properties of cell specification pathways. To test this hypothesis we will, for the first time, determine early developmental cell lineages from single cell transcriptomic data. Finally, we will attempt to unify the molecular signatures of conserved stages in disparate phyla under a framework in which they can be systematically compared. This research collectively represents the first time that developmental gene pathways are examined in an unbiased manner contributing to a theory of molecular variation that explains the evolutionary processes that underlie phenotypic novelty.
Summary
The staggering diversity of the living world is a testament to the amount of variation available to the agency of natural selection. While it has been assumed that variation is entirely uniform and unbiased, recent work has challenged this notion. Evolutionary developmental biology seeks to understand the biases on variation imposed by developmental processes and their distinction from selective constraints. Metazoan development is best described by developmental gene pathways which are composed of transcription factors, signaling molecules, and terminal differentiation genes. A systematic comparison of such pathways across species would reveal the patterns of conservation and divergence; however this has not yet been achieved. In the EvoDevoPaths project we will develop a new approach to unravel pathways using both single-cell and tissue-specific transcriptomics. Our aim is to elucidate the evolution of developmental gene pathways using intricate embryology in the nematode phylum, a single-cell transcriptomic method we have developed, and sophisticated computational approaches for pathway comparisons. We will ask how variation is distributed across the specification and differentiation modules of a pathway using the nematode endoderm pathway as a model system. We further propose that the evolutionary change in the tissue specification pathways of early cell lineages is constrained by the properties of cell specification pathways. To test this hypothesis we will, for the first time, determine early developmental cell lineages from single cell transcriptomic data. Finally, we will attempt to unify the molecular signatures of conserved stages in disparate phyla under a framework in which they can be systematically compared. This research collectively represents the first time that developmental gene pathways are examined in an unbiased manner contributing to a theory of molecular variation that explains the evolutionary processes that underlie phenotypic novelty.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym EVOEPIC
Project Evolutionary mechanisms of epigenomic and chromosomal aberrations in cancer
Researcher (PI) Amos Tanay
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS2, ERC-2012-StG_20111109
Summary Our working hypothesis is that tumorigenesis is an evolutionary process that fundamentally couples few major driving events (point mutations, rearrangements) with a complex flux of minor aberrations, many of which are epigenetic. We believe that these minor events are critical factors in the emergence of the cancer phenotype, and that understanding them is essential to the characterization of the disease. In particular, we hypothesize that a quantitative and principled evolutionary model for carcinogenesis is imperative for understanding the heterogeneity within tumor cell populations and predicting the effects of cancer therapies. We will therefore develop an interdisciplinary scheme that combines theoretical models of cancer evolution with in vitro evolutionary experiments and new methods for assaying the population heterogeneity of epigenomic organization. By developing techniques to interrogate DNA methylation and its interaction with other key epigenetic marks at the single-cell level, we will allow quantitative theoretical predictions to be scrutinized and refined. By combining models describing epigenetic aberrations with direct measurements of chromatin organization using Hi-C and 4C-seq, we shall revisit fundamental questions on the causative nature of epigenetic changes during carcinogenesis. Ultimately, we will apply both theoretical and experimental methodologies to assay and characterize the evolutionary histories of tumor cell populations from multiple mouse models and clinical patient samples.
Summary
Our working hypothesis is that tumorigenesis is an evolutionary process that fundamentally couples few major driving events (point mutations, rearrangements) with a complex flux of minor aberrations, many of which are epigenetic. We believe that these minor events are critical factors in the emergence of the cancer phenotype, and that understanding them is essential to the characterization of the disease. In particular, we hypothesize that a quantitative and principled evolutionary model for carcinogenesis is imperative for understanding the heterogeneity within tumor cell populations and predicting the effects of cancer therapies. We will therefore develop an interdisciplinary scheme that combines theoretical models of cancer evolution with in vitro evolutionary experiments and new methods for assaying the population heterogeneity of epigenomic organization. By developing techniques to interrogate DNA methylation and its interaction with other key epigenetic marks at the single-cell level, we will allow quantitative theoretical predictions to be scrutinized and refined. By combining models describing epigenetic aberrations with direct measurements of chromatin organization using Hi-C and 4C-seq, we shall revisit fundamental questions on the causative nature of epigenetic changes during carcinogenesis. Ultimately, we will apply both theoretical and experimental methodologies to assay and characterize the evolutionary histories of tumor cell populations from multiple mouse models and clinical patient samples.
Max ERC Funding
1 499 998 €
Duration
Start date: 2012-12-01, End date: 2017-11-30
Project acronym GAtransport
Project A direct, multi-faceted approach to investigate plant hormones spatial regulation: the case of gibberellins
Researcher (PI) Roy Weinstain
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS3, ERC-2015-STG
Summary Plants evolved a unique molecular mechanism that spatially regulate auxin, forming finely tuned gradients and local maxima of auxin that inform and direct developmental patterning and adaptive growth processes. Recent findings call into question the uniqueness of polar auxin transport in the sense that more plant hormones seem to be actively transported. Although still lacking many mechanistic details, as well as comprehensive functional connotations, these findings warrant a more thorough investigation into the prospect of a broader scope for plants spatial regulation capacity in the context of additional hormones. Critically, we lack an effective set of tools to directly investigate and dissect the particulars of plant hormones mobility at the molecular level. My long-term goal is to provide a molecular and mechanistic understanding of plant hormones dynamics that will augment our evolving model of how they are regulated and how they convey information. Here, I hypothesize that GA mobility in plants is controlled and directed by an active transport mechanism to form distinct distribution patterns that affect signaling. I will test my hypothesis with a multi-faceted and multi-disciplinary approach, combining: fluorescent labeling of key gibberellins to map their accumulation sites in whole plants and at the sub-cellular level; chemical-biology strategies that facilitate manipulation of GA “origin point” in planta to map and quantify GA flow pathways; probe-based genetic screens and un-biased photo-affinity labeling to identify proteins affecting GA mobility; and genetic and molecular biology techniques to characterize identified proteins’ functions. I expect to offer an exceptional, detailed view into the inner workings of gibberellins dynamics in planta and into the mechanisms driving it. I further anticipate that the strategies developed here to specifically address gibberellins could be straightforwardly re-tailored to investigate additional plant hormones.
Summary
Plants evolved a unique molecular mechanism that spatially regulate auxin, forming finely tuned gradients and local maxima of auxin that inform and direct developmental patterning and adaptive growth processes. Recent findings call into question the uniqueness of polar auxin transport in the sense that more plant hormones seem to be actively transported. Although still lacking many mechanistic details, as well as comprehensive functional connotations, these findings warrant a more thorough investigation into the prospect of a broader scope for plants spatial regulation capacity in the context of additional hormones. Critically, we lack an effective set of tools to directly investigate and dissect the particulars of plant hormones mobility at the molecular level. My long-term goal is to provide a molecular and mechanistic understanding of plant hormones dynamics that will augment our evolving model of how they are regulated and how they convey information. Here, I hypothesize that GA mobility in plants is controlled and directed by an active transport mechanism to form distinct distribution patterns that affect signaling. I will test my hypothesis with a multi-faceted and multi-disciplinary approach, combining: fluorescent labeling of key gibberellins to map their accumulation sites in whole plants and at the sub-cellular level; chemical-biology strategies that facilitate manipulation of GA “origin point” in planta to map and quantify GA flow pathways; probe-based genetic screens and un-biased photo-affinity labeling to identify proteins affecting GA mobility; and genetic and molecular biology techniques to characterize identified proteins’ functions. I expect to offer an exceptional, detailed view into the inner workings of gibberellins dynamics in planta and into the mechanisms driving it. I further anticipate that the strategies developed here to specifically address gibberellins could be straightforwardly re-tailored to investigate additional plant hormones.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-02-01, End date: 2021-01-31
Project acronym Gendever
Project Genome, the Edited Version: DNA and RNA Editing of Mammalian Retroelements
Researcher (PI) Erez Levanon
Host Institution (HI) BAR ILAN UNIVERSITY
Call Details Starting Grant (StG), LS2, ERC-2012-StG_20111109
Summary It is generally thought that an organism contains the exactly same genomic information in all its cells and that a genome remains unaltered throughout the organism’s life, with the exception of rare and random somatic mutations that might occur. This genomic information will also serve as a template for exact RNA copies. However, endogenous and powerful means of creating inner genomic diversity are known to exist: (1) RNA editing that leads to alteration of one nucleotide into another, (mainly A-to-I); (2) DNA editing that changes the DNA’s content by shifting C-into-U; (3) active retroelements that can insert copies of their sequences into new locations in a genome.
Recently, we and others have found that although considered extremely rare, all three mechanisms are active somatically or at least leave traces of their occurrence in the genome, and are linked together, as most editing events occur in retroelements. However, the magnitude and scope of these mechanisms, which can lead to huge diversity and complexity within an organism and even within a cell, are still a mystery. This explosion of genomic variety can have dramatic effect on diverse biological processes, such as brain complexity, cancer and evolution acceleration.
In GENEDVER, we aim to perform the first genome-wide mapping of editing and active retroelements in various genomes using a combination of computational and genomic approaches. Specifically, we will develop a strategy to detect RNA and DNA editing in retroelements, scan for editing events in various genomes, and build the first global editing atlas. In addition, we will exploit the close association between editing and retroelements in to produce a genome-wide approach to detect active retroelements. Finally, we will screen for editing events and retrotranspositions in various biological conditions, in order to expose their involvement in many biological states and evolution.
Summary
It is generally thought that an organism contains the exactly same genomic information in all its cells and that a genome remains unaltered throughout the organism’s life, with the exception of rare and random somatic mutations that might occur. This genomic information will also serve as a template for exact RNA copies. However, endogenous and powerful means of creating inner genomic diversity are known to exist: (1) RNA editing that leads to alteration of one nucleotide into another, (mainly A-to-I); (2) DNA editing that changes the DNA’s content by shifting C-into-U; (3) active retroelements that can insert copies of their sequences into new locations in a genome.
Recently, we and others have found that although considered extremely rare, all three mechanisms are active somatically or at least leave traces of their occurrence in the genome, and are linked together, as most editing events occur in retroelements. However, the magnitude and scope of these mechanisms, which can lead to huge diversity and complexity within an organism and even within a cell, are still a mystery. This explosion of genomic variety can have dramatic effect on diverse biological processes, such as brain complexity, cancer and evolution acceleration.
In GENEDVER, we aim to perform the first genome-wide mapping of editing and active retroelements in various genomes using a combination of computational and genomic approaches. Specifically, we will develop a strategy to detect RNA and DNA editing in retroelements, scan for editing events in various genomes, and build the first global editing atlas. In addition, we will exploit the close association between editing and retroelements in to produce a genome-wide approach to detect active retroelements. Finally, we will screen for editing events and retrotranspositions in various biological conditions, in order to expose their involvement in many biological states and evolution.
Max ERC Funding
1 499 249 €
Duration
Start date: 2012-10-01, End date: 2017-09-30
Project acronym GeneBodyMethylation
Project Resolving the Nuts and Bolts of Gene Body Methylation
Researcher (PI) Assaf Zemach
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS2, ERC-2015-STG
Summary DNA methylation, the covalent binding of a methyl group (CH3) to cytosine base, regulates the genome activity and plays a fundamental developmental role in eukaryotes. Its epigenetic characteristics of regulating transcription without changing the genetic code together with the ability to be transmitted through DNA replication allow organisms to memorize cellular events for many generations. DNA methylation is mostly known for its role in transcriptional silencing; however, its functional output is more complex and is influenced in part by its DNA context. Recent genomic studies, have found DNA methylation to be targeted inside sequences of actively transcribed genes, thus termed gene body methylation. Despite being an evolutionary conserved and a robust methylation pathway targeted to thousands of genes in animal and plant genomes, the function of gene body methylation is still poorly understood at both the molecular and functional level. Similar to the chicken and egg conundrum, because we do not know what gene body methylation does, therefore scientists could not apply its function to discover its regulators either. Gene body methylation is targeted to a very specific subset and subregions of genes, thus we strongly believe that specific factors exist and are missing simply because that no one has ever searched for them before. Hence, to make major breakthroughs in the field, our approach is to artificially generate gene-body-specific hypomethylated plants that together with customized genetic and biochemical systems will allow us to discover regulators and interpreters of gene body methylation. Using these unique genetic tools and novel molecular factors, we will be able to ultimately explore the particular biological roles of gene body methylation. These findings will fill the gap towards a full comprehension of the entire functional array of DNA methylation, and to its more precise exploitation in yielding better crops and in treating human diseases.
Summary
DNA methylation, the covalent binding of a methyl group (CH3) to cytosine base, regulates the genome activity and plays a fundamental developmental role in eukaryotes. Its epigenetic characteristics of regulating transcription without changing the genetic code together with the ability to be transmitted through DNA replication allow organisms to memorize cellular events for many generations. DNA methylation is mostly known for its role in transcriptional silencing; however, its functional output is more complex and is influenced in part by its DNA context. Recent genomic studies, have found DNA methylation to be targeted inside sequences of actively transcribed genes, thus termed gene body methylation. Despite being an evolutionary conserved and a robust methylation pathway targeted to thousands of genes in animal and plant genomes, the function of gene body methylation is still poorly understood at both the molecular and functional level. Similar to the chicken and egg conundrum, because we do not know what gene body methylation does, therefore scientists could not apply its function to discover its regulators either. Gene body methylation is targeted to a very specific subset and subregions of genes, thus we strongly believe that specific factors exist and are missing simply because that no one has ever searched for them before. Hence, to make major breakthroughs in the field, our approach is to artificially generate gene-body-specific hypomethylated plants that together with customized genetic and biochemical systems will allow us to discover regulators and interpreters of gene body methylation. Using these unique genetic tools and novel molecular factors, we will be able to ultimately explore the particular biological roles of gene body methylation. These findings will fill the gap towards a full comprehension of the entire functional array of DNA methylation, and to its more precise exploitation in yielding better crops and in treating human diseases.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym GPS-Bat
Project Foraging Decision Making in the Real World – revealed from a bat’s point of view by on-board miniature sensors
Researcher (PI) Yosef Gershon Yovel
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS8, ERC-2015-STG
Summary How animals make decisions in the wild is an open key-question in biology. Our lack of knowledge results from a technological gap – the difficulty to track animals over long periods while monitoring their behaviour; and from a conceptual gap – how to identify animals’ decision-points outdoors? We suggest applying our innovative on-board miniature sensors, to study decision making in the wild. We focus on one of the most fundamental contexts of decision making – foraging for food. We will study bats, which constitute over 20% of mammalian species and are extremely diverse, enabling to examine different aspects of decision making. Importantly, echolocating bats emit sound to perceive their environment, allowing us to infer their behavior (attacks on prey and interactions with conspecifics) via sound recording. Our miniature sensors include a GPS and an ultrasonic microphone, which enables us to reveal not only bats’ movements, but also their behavior and accordingly the factors underlying their decisions.
We will study three bat species to elucidate different aspects of foraging decisions: (1) How does animal sociality facilitate decision making? We have developed a system to monitor an entire colony including all conspecific-interactions when bats are in the roost or foraging outside. (2) How do animals weigh current input against previous experience? We will study a bat that must nightly search large areas over sea to find food. (3) How flexible are animal decisions? We will manipulate the natural environment of specific individuals to study how they adjust their foraging.
Our results will have far-reaching implications in many fields, from animal conservation to robotics. The operational and technical difficulty of performing controlled manipulations in the wild drives most disciplines to perform experiments exclusively in artificial laboratory conditions. Our approach opens new opportunities to conduct controlled studies in the natural environment.
Summary
How animals make decisions in the wild is an open key-question in biology. Our lack of knowledge results from a technological gap – the difficulty to track animals over long periods while monitoring their behaviour; and from a conceptual gap – how to identify animals’ decision-points outdoors? We suggest applying our innovative on-board miniature sensors, to study decision making in the wild. We focus on one of the most fundamental contexts of decision making – foraging for food. We will study bats, which constitute over 20% of mammalian species and are extremely diverse, enabling to examine different aspects of decision making. Importantly, echolocating bats emit sound to perceive their environment, allowing us to infer their behavior (attacks on prey and interactions with conspecifics) via sound recording. Our miniature sensors include a GPS and an ultrasonic microphone, which enables us to reveal not only bats’ movements, but also their behavior and accordingly the factors underlying their decisions.
We will study three bat species to elucidate different aspects of foraging decisions: (1) How does animal sociality facilitate decision making? We have developed a system to monitor an entire colony including all conspecific-interactions when bats are in the roost or foraging outside. (2) How do animals weigh current input against previous experience? We will study a bat that must nightly search large areas over sea to find food. (3) How flexible are animal decisions? We will manipulate the natural environment of specific individuals to study how they adjust their foraging.
Our results will have far-reaching implications in many fields, from animal conservation to robotics. The operational and technical difficulty of performing controlled manipulations in the wild drives most disciplines to perform experiments exclusively in artificial laboratory conditions. Our approach opens new opportunities to conduct controlled studies in the natural environment.
Max ERC Funding
1 928 750 €
Duration
Start date: 2016-03-01, End date: 2021-02-28
Project acronym GutBCells
Project Cellular Dynamics of Intestinal Antibody-Mediated Immune Response
Researcher (PI) Ziv Shulman Ben-Avraham
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE LTD
Call Details Starting Grant (StG), LS6, ERC-2015-STG
Summary Vaccination is widely used to prevent human diseases by inducing the formation of cellular and antibody-mediated immune responses for induction of long lasting immunological memory. Although most studies focus on immune responses elicited against injected immunizations, the simplest delivery of a vaccine regimen is by oral administration. The cellular and molecular components of the antibody immune response in peripheral lymph nodes in response to immunization are well described, however, much less is known about the dynamics of immune cells in gut associate lymphoid tissues (GALT) and adjust intestinal mucosal tissues. In the proposed research plan I will implicate intravital in vivo imaging for analysis of the cellular component of the antibody immune response in intestinal tissues. My goals are: 1. To track germinal center (GC) T cells for prolong time periods in peripheral lymph nodes and GALT and determine if they enter the memory compartment. For this purpose I will develop a new photoactivation method for permanently labeling immune cells and fate tracing of their daughter cells. 2. To examine T-B interactions and their regulation by intraceullar signaling pathways in GALT and to determine where and when class switch recombination to IgA takes place. For this purpose I will use intravital imaging of fluorescent reporter mice. 3. I will analyze the dynamics of plasma cell migration from Peyer’s patches to the mucosa by implementing state of the art photoactivation and imaging techniques that allow prolonged cell tracking. I will also use photoactivation approaches for sorting plasma cells from specific intestinal layers and perform gene expression analysis. 4. I will develop a new method to study dynamics and fate of B cells specific for commensal microbes in the GC, memory and plasma cell compartments. This research plan will extend our knowledge of the antibody immune response in intestinal tissues towards the future design of improved oral vaccinations.
Summary
Vaccination is widely used to prevent human diseases by inducing the formation of cellular and antibody-mediated immune responses for induction of long lasting immunological memory. Although most studies focus on immune responses elicited against injected immunizations, the simplest delivery of a vaccine regimen is by oral administration. The cellular and molecular components of the antibody immune response in peripheral lymph nodes in response to immunization are well described, however, much less is known about the dynamics of immune cells in gut associate lymphoid tissues (GALT) and adjust intestinal mucosal tissues. In the proposed research plan I will implicate intravital in vivo imaging for analysis of the cellular component of the antibody immune response in intestinal tissues. My goals are: 1. To track germinal center (GC) T cells for prolong time periods in peripheral lymph nodes and GALT and determine if they enter the memory compartment. For this purpose I will develop a new photoactivation method for permanently labeling immune cells and fate tracing of their daughter cells. 2. To examine T-B interactions and their regulation by intraceullar signaling pathways in GALT and to determine where and when class switch recombination to IgA takes place. For this purpose I will use intravital imaging of fluorescent reporter mice. 3. I will analyze the dynamics of plasma cell migration from Peyer’s patches to the mucosa by implementing state of the art photoactivation and imaging techniques that allow prolonged cell tracking. I will also use photoactivation approaches for sorting plasma cells from specific intestinal layers and perform gene expression analysis. 4. I will develop a new method to study dynamics and fate of B cells specific for commensal microbes in the GC, memory and plasma cell compartments. This research plan will extend our knowledge of the antibody immune response in intestinal tissues towards the future design of improved oral vaccinations.
Max ERC Funding
1 375 000 €
Duration
Start date: 2016-05-01, End date: 2021-04-30
Project acronym HIRESMEMMANIP
Project Spiking network mechanisms underlying short term memory
Researcher (PI) Eran Stark
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS5, ERC-2015-STG
Summary Short term memory (STM) is impaired at old age and a host of neuropsychiatric disorders, and has been the focus of a multitude of psychological and theoretical studies. However, the underlying neuronal circuit mechanisms remain elusive, mainly due to the lack of experimental tools: we suggest that rapid manipulations at the neuronal level are required for deciphering underlying mechanisms. We have developed an approach combining large-scale extracellular recordings and high density multi-site/multi-color optical stimulation (“diode-probes”), which enables high resolution closed-loop manipulation of multiple circuit elements in intact, free, behaving rodents. After training mice and rats to perform bridging-free STM-tasks, we will evaluate local circuit mechanisms in hippocampus and prefrontal cortex. Two broad classes of manipulations will be used: First, necessary components and timescales needed for STM maintenance will be established by focal real-time silencing of specific cell types and real-time jittering of spiking in those cells. Second, sufficient components (neuronal codes) will be determined by a circuit-training phase, in which novel associations between synthetic brain patterns and behaviorally-relevant short-term memory traces will be established. The first class is equivalent to erasing memories and the second to their writing. Together, these manipulations are expected to reveal global and local circuit mechanisms that facilitate STM maintenance in intact animals
Summary
Short term memory (STM) is impaired at old age and a host of neuropsychiatric disorders, and has been the focus of a multitude of psychological and theoretical studies. However, the underlying neuronal circuit mechanisms remain elusive, mainly due to the lack of experimental tools: we suggest that rapid manipulations at the neuronal level are required for deciphering underlying mechanisms. We have developed an approach combining large-scale extracellular recordings and high density multi-site/multi-color optical stimulation (“diode-probes”), which enables high resolution closed-loop manipulation of multiple circuit elements in intact, free, behaving rodents. After training mice and rats to perform bridging-free STM-tasks, we will evaluate local circuit mechanisms in hippocampus and prefrontal cortex. Two broad classes of manipulations will be used: First, necessary components and timescales needed for STM maintenance will be established by focal real-time silencing of specific cell types and real-time jittering of spiking in those cells. Second, sufficient components (neuronal codes) will be determined by a circuit-training phase, in which novel associations between synthetic brain patterns and behaviorally-relevant short-term memory traces will be established. The first class is equivalent to erasing memories and the second to their writing. Together, these manipulations are expected to reveal global and local circuit mechanisms that facilitate STM maintenance in intact animals
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-10-01, End date: 2021-09-30
Project acronym MechBiolRep
Project Mechanobiology of Bovine Reproduction
Researcher (PI) Amnon Buxboim
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS9, ERC-2015-STG
Summary The global demand for dairy products is expected to surge by 36% over the next decade in a manner that is progressively insatiable by existing technologies. The dairy industry relies on bovine reproduction, yet cow fertility is declining and the exact causes are not fully understood. It is clear, however, that the quality of bovine oocytes is decreasing.
In mammals, the ovarian reserve of oocytes stored within quiescent primordial follicles is non-renewable. Oocyte develop and mature within distinctive follicular microenvironments under tightly regulated molecular and physical conditions. Similarly, preimplantation embryo development is supported within a specialized microenvironment that is surrounded by the zona pellucida and insulated from external soluble and mechanical inputs. Characterizing and understanding these environments and how they affect reproductive processes is a key toward improving assisted reproductive technologies in bovine species.
Our premise is that molecular characterization of endocrine and paracrine signalling pathways must be complemented with understanding the mechanical regulation of reproductive biology. This premise is supported by recent finding showing that physical stresses and the mechanical compliance of the extracellular surroundings serve as potent regulators of cell fates in regeneration processes, development, and disease.
I propose to employ a biophysical and computational toolbox to study the mechanobiology of reproduction with application to bovine embryo-based technologies. By mimicking the mechanical properties of the ovarian cortical niche, which I will characterize using freshly derived ovaries, I will design an in vitro system for supporting follicle growth. Mechanical profiling of the entire developmental course from oocyte maturation to preimplantation embryogenesis will generate mechanistic insights into the physical regulation of reproductive processes.
Summary
The global demand for dairy products is expected to surge by 36% over the next decade in a manner that is progressively insatiable by existing technologies. The dairy industry relies on bovine reproduction, yet cow fertility is declining and the exact causes are not fully understood. It is clear, however, that the quality of bovine oocytes is decreasing.
In mammals, the ovarian reserve of oocytes stored within quiescent primordial follicles is non-renewable. Oocyte develop and mature within distinctive follicular microenvironments under tightly regulated molecular and physical conditions. Similarly, preimplantation embryo development is supported within a specialized microenvironment that is surrounded by the zona pellucida and insulated from external soluble and mechanical inputs. Characterizing and understanding these environments and how they affect reproductive processes is a key toward improving assisted reproductive technologies in bovine species.
Our premise is that molecular characterization of endocrine and paracrine signalling pathways must be complemented with understanding the mechanical regulation of reproductive biology. This premise is supported by recent finding showing that physical stresses and the mechanical compliance of the extracellular surroundings serve as potent regulators of cell fates in regeneration processes, development, and disease.
I propose to employ a biophysical and computational toolbox to study the mechanobiology of reproduction with application to bovine embryo-based technologies. By mimicking the mechanical properties of the ovarian cortical niche, which I will characterize using freshly derived ovaries, I will design an in vitro system for supporting follicle growth. Mechanical profiling of the entire developmental course from oocyte maturation to preimplantation embryogenesis will generate mechanistic insights into the physical regulation of reproductive processes.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
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 MorphoNotch
Project Multi-scale analysis of the interplay between cell morphology and cell-cell signaling
Researcher (PI) David Sprinzak
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Consolidator Grant (CoG), LS3, ERC-2015-CoG
Summary Signaling, genetic regulatory circuits, and tissue morphology are inherently coupled to each other during embryonic development. Although changes in cellular and tissue morphology are commonly treated as a downstream consequence of cell fate decision processes, there are multiple examples where morphological changes occur concurrently with the differentiation processes. This suggests that a feedback between cell morphology and regulatory processes can play an important role in coordinating tissue development. Currently, however, we lack the experimental, theoretical, and conceptual tools to understand this interplay between cell morphology, signaling, and regulatory circuits. In particular, we need to understand (1) how intercellular signaling depends on the cellular morphology and on the properties of the boundary between cells, and (2) how intercellular signaling, genetic circuits, and cell morphology integrate to generate robust differentiation patterns. Here, I propose to combine quantitative in-vitro and in-vivo experiments with mathematical modeling to address these questions in the context of Notch signaling and Notch mediated patterning, typically used for coordinating differentiation between neighboring cells during development. We will utilize novel reporters and micropatterning technology to analyze Notch signaling between pairs of cells. We will elucidate how the geometry and the molecular composition of the boundary between cells affect signaling. At the tissue level, we will study how the interplay between cell morphology and Notch signaling gives rise to robust patterning in the mammalian inner ear. We will use cochlear inner ear explant imaging to track the transition from disordered undifferentiated state to ordered pattern of hair and supporting cells in the cochlea. Together with a novel hybrid modeling approach, we will provide the foundation for a systems level understanding of development that interconnects morphology and regulatory circuits.
Summary
Signaling, genetic regulatory circuits, and tissue morphology are inherently coupled to each other during embryonic development. Although changes in cellular and tissue morphology are commonly treated as a downstream consequence of cell fate decision processes, there are multiple examples where morphological changes occur concurrently with the differentiation processes. This suggests that a feedback between cell morphology and regulatory processes can play an important role in coordinating tissue development. Currently, however, we lack the experimental, theoretical, and conceptual tools to understand this interplay between cell morphology, signaling, and regulatory circuits. In particular, we need to understand (1) how intercellular signaling depends on the cellular morphology and on the properties of the boundary between cells, and (2) how intercellular signaling, genetic circuits, and cell morphology integrate to generate robust differentiation patterns. Here, I propose to combine quantitative in-vitro and in-vivo experiments with mathematical modeling to address these questions in the context of Notch signaling and Notch mediated patterning, typically used for coordinating differentiation between neighboring cells during development. We will utilize novel reporters and micropatterning technology to analyze Notch signaling between pairs of cells. We will elucidate how the geometry and the molecular composition of the boundary between cells affect signaling. At the tissue level, we will study how the interplay between cell morphology and Notch signaling gives rise to robust patterning in the mammalian inner ear. We will use cochlear inner ear explant imaging to track the transition from disordered undifferentiated state to ordered pattern of hair and supporting cells in the cochlea. Together with a novel hybrid modeling approach, we will provide the foundation for a systems level understanding of development that interconnects morphology and regulatory circuits.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-06-01, End date: 2021-05-31
Project acronym MuDLOC
Project Multi-Dimensional Lab-On-Chip
Researcher (PI) Doron Gerber
Host Institution (HI) BAR ILAN UNIVERSITY
Call Details Starting Grant (StG), LS9, ERC-2012-StG_20111109
Summary There are several bottlenecks that hinder certain aspects of proteomics, in particular, incompatibility of high throughput technologies with certain protein types or modifications, low sensitivity and lack of quantitative data. I have developed a microfluidics affinity assay compatible with transmembrane proteins and post-translational modifications that is highly sensitive and can provide quantitative data.
The primary objective of this proposal is to bioengineer, using the abovementioned building blocks, a multi-functional microfluidic-based human protein arrays. The platform will enable addressing important scientific questions not otherwise possible. Specifically, the process of DNA demethylation, which is poorly characterised due to technological limitations. The biological aspects of chromatin methylation and their regulators that are crucial for cell differentiation and disease will be studied.
Work in MuDLOC will include the following: i) Bioengineering of a microfluidic-based platform that expresses thousands of human genes; ii) Design new tools for post-translational modifications and chromatin modifications; iii) Search for chromatin modifiers and their regulators; and iv) Exploration of specific inhibitors using a microfluidic inhibitor screen.
Beyond studying chromatin methylation from a new perspective, MuDLOC will greatly benefit a plethora of disciplines, such as proteomics, genomics and cancer research. At the end of the project my vision is to capture under one platform a whole pathway, including protein interactions, post-translational modifications and chromatin modifications.
Summary
There are several bottlenecks that hinder certain aspects of proteomics, in particular, incompatibility of high throughput technologies with certain protein types or modifications, low sensitivity and lack of quantitative data. I have developed a microfluidics affinity assay compatible with transmembrane proteins and post-translational modifications that is highly sensitive and can provide quantitative data.
The primary objective of this proposal is to bioengineer, using the abovementioned building blocks, a multi-functional microfluidic-based human protein arrays. The platform will enable addressing important scientific questions not otherwise possible. Specifically, the process of DNA demethylation, which is poorly characterised due to technological limitations. The biological aspects of chromatin methylation and their regulators that are crucial for cell differentiation and disease will be studied.
Work in MuDLOC will include the following: i) Bioengineering of a microfluidic-based platform that expresses thousands of human genes; ii) Design new tools for post-translational modifications and chromatin modifications; iii) Search for chromatin modifiers and their regulators; and iv) Exploration of specific inhibitors using a microfluidic inhibitor screen.
Beyond studying chromatin methylation from a new perspective, MuDLOC will greatly benefit a plethora of disciplines, such as proteomics, genomics and cancer research. At the end of the project my vision is to capture under one platform a whole pathway, including protein interactions, post-translational modifications and chromatin modifications.
Max ERC Funding
1 497 990 €
Duration
Start date: 2013-02-01, End date: 2018-01-31
Project acronym NATURAL_BAT_NAV
Project Neural basis of natural navigation: Representation of goals, 3-D spaces and 1-km distances in the bat hippocampal formation – the role of experience
Researcher (PI) Nachum Ulanovsky
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS5, ERC-2015-CoG
Summary The mammalian hippocampal formation contains place cells, grid cells, head-direction cells and border cells, which collectively represent the animal’s position (‘map’), distance traveled (‘odometer’) and direction (‘compass’), and are thought to underlie navigation. These neurons are typically studied in rodents running on linear tracks or in small empty boxes, ~1×1 m in size. However, real-world navigation differs dramatically from typical laboratory setups, in at least three ways – which we plan to study:
(1) The world is not empty, but contains objects and goals. Almost nothing is known about how neural circuits represent goal location – which is essential for navigating towards the goal. We will record single-neuron activity in bats flying towards spatial goals, in search for cells that encode vectorial information about the direction and distance to the goal. Preliminary results support the existence of such cells in the bat hippocampal formation. This new functional cell class of vectorial goal-encoding neurons may underlie goal-directed navigation.
(2) The world is not flat, but three-dimensional (3-D). We will train bats to fly in a large flight-room and examine 3-D grid cells and 3-D border cells.
(3) The world is not 1-m in size, and both rodents and bats navigate over kilometer-scale distances. Nothing is known about how the brain supports such real-life navigation. We will utilize a 1-km long test facility at the Weizmann Institute of Science, and record place cells and grid cells in bats navigating over biologically relevant spatial scales. Further, we will compare neural codes for space in wild-born bats versus bats born in the lab – which have never experienced a 1-km distance – to illuminate the role of experience in mammalian spatial cognition.
Taken together, this set of studies will bridge the gap – a conceptual gap and a gap in spatial scale – between hippocampal laboratory studies and real-world natural navigation.
Summary
The mammalian hippocampal formation contains place cells, grid cells, head-direction cells and border cells, which collectively represent the animal’s position (‘map’), distance traveled (‘odometer’) and direction (‘compass’), and are thought to underlie navigation. These neurons are typically studied in rodents running on linear tracks or in small empty boxes, ~1×1 m in size. However, real-world navigation differs dramatically from typical laboratory setups, in at least three ways – which we plan to study:
(1) The world is not empty, but contains objects and goals. Almost nothing is known about how neural circuits represent goal location – which is essential for navigating towards the goal. We will record single-neuron activity in bats flying towards spatial goals, in search for cells that encode vectorial information about the direction and distance to the goal. Preliminary results support the existence of such cells in the bat hippocampal formation. This new functional cell class of vectorial goal-encoding neurons may underlie goal-directed navigation.
(2) The world is not flat, but three-dimensional (3-D). We will train bats to fly in a large flight-room and examine 3-D grid cells and 3-D border cells.
(3) The world is not 1-m in size, and both rodents and bats navigate over kilometer-scale distances. Nothing is known about how the brain supports such real-life navigation. We will utilize a 1-km long test facility at the Weizmann Institute of Science, and record place cells and grid cells in bats navigating over biologically relevant spatial scales. Further, we will compare neural codes for space in wild-born bats versus bats born in the lab – which have never experienced a 1-km distance – to illuminate the role of experience in mammalian spatial cognition.
Taken together, this set of studies will bridge the gap – a conceptual gap and a gap in spatial scale – between hippocampal laboratory studies and real-world natural navigation.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-11-01, End date: 2021-10-31
Project acronym NEURO-POPCODE
Project Learning to read the code of large neural populations
Researcher (PI) Elad Schneidman
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS5, ERC-2012-StG_20111109
Summary Information is represented and transmitted in the brain by the joint activity of large groups of neurons. Understanding how information is “written” in these population patterns, and how it is read and processed, is a fundamental question in neuroscience. Yet, because of the huge number of potential activity patterns and complexity of natural stimuli, most of our understanding of the code relies on single neuron studies. We will extend and apply mathematical tools from information theory, machine learning, and physics, to overcome this ‘curse of dimensionality’ and build neural dictionaries relating activity and stimuli at an unparalleled resolution of hundreds of neurons. To identify the fundamental design principles of neural population codes we will study the spatial and spatio-temporal activity of hundreds of neurons from the retina, tectum, and cortical networks responding to naturalistic and artificial stimuli. Our primary goals are: (a) to characterize the encoding ‘codebooks’ of large populations of neurons, and the effect of network noise on encoding, and thus construct a thesaurus for neural populations, (b) use this thesaurus to develop new family of decoders of population activity which would be biologically plausible and accurate for natural stimuli, (c) characterize adaptation at the level of the code of networks of neurons, and the effect of learning on population neural codes, (d) explore “learnability” as a key feature of the neural code, and construct biologically plausible models of how the brain can learn to read population codes and compute, and (e) merge these ideas into a new mathematical framework that will connect the architecture of neural interaction networks and the properties of their neural codes. Our work will establish a new mathematical framework for studying the neural code, which will entail important implications for neural prostheses and brain machine interfaces, as well as brain-inspired learning algorithms.
Summary
Information is represented and transmitted in the brain by the joint activity of large groups of neurons. Understanding how information is “written” in these population patterns, and how it is read and processed, is a fundamental question in neuroscience. Yet, because of the huge number of potential activity patterns and complexity of natural stimuli, most of our understanding of the code relies on single neuron studies. We will extend and apply mathematical tools from information theory, machine learning, and physics, to overcome this ‘curse of dimensionality’ and build neural dictionaries relating activity and stimuli at an unparalleled resolution of hundreds of neurons. To identify the fundamental design principles of neural population codes we will study the spatial and spatio-temporal activity of hundreds of neurons from the retina, tectum, and cortical networks responding to naturalistic and artificial stimuli. Our primary goals are: (a) to characterize the encoding ‘codebooks’ of large populations of neurons, and the effect of network noise on encoding, and thus construct a thesaurus for neural populations, (b) use this thesaurus to develop new family of decoders of population activity which would be biologically plausible and accurate for natural stimuli, (c) characterize adaptation at the level of the code of networks of neurons, and the effect of learning on population neural codes, (d) explore “learnability” as a key feature of the neural code, and construct biologically plausible models of how the brain can learn to read population codes and compute, and (e) merge these ideas into a new mathematical framework that will connect the architecture of neural interaction networks and the properties of their neural codes. Our work will establish a new mathematical framework for studying the neural code, which will entail important implications for neural prostheses and brain machine interfaces, as well as brain-inspired learning algorithms.
Max ERC Funding
1 438 996 €
Duration
Start date: 2013-01-01, End date: 2018-10-31
Project acronym NEUROCOMMUNICATION
Project The Molecular Communication Mechanism of Motor Neuron Survival and Synapse Maintenance
Researcher (PI) Eran Perlson
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS5, ERC-2012-StG_20111109
Summary In order to survive and maintain normal function, the cell depends on a dynamic system of spatial specificity and fidelity of signaling pathways that can respond to both internal and external changes over space and time. This cell-cell communication is mediated by ligand-receptor mechanisms. In the case of highly polarized cells such as neurons trafficking mechanisms mediated by motor proteins are used to achieve precise signal targeting. Alterations in the trafficking machinery may results in incorrect signaling, that in some cases leads to neurodegeneration. An example for such phenomenon may be found in Amyotrophic Lateral Sclerosis (ALS). ALS is a motor neuron disease characterized by a non-cell autonomous neurodegeneration process, which involves neighboring cells via an unknown mechanism. This proposal focuses on the elucidation of basic cell-cell communication mechanisms by using the motor neuron degeneration process as a model. I aim to reveal critical communication mechanisms between the neuron and its environment for cell survival and synapse maintenance. My working hypothesis is that alterations in extrinsic and intrinsic signals may lead to the neurodegeneration seen in ALS. I will develop unique compartmental platforms mimicking the natural environment of the motor neuron. Then using differential “omics” approaches followed by functional assays I will reveal and characterized vital factors essential to neuron synapse integrity and neuron survival. Using state of the art live-cell imaging techniques I will reveal also the molecular mechanism for signals localization and targeting driven by the motor protein dynein. I will elucidate the molecular mechanism of neuronal communication with its diverse environment essential to its survival and proper function. The project will bring revolutionary new mechanistic insight to a truly fundamental problem in cell biology, how the cell communicates and how signals arrive at the right place at the right time?
Summary
In order to survive and maintain normal function, the cell depends on a dynamic system of spatial specificity and fidelity of signaling pathways that can respond to both internal and external changes over space and time. This cell-cell communication is mediated by ligand-receptor mechanisms. In the case of highly polarized cells such as neurons trafficking mechanisms mediated by motor proteins are used to achieve precise signal targeting. Alterations in the trafficking machinery may results in incorrect signaling, that in some cases leads to neurodegeneration. An example for such phenomenon may be found in Amyotrophic Lateral Sclerosis (ALS). ALS is a motor neuron disease characterized by a non-cell autonomous neurodegeneration process, which involves neighboring cells via an unknown mechanism. This proposal focuses on the elucidation of basic cell-cell communication mechanisms by using the motor neuron degeneration process as a model. I aim to reveal critical communication mechanisms between the neuron and its environment for cell survival and synapse maintenance. My working hypothesis is that alterations in extrinsic and intrinsic signals may lead to the neurodegeneration seen in ALS. I will develop unique compartmental platforms mimicking the natural environment of the motor neuron. Then using differential “omics” approaches followed by functional assays I will reveal and characterized vital factors essential to neuron synapse integrity and neuron survival. Using state of the art live-cell imaging techniques I will reveal also the molecular mechanism for signals localization and targeting driven by the motor protein dynein. I will elucidate the molecular mechanism of neuronal communication with its diverse environment essential to its survival and proper function. The project will bring revolutionary new mechanistic insight to a truly fundamental problem in cell biology, how the cell communicates and how signals arrive at the right place at the right time?
Max ERC Funding
1 499 800 €
Duration
Start date: 2013-02-01, End date: 2019-01-31
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 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 ORDER IN DISORDER
Project Order in Disorder: Context-dependent strategies for integrating peptide-mediated interactions
Researcher (PI) Ora Schueler-Furman
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS2, ERC-2012-StG_20111109
Summary Peptide-mediated protein interactions are emerging as key regulators of many important regulatory processes in the cell. Short motifs, usually embedded into disordered regions of the protein, interact with specific sets of protein partners in a transient way and allow efficient and malleable propagation of signals towards their targets. While regular protein interactions have been intensively studied, many aspects of peptide-mediated interactions have not yet been elucidated.
The aim of this proposal is to improve our understanding of the basic strategies that are employed by peptide-mediated interactions to achieve different types of outcomes in different settings, and how the context of the peptide influences this outcome. Towards this aim, we will establish two complementary strategies, namely (1) a significant extension of our modeling tools for peptide-protein complex structures that will allow modeling of effects of the surrounding flexible linker, and (2) the establishment of an experimental lab that will allow us to independently validate and complement our modeling results. Targeted modulation of peptide affinity, specificity, and linker length and sequence, using both computational design as well as experimental in vitro evolution, will dissect different contributions to the functional outcome of a peptide-mediated interaction within its context. We can thus study in detail the interplay of the interaction with additional features in the linker sequence, such as posttranslational modification sites, as well as additional peptide binding motifs and interactions.
Interactions mediated by intrinsically disordered regions are omnipresent. Their accurate characterization, modeling and manipulation holds therefore many promises towards applications for the development of better drugs and basic insights for better fundamental understanding of the underlying basis of regulatory interactions.
Summary
Peptide-mediated protein interactions are emerging as key regulators of many important regulatory processes in the cell. Short motifs, usually embedded into disordered regions of the protein, interact with specific sets of protein partners in a transient way and allow efficient and malleable propagation of signals towards their targets. While regular protein interactions have been intensively studied, many aspects of peptide-mediated interactions have not yet been elucidated.
The aim of this proposal is to improve our understanding of the basic strategies that are employed by peptide-mediated interactions to achieve different types of outcomes in different settings, and how the context of the peptide influences this outcome. Towards this aim, we will establish two complementary strategies, namely (1) a significant extension of our modeling tools for peptide-protein complex structures that will allow modeling of effects of the surrounding flexible linker, and (2) the establishment of an experimental lab that will allow us to independently validate and complement our modeling results. Targeted modulation of peptide affinity, specificity, and linker length and sequence, using both computational design as well as experimental in vitro evolution, will dissect different contributions to the functional outcome of a peptide-mediated interaction within its context. We can thus study in detail the interplay of the interaction with additional features in the linker sequence, such as posttranslational modification sites, as well as additional peptide binding motifs and interactions.
Interactions mediated by intrinsically disordered regions are omnipresent. Their accurate characterization, modeling and manipulation holds therefore many promises towards applications for the development of better drugs and basic insights for better fundamental understanding of the underlying basis of regulatory interactions.
Max ERC Funding
1 499 808 €
Duration
Start date: 2012-12-01, End date: 2017-11-30
Project acronym PARASITENUTRISENSING
Project Nutrient sensing by parasites
Researcher (PI) Maria Manuel Dias Da Mota
Host Institution (HI) INSTITUTO DE MEDICINA MOLECULAR JOAO LOBO ANTUNES
Call Details Starting Grant (StG), LS6, ERC-2012-StG_20111109
Summary As any other obligate parasite, Plasmodium depends on its hosts and on the nutrients they provide to survive and complete its life cycle. Surprisingly, nothing is know about Plasmodium’s capacity to sense nutrients or its host’s nutritional status and thereby reprogram its metabolism. Our preliminary data provides unequivocal evidence that Plasmodium has the ability to sense the host low-nutrient status and adapt to it by decreasing its multiplication rate. Thus, the overall goal of the present proposal is to unveil the molecular mechanisms by which parasites are capable to sense and adapt to environmental signals originated from nutrients and to determine its impact on the course and virulence of infection. To that end we propose to: (i) Identify Plasmodium pathway(s) that sense (host) nutritional changes; (ii) Uncover which molecules are sensed by Plasmodium during its intracellular development; (iii) Study the impact of the parasite’s nutrient sensing pathways activity on the course of infection; and (iv) Evaluate host nutritional status sensing as a common feature in parasites. The present proposal moves towards a change of paradigm on how host-parasite interactions are viewed. By definition, since a parasite requires a host in order to survive, a decrease in the availability of an essential molecule obtained from the host will weaken the parasite and render it incapable of succeeding in its life cycle. The rationale behind this proposal is that parasites monitor host nutritional environment and, prior to any nutrient(s) becoming limiting, are able to respond and adapt to the sensed alteration(s). Multidisciplinary approaches that combine genetic, genomic, cell biological and physiological methodologies will be used. Results arising from the present proposal will provide novel insights into the cell biology of these parasites and will increase our understanding of the interactions that these parasites maintain with their hosts.
Summary
As any other obligate parasite, Plasmodium depends on its hosts and on the nutrients they provide to survive and complete its life cycle. Surprisingly, nothing is know about Plasmodium’s capacity to sense nutrients or its host’s nutritional status and thereby reprogram its metabolism. Our preliminary data provides unequivocal evidence that Plasmodium has the ability to sense the host low-nutrient status and adapt to it by decreasing its multiplication rate. Thus, the overall goal of the present proposal is to unveil the molecular mechanisms by which parasites are capable to sense and adapt to environmental signals originated from nutrients and to determine its impact on the course and virulence of infection. To that end we propose to: (i) Identify Plasmodium pathway(s) that sense (host) nutritional changes; (ii) Uncover which molecules are sensed by Plasmodium during its intracellular development; (iii) Study the impact of the parasite’s nutrient sensing pathways activity on the course of infection; and (iv) Evaluate host nutritional status sensing as a common feature in parasites. The present proposal moves towards a change of paradigm on how host-parasite interactions are viewed. By definition, since a parasite requires a host in order to survive, a decrease in the availability of an essential molecule obtained from the host will weaken the parasite and render it incapable of succeeding in its life cycle. The rationale behind this proposal is that parasites monitor host nutritional environment and, prior to any nutrient(s) becoming limiting, are able to respond and adapt to the sensed alteration(s). Multidisciplinary approaches that combine genetic, genomic, cell biological and physiological methodologies will be used. Results arising from the present proposal will provide novel insights into the cell biology of these parasites and will increase our understanding of the interactions that these parasites maintain with their hosts.
Max ERC Funding
1 500 000 €
Duration
Start date: 2012-12-01, End date: 2017-11-30
Project acronym PhageResist
Project Beyond CRISPR: Systematic characterization of novel anti-phage defense systems in the microbial pan-genome
Researcher (PI) Rotem Sorek
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE LTD
Call Details Consolidator Grant (CoG), LS2, ERC-2015-CoG
Summary The perpetual arms race between bacteria and phage has resulted in the evolution of efficient resistance systems that protect bacteria from phage infection. Such systems, which include restriction enzymes and CRISPR-Cas, have major influence on the evolution of both bacteria and phage, and have also proven to be invaluable for molecular and biotechnological applications. Although much have been learned on the biology of bacterial defense against phage, more than half of all sequenced bacteria do not contain CRISPR-Cas, and it is estimated that many additional, yet-uncharacterized anti-phage defense systems are encoded in bacterial genomes.
The goal of this project is to systematically understand the arsenal of defense mechanisms that are at the disposal of microbes in their struggle against phages. The project combines computational genomics, synthetic biology, high-throughput robotic assays, and deep genetic and biochemical experiments to discover, verify, and study the properties of anti-phage defense systems.
Summary
The perpetual arms race between bacteria and phage has resulted in the evolution of efficient resistance systems that protect bacteria from phage infection. Such systems, which include restriction enzymes and CRISPR-Cas, have major influence on the evolution of both bacteria and phage, and have also proven to be invaluable for molecular and biotechnological applications. Although much have been learned on the biology of bacterial defense against phage, more than half of all sequenced bacteria do not contain CRISPR-Cas, and it is estimated that many additional, yet-uncharacterized anti-phage defense systems are encoded in bacterial genomes.
The goal of this project is to systematically understand the arsenal of defense mechanisms that are at the disposal of microbes in their struggle against phages. The project combines computational genomics, synthetic biology, high-throughput robotic assays, and deep genetic and biochemical experiments to discover, verify, and study the properties of anti-phage defense systems.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-07-01, End date: 2021-06-30
Project acronym PhotoPhage
Project The role of viral photosynthetic proteins in oceanic photosynthesis
Researcher (PI) Oded Beja
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Advanced Grant (AdG), LS8, ERC-2012-ADG_20120314
Summary Cyanobacteria play a key role in marine photosynthesis, contributing almost 50% of primary production in oligotrophic regions of the ocean. Marine cyanophages were recently discovered to carry photosystem II (PSII) genes, and it was suggested that these genes increase phage fitness by helping the phages to maintain photosynthesis in the infected bacterial cells. We recently showed evidence for the presence of photosystem I (PSI) genes in genomes of marine cyanophages [Sharon et al. 2009 Nature 461, 258-262]. Cyanobacterial core PSI gene cassettes, containing psaJFABCDEK, or psaDCAB gene cassettes forms unique clusters in cyanophage genomes, suggestive of selection for a distinct function in virus reproduction. Potentially, the proteins encoded by the viral genes are sufficient for forming intact monomeric PSI complexes. Projection of viral predicted peptides on the cyanobacterial PSI crystal structure suggests that the viral PSI components provide a unique way for funneling reducing power from respiratory and other electron transfer chains to PSI, therefore bypassing the need to rely solely on reducing power from the photosystem electron transfer chain.
The main goals of this proposal are:
(1) To determine how much of oceanic photosynthesis is actually performed with viral proteins.
(2) To establish a model system to understand the role of modified photosynthetic viral proteins in photosynthesis
We hypothesize that viral photosynthetic peptides are integrated into the bacterial photosynthetic membranes in order to maintain photosynthesis in infected cells, that otherwise stop to photosynthesize, and that changes are introduced to the system as a whole.
The proposed research will integrate concepts and techniques from metagenomics, metaproteomics and bioinformatics techniques to explore the interaction of viral PSII and PSI proteins with their host reaction center complexes, and to examine their influence on global marine photosynthesis production
Summary
Cyanobacteria play a key role in marine photosynthesis, contributing almost 50% of primary production in oligotrophic regions of the ocean. Marine cyanophages were recently discovered to carry photosystem II (PSII) genes, and it was suggested that these genes increase phage fitness by helping the phages to maintain photosynthesis in the infected bacterial cells. We recently showed evidence for the presence of photosystem I (PSI) genes in genomes of marine cyanophages [Sharon et al. 2009 Nature 461, 258-262]. Cyanobacterial core PSI gene cassettes, containing psaJFABCDEK, or psaDCAB gene cassettes forms unique clusters in cyanophage genomes, suggestive of selection for a distinct function in virus reproduction. Potentially, the proteins encoded by the viral genes are sufficient for forming intact monomeric PSI complexes. Projection of viral predicted peptides on the cyanobacterial PSI crystal structure suggests that the viral PSI components provide a unique way for funneling reducing power from respiratory and other electron transfer chains to PSI, therefore bypassing the need to rely solely on reducing power from the photosystem electron transfer chain.
The main goals of this proposal are:
(1) To determine how much of oceanic photosynthesis is actually performed with viral proteins.
(2) To establish a model system to understand the role of modified photosynthetic viral proteins in photosynthesis
We hypothesize that viral photosynthetic peptides are integrated into the bacterial photosynthetic membranes in order to maintain photosynthesis in infected cells, that otherwise stop to photosynthesize, and that changes are introduced to the system as a whole.
The proposed research will integrate concepts and techniques from metagenomics, metaproteomics and bioinformatics techniques to explore the interaction of viral PSII and PSI proteins with their host reaction center complexes, and to examine their influence on global marine photosynthesis production
Max ERC Funding
1 933 800 €
Duration
Start date: 2013-02-01, End date: 2018-01-31
Project acronym POSTTRANS
Project An interdisciplinary genome-wide study of posttranscriptional regulation by small RNAs: from individual interactions to networks and evolution
Researcher (PI) Hanah Margalit
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS2, ERC-2012-ADG_20120314
Summary Deciphering the interactions within and between the three major components of living organisms, DNA, RNA and Protein, is at the heart of biological research. New large-scale experimental methods have dramatically advanced genome-wide detection of protein-protein, protein-DNA, protein-RNA and protein-mediated RNA-RNA interactions. However, at present there is no large-scale method that could detect all RNA-RNA interactions independent of a mediator protein, or when the mediator protein is unknown. Attaining such a method is of utmost importance and is very timely, as it is now evident that RNA-RNA interactions play central roles in cellular life. In particular, hundreds of expressed small RNA (sRNA) molecules were discovered in both pro- and eukaryotes, many of which act as posttranscriptional regulators of gene expression by base-pairing with their mRNA targets. It seems that in many organisms the layer of posttranscriptional regulation is as widespread as transcription regulation, presenting a major challenge towards achieving functional and mechanistic understanding of this regulation level. Here we propose to develop an innovative methodology for genome-wide detection of the sRNA targetome, all mRNA targets of cellular sRNAs. This new methodology combines in vivo structural probing with deep sequencing and is independent of protein considerations. We will apply this method to deciper the sRNA targetome of the model organism Escherichia coli, which encodes over 100 sRNAs. We will use the sRNA targetome data as the foundation for a systematic ‘bottom-up’ computational analysis of multifaceted aspects of sRNA-mediated posttranscriptional regulation, encompassing the basic underlying rules of sRNA-mRNA target recognition, the design principles of the posttranscriptional regulatory network and its integration with the transcriptional and metabolic networks, and the evolution of posttranscriptional regulation.
Summary
Deciphering the interactions within and between the three major components of living organisms, DNA, RNA and Protein, is at the heart of biological research. New large-scale experimental methods have dramatically advanced genome-wide detection of protein-protein, protein-DNA, protein-RNA and protein-mediated RNA-RNA interactions. However, at present there is no large-scale method that could detect all RNA-RNA interactions independent of a mediator protein, or when the mediator protein is unknown. Attaining such a method is of utmost importance and is very timely, as it is now evident that RNA-RNA interactions play central roles in cellular life. In particular, hundreds of expressed small RNA (sRNA) molecules were discovered in both pro- and eukaryotes, many of which act as posttranscriptional regulators of gene expression by base-pairing with their mRNA targets. It seems that in many organisms the layer of posttranscriptional regulation is as widespread as transcription regulation, presenting a major challenge towards achieving functional and mechanistic understanding of this regulation level. Here we propose to develop an innovative methodology for genome-wide detection of the sRNA targetome, all mRNA targets of cellular sRNAs. This new methodology combines in vivo structural probing with deep sequencing and is independent of protein considerations. We will apply this method to deciper the sRNA targetome of the model organism Escherichia coli, which encodes over 100 sRNAs. We will use the sRNA targetome data as the foundation for a systematic ‘bottom-up’ computational analysis of multifaceted aspects of sRNA-mediated posttranscriptional regulation, encompassing the basic underlying rules of sRNA-mRNA target recognition, the design principles of the posttranscriptional regulatory network and its integration with the transcriptional and metabolic networks, and the evolution of posttranscriptional regulation.
Max ERC Funding
2 329 360 €
Duration
Start date: 2013-02-01, End date: 2019-01-31
Project acronym ProteinLocalization
Project Finding New Mechanisms for Protein Localization in Bacteria
Researcher (PI) Mariana Luisa Tomàs Gomes De Pinho
Host Institution (HI) INSTITUTO DE TECNOLOGIA QUIMICA E BIOLOGICA - UNIVERSIDADE NOVA DE LISBOA
Call Details Starting Grant (StG), LS6, ERC-2012-StG_20111109
Summary During infection, the host immune system interacts with the bacterial cell surface, a complex structure made of peptidoglycan, wall teichoic acids, lipoteichoic acids, capsule polysaccharide and peptidoglycan-attached proteins. A lot is known about the metabolic pathways for the synthesis of each individual cell surface component. Almost nothing is known about the coordination between the synthesis of the peptidoglycan, the major structural component of the cell surface and the main inflammatory component of gram-positive bacteria, and the synthesis of the other molecules present at the surface. However, this coordination is essential for the construction of a surface capable not only of performing its biological functions in cell protection and morphology, but also of masking its inflammatory components for evasion from host recognition.
Using the clinical pathogen Staphylococcus aureus as a model organism, we propose to investigate the temporal and spatial regulation of the enzymes responsible for the synthesis of the cell surface components, as well as their dependence on the underlying divisome.
We will (i) use state-of –the art fluorescence microscopy to localize fluorescent derivatives of enzymes required for cell surface synthesis; (ii) use libraries of antibiotics, of antisense RNA expression plasmids, and of transposon mutants to identify the order of assembly and requirements for the localization of cell surface synthesis enzymes; (iii) identify the exact metabolic compound/protein/geometric cue responsible for the localization of key enzymes; (iv) determine if cells with impaired surface synthesis due to protein delocalization are more susceptible to host recognition and therefore less capable of causing infections.
This project will result in the identification of new mechanisms of protein localization, a fundamental question in cell biology, and in a better understanding of the assembly of the bacterial cell surface of successful bacterial pathogens
Summary
During infection, the host immune system interacts with the bacterial cell surface, a complex structure made of peptidoglycan, wall teichoic acids, lipoteichoic acids, capsule polysaccharide and peptidoglycan-attached proteins. A lot is known about the metabolic pathways for the synthesis of each individual cell surface component. Almost nothing is known about the coordination between the synthesis of the peptidoglycan, the major structural component of the cell surface and the main inflammatory component of gram-positive bacteria, and the synthesis of the other molecules present at the surface. However, this coordination is essential for the construction of a surface capable not only of performing its biological functions in cell protection and morphology, but also of masking its inflammatory components for evasion from host recognition.
Using the clinical pathogen Staphylococcus aureus as a model organism, we propose to investigate the temporal and spatial regulation of the enzymes responsible for the synthesis of the cell surface components, as well as their dependence on the underlying divisome.
We will (i) use state-of –the art fluorescence microscopy to localize fluorescent derivatives of enzymes required for cell surface synthesis; (ii) use libraries of antibiotics, of antisense RNA expression plasmids, and of transposon mutants to identify the order of assembly and requirements for the localization of cell surface synthesis enzymes; (iii) identify the exact metabolic compound/protein/geometric cue responsible for the localization of key enzymes; (iv) determine if cells with impaired surface synthesis due to protein delocalization are more susceptible to host recognition and therefore less capable of causing infections.
This project will result in the identification of new mechanisms of protein localization, a fundamental question in cell biology, and in a better understanding of the assembly of the bacterial cell surface of successful bacterial pathogens
Max ERC Funding
1 656 960 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
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 ReachingCompleteness
Project The Molecular Basis of Somatic Nuclear Reprogramming
Researcher (PI) Yosef Buganim
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Starting Grant (StG), LS3, ERC-2015-STG
Summary The direct conversion approach and the generation of induced pluripotent stem cells (iPSCs) provide an invaluable resource of cells for disease modelling, drug screening, and patient-specific cell-based therapy. However, the directly converted cells are not stable, and the vast majority of iPSCs exhibit poor developmental potential as measured by stringent pluripotency tests. This suggests that the prevailing method of reprogramming is not ideal and leads to aberrant/incomplete conversion. To improve the quality of the converted cells, efforts should be focused on uncovering the molecular mechanisms that characterize the nuclear reprogramming process. There are two critical hurdles that hinder the progress of deciphering the elements that dictate successful reprogramming: (1) The ability to detect and capture solely the rare cells that eventually will be converted and (2) to monitor the transcriptional profile of cells at the single-cell level. Single-cell technology is in its infancy and many of the methods used today are characterized by high noise to signal ratio. In this grant proposal we intend to overcome these limitations by (1) establishing a complex fluorescent knock-in reporter system using the CRISPR/Cas9 method to capture the early rare reprogrammable cells and by (2) employing several cutting-edge single-cell technologies, RNA-Seq, Fluidigm BioMark and single-molecule mRNA-FISH, to segregate the real signal from the noise. To identify common and more global elements that facilitate nuclear reprogramming at large, we will trace in parallel, reprogrammable cells from two different somatic cell conversion models that reach high degree of nuclear reprogramming, and analyse their transcriptome using sophisticated bioinformatic tools. This study will provide a general overview of the changes that occur during the conversion of various cell types and will uncover the basic features that are essential to reach safe and complete conversion.
Summary
The direct conversion approach and the generation of induced pluripotent stem cells (iPSCs) provide an invaluable resource of cells for disease modelling, drug screening, and patient-specific cell-based therapy. However, the directly converted cells are not stable, and the vast majority of iPSCs exhibit poor developmental potential as measured by stringent pluripotency tests. This suggests that the prevailing method of reprogramming is not ideal and leads to aberrant/incomplete conversion. To improve the quality of the converted cells, efforts should be focused on uncovering the molecular mechanisms that characterize the nuclear reprogramming process. There are two critical hurdles that hinder the progress of deciphering the elements that dictate successful reprogramming: (1) The ability to detect and capture solely the rare cells that eventually will be converted and (2) to monitor the transcriptional profile of cells at the single-cell level. Single-cell technology is in its infancy and many of the methods used today are characterized by high noise to signal ratio. In this grant proposal we intend to overcome these limitations by (1) establishing a complex fluorescent knock-in reporter system using the CRISPR/Cas9 method to capture the early rare reprogrammable cells and by (2) employing several cutting-edge single-cell technologies, RNA-Seq, Fluidigm BioMark and single-molecule mRNA-FISH, to segregate the real signal from the noise. To identify common and more global elements that facilitate nuclear reprogramming at large, we will trace in parallel, reprogrammable cells from two different somatic cell conversion models that reach high degree of nuclear reprogramming, and analyse their transcriptome using sophisticated bioinformatic tools. This study will provide a general overview of the changes that occur during the conversion of various cell types and will uncover the basic features that are essential to reach safe and complete conversion.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-03-01, End date: 2021-02-28
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 SynapticMitochondria
Project Quality Control and Maintenance of Synaptic Mitochondria
Researcher (PI) Vanessa Alexandra Dos Santos Morais Epifânio
Host Institution (HI) INSTITUTO DE MEDICINA MOLECULAR JOAO LOBO ANTUNES
Call Details Starting Grant (StG), LS5, ERC-2015-STG
Summary Mitochondria at the synapse have a pivotal role in neurotransmitter release, but almost nothing is known about synaptic mitochondria composition or specific functions. Synaptic mitochondria compared to mitochondria in other cells, need to cope with increased calcium load, more oxidative stress, and high demands of energy generation during synaptic activity. My hypothesis is that synaptic mitochondria have acquired specific mechanisms to manage local stress and that disruption of these mechanisms contributes to neurodegeneration.
How mitochondria sense their dysfunction is unclear. Even more intriguing is the question how they decide whether their failure should lead to removal of the organelle or dismissal of the complete neuron via cell death. We anticipate that these decisions are not only operational during disease, but might constitute a fundamental mechanism relevant for maintenance of synaptic activity and establishment of new synapses.
Recent studies have revealed several genes implicated in neurodegenerative disorders involved in mitochondrial maintenance. However the function of these genes at the synapse, where the initial damage occurs, remains to be clarified. These genes provide excellent starting points to decipher the molecular mechanisms discussed above. Furthermore I propose to use proteomic approaches to identify the protein fingerprint of synaptic mitochondria and to compare them to mitochondria from other tissues. I plan to identify key players of the proposed regulatory pathways involved in intrinsic mitochondria quality control. In a complimentary approach, I will exploit our findings and use in vitro and in vivo experimental approaches to measure mitochondrial function of synaptic versus non-synaptic mitochondria and the relevance of those changes for synaptic function. Our work will unravel the specific properties of synaptic mitochondria and provide much needed insight in their hypothesized predominant role in neurodegenerative disorders.
Summary
Mitochondria at the synapse have a pivotal role in neurotransmitter release, but almost nothing is known about synaptic mitochondria composition or specific functions. Synaptic mitochondria compared to mitochondria in other cells, need to cope with increased calcium load, more oxidative stress, and high demands of energy generation during synaptic activity. My hypothesis is that synaptic mitochondria have acquired specific mechanisms to manage local stress and that disruption of these mechanisms contributes to neurodegeneration.
How mitochondria sense their dysfunction is unclear. Even more intriguing is the question how they decide whether their failure should lead to removal of the organelle or dismissal of the complete neuron via cell death. We anticipate that these decisions are not only operational during disease, but might constitute a fundamental mechanism relevant for maintenance of synaptic activity and establishment of new synapses.
Recent studies have revealed several genes implicated in neurodegenerative disorders involved in mitochondrial maintenance. However the function of these genes at the synapse, where the initial damage occurs, remains to be clarified. These genes provide excellent starting points to decipher the molecular mechanisms discussed above. Furthermore I propose to use proteomic approaches to identify the protein fingerprint of synaptic mitochondria and to compare them to mitochondria from other tissues. I plan to identify key players of the proposed regulatory pathways involved in intrinsic mitochondria quality control. In a complimentary approach, I will exploit our findings and use in vitro and in vivo experimental approaches to measure mitochondrial function of synaptic versus non-synaptic mitochondria and the relevance of those changes for synaptic function. Our work will unravel the specific properties of synaptic mitochondria and provide much needed insight in their hypothesized predominant role in neurodegenerative disorders.
Max ERC Funding
1 300 000 €
Duration
Start date: 2016-09-01, End date: 2021-08-31
Project acronym Temporal Coding
Project Do behaving animals extract information from precise spike timing? – The use of temporal codes
Researcher (PI) Moshe Parnas
Host Institution (HI) TEL AVIV UNIVERSITY
Call Details Starting Grant (StG), LS5, ERC-2015-STG
Summary Neural temporal codes have come to dominate our way of thinking on how information is coded in the brain. When precise spike timing is found to carry information, the neural code is defined as a temporal code. In spite of the importance of temporal codes, whether behaving animals actually use this type of coding is still an unresolved question. To date studying temporal codes was technically impossible due to the inability to manipulate spike timing in behaving animals. However, very recent developments in optogenetics solved this problem. Despite these modern tools, this key question is very difficult to resolve in mammals, because the meaning of manipulating a part of a neural circuit without knowledge of the neural activity of all the neurons involved in the coding is unclear.
The fly is an ideal model system to study temporal codes because its small number of neurons allows for complete mapping of the neural activity of all the neurons involved. Since temporal codes are suggested to be involved in olfactory intensity coding, I will study this process. I will device a multidisciplinary approach of electrophysiology, two-photon imaging and behavior.
I aim to examine for the first time directly whether temporal coding is used by behaving animals and to unravel the circuits and mechanisms that underlie intensity coding. To do so, I will manipulate the temporal codes in behaving animals and examine whether the behavioral responses change accordingly. To guide this study I will generate three novel databases of: i. the temporal activity of all neurons involved in Drosophila olfactory intensity coding. ii. The functional connectivity between the two brain regions that are involved in the intensity coding and iii. behavioral responses to different odors and intensities.
Thus, this research will use cutting edge techniques to resolve a long standing basic question in neuroscience: how does the brain actually code information?
Summary
Neural temporal codes have come to dominate our way of thinking on how information is coded in the brain. When precise spike timing is found to carry information, the neural code is defined as a temporal code. In spite of the importance of temporal codes, whether behaving animals actually use this type of coding is still an unresolved question. To date studying temporal codes was technically impossible due to the inability to manipulate spike timing in behaving animals. However, very recent developments in optogenetics solved this problem. Despite these modern tools, this key question is very difficult to resolve in mammals, because the meaning of manipulating a part of a neural circuit without knowledge of the neural activity of all the neurons involved in the coding is unclear.
The fly is an ideal model system to study temporal codes because its small number of neurons allows for complete mapping of the neural activity of all the neurons involved. Since temporal codes are suggested to be involved in olfactory intensity coding, I will study this process. I will device a multidisciplinary approach of electrophysiology, two-photon imaging and behavior.
I aim to examine for the first time directly whether temporal coding is used by behaving animals and to unravel the circuits and mechanisms that underlie intensity coding. To do so, I will manipulate the temporal codes in behaving animals and examine whether the behavioral responses change accordingly. To guide this study I will generate three novel databases of: i. the temporal activity of all neurons involved in Drosophila olfactory intensity coding. ii. The functional connectivity between the two brain regions that are involved in the intensity coding and iii. behavioral responses to different odors and intensities.
Thus, this research will use cutting edge techniques to resolve a long standing basic question in neuroscience: how does the brain actually code information?
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-01-01, End date: 2020-12-31
Project acronym TENDONTOBONE
Project The mechanisms that underlie the development of a tendon-bone attachment unit
Researcher (PI) Elazar Zelzer
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Starting Grant (StG), LS3, ERC-2012-StG_20111109
Summary We walk, run and jump using the complex and ingenious musculoskeletal system. It is therefore puzzling that although each of its components has been extensively studied, research of the musculoskeleton as an integrated system and, in particular, of its assembly has been scarce. In recent years, studies conducted in my lab have demonstrated the centrality of cross regulation between musculoskeletal tissues in skeletogenesis. These works have provided me with the inspiration for a revolutionary hypothesis on the way tendons connect to bones, along with sufficient preliminary data on which to base it.
The critical component in the assembly of the musculoskeleton is the formation of an attachment unit, where a tendon is inserted into a bone. Instead of two tissues that attach to each other, my novel hypothesis suggests that the entire attachment unit originates from a single pool of progenitor cells, which following differentiation diverges to form a tendon attached to cartilage.
With the support of the ERC scheme, I will uncover the previously uncharacterized cellular origin of the attachment unit and the genetic program underlying its development. The attachment unit is a compound tissue, as it is composed of chondrocytes at one end and of tenocytes at the other end. We will investigate the mechanisms that facilitate in situ differentiation of mesenchymal progenitor cells into two distinct cell fates, under one defined niche. In addition, I will identify the contribution of both mechanical stimuli and molecular signals to the development of the attachment unit.
The ultimate goal of this program is to provide a complete picture of attachment unit development, in order to promote understanding of musculoskeletal assembly. The acquired knowledge may provide the basis for new therapies for enthesopathies, through tissue engineering or repair.
Summary
We walk, run and jump using the complex and ingenious musculoskeletal system. It is therefore puzzling that although each of its components has been extensively studied, research of the musculoskeleton as an integrated system and, in particular, of its assembly has been scarce. In recent years, studies conducted in my lab have demonstrated the centrality of cross regulation between musculoskeletal tissues in skeletogenesis. These works have provided me with the inspiration for a revolutionary hypothesis on the way tendons connect to bones, along with sufficient preliminary data on which to base it.
The critical component in the assembly of the musculoskeleton is the formation of an attachment unit, where a tendon is inserted into a bone. Instead of two tissues that attach to each other, my novel hypothesis suggests that the entire attachment unit originates from a single pool of progenitor cells, which following differentiation diverges to form a tendon attached to cartilage.
With the support of the ERC scheme, I will uncover the previously uncharacterized cellular origin of the attachment unit and the genetic program underlying its development. The attachment unit is a compound tissue, as it is composed of chondrocytes at one end and of tenocytes at the other end. We will investigate the mechanisms that facilitate in situ differentiation of mesenchymal progenitor cells into two distinct cell fates, under one defined niche. In addition, I will identify the contribution of both mechanical stimuli and molecular signals to the development of the attachment unit.
The ultimate goal of this program is to provide a complete picture of attachment unit development, in order to promote understanding of musculoskeletal assembly. The acquired knowledge may provide the basis for new therapies for enthesopathies, through tissue engineering or repair.
Max ERC Funding
1 499 999 €
Duration
Start date: 2013-01-01, End date: 2017-12-31
Project acronym Tolerome
Project Evolution of antibiotic tolerance in the 'wild': A quantitative approach
Researcher (PI) Nathalie Balaban
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Consolidator Grant (CoG), LS8, ERC-2015-CoG
Summary Bacterial ability to evolve strategies for evading antibiotic treatment is a fascinating example of an evolutionary process, as well as a major health threat. Despite efforts to understand treatment failure, we lack the means to prevent evolution of resistance when a new drug is released to the market. Most efforts are directed towards understanding the mechanisms of antibiotic resistance. Whereas ‘resistance’ is due to mutations that enable microorganisms to grow even at high concentrations of the drug, ‘tolerance’ is the ability to sustain a transient treatment, for example by entering a mode of transient dormancy. The importance of tolerance in the clinic has not been investigated as thoroughly as resistance. The presence of tolerant bacteria is not detected in the clinic because of the inherent difficulty of tracking dormant bacteria that often constitute only a minute fraction of the bacterial population. I hypothesize that bacterial dormancy may evolve quickly in the host under antibiotic treatment. This hypothesis is strengthened by our recent results demonstrating the rapid evolution of dormancy leading to tolerance in vitro, and by the increasing number of cases of treatment failure in the clinic not explained by resistance. My goal is to develop a multidisciplinary approach to detect, quantify and characterize tolerant bacteria in the clinic. Using my background in quantitative single-cell analyses, I will develop microfluidic devices for the rapid detection of tolerant bacteria in the clinic, systems biology tools to isolate and analyze dormant sub-populations directly from clinical isolates. I will search for the genetic mutations leading to tolerance, namely build what I term here the ‘tolerome’. The results will be analyzed in a mathematical framework of tolerance evolution. This approach should reveal the role of tolerance in the clinic and may lead to a paradigm shift in the way bacterial infections are characterized and treated.
Summary
Bacterial ability to evolve strategies for evading antibiotic treatment is a fascinating example of an evolutionary process, as well as a major health threat. Despite efforts to understand treatment failure, we lack the means to prevent evolution of resistance when a new drug is released to the market. Most efforts are directed towards understanding the mechanisms of antibiotic resistance. Whereas ‘resistance’ is due to mutations that enable microorganisms to grow even at high concentrations of the drug, ‘tolerance’ is the ability to sustain a transient treatment, for example by entering a mode of transient dormancy. The importance of tolerance in the clinic has not been investigated as thoroughly as resistance. The presence of tolerant bacteria is not detected in the clinic because of the inherent difficulty of tracking dormant bacteria that often constitute only a minute fraction of the bacterial population. I hypothesize that bacterial dormancy may evolve quickly in the host under antibiotic treatment. This hypothesis is strengthened by our recent results demonstrating the rapid evolution of dormancy leading to tolerance in vitro, and by the increasing number of cases of treatment failure in the clinic not explained by resistance. My goal is to develop a multidisciplinary approach to detect, quantify and characterize tolerant bacteria in the clinic. Using my background in quantitative single-cell analyses, I will develop microfluidic devices for the rapid detection of tolerant bacteria in the clinic, systems biology tools to isolate and analyze dormant sub-populations directly from clinical isolates. I will search for the genetic mutations leading to tolerance, namely build what I term here the ‘tolerome’. The results will be analyzed in a mathematical framework of tolerance evolution. This approach should reveal the role of tolerance in the clinic and may lead to a paradigm shift in the way bacterial infections are characterized and treated.
Max ERC Funding
1 978 750 €
Duration
Start date: 2016-05-01, End date: 2021-04-30
Project acronym TranslationRegCode
Project Cracking the Translation Regulatory Code
Researcher (PI) Reut Gitit Shalgi
Host Institution (HI) TECHNION - ISRAEL INSTITUTE OF TECHNOLOGY
Call Details Starting Grant (StG), LS2, ERC-2015-STG
Summary Organisms across all kingdoms share several systems that are essential to life, one of the most central being protein synthesis. Living in a continuously changing environment, cells need to constantly respond to various environmental cues and change their protein landscape. In extreme cases, cells globally shut down protein synthesis and upregulate stress-protective proteins.
Mechanisms of translational repression or selective enhancement of stress-induced proteins have been characterized, but their effects were demonstrated on an individual mRNA basis. Which target mRNAs are translationally regulated in response to different environmental cues, and what are the cis-regulatory elements involved, largely remain as open questions. Using ribosome footprint profiling, I recently discovered a novel mode of translational control in stress, underscoring the potential of new technologies to uncover novel regulatory mechanisms. But while transcription cis-regulatory elements have been thoroughly mapped in the past decade, and splicing regulatory elements are accumulating, the identification of translation cis-regulatory elements is lagging behind.
Here I propose to crack the mammalian translation regulatory code, and close this long-standing gap. I present a novel interdisciplinary framework to comprehensively identify translation cis-regulatory elements, and map their mRNAs targets in a variety of cellular perturbations. Importantly, we plan to explore mechanisms underlying novel cis-regulatory elements, and create the first genome-wide functionally annotated translation regulatory code.
The translation regulatory code will map targets of existing mechanisms and shed light on newly identified pathways that play a role in stress-induced translational control. The proposed project is an imperative stepping stone to understanding translational regulation by cis-regulatory elements, opening new avenues in the functional genomics research of translational control.
Summary
Organisms across all kingdoms share several systems that are essential to life, one of the most central being protein synthesis. Living in a continuously changing environment, cells need to constantly respond to various environmental cues and change their protein landscape. In extreme cases, cells globally shut down protein synthesis and upregulate stress-protective proteins.
Mechanisms of translational repression or selective enhancement of stress-induced proteins have been characterized, but their effects were demonstrated on an individual mRNA basis. Which target mRNAs are translationally regulated in response to different environmental cues, and what are the cis-regulatory elements involved, largely remain as open questions. Using ribosome footprint profiling, I recently discovered a novel mode of translational control in stress, underscoring the potential of new technologies to uncover novel regulatory mechanisms. But while transcription cis-regulatory elements have been thoroughly mapped in the past decade, and splicing regulatory elements are accumulating, the identification of translation cis-regulatory elements is lagging behind.
Here I propose to crack the mammalian translation regulatory code, and close this long-standing gap. I present a novel interdisciplinary framework to comprehensively identify translation cis-regulatory elements, and map their mRNAs targets in a variety of cellular perturbations. Importantly, we plan to explore mechanisms underlying novel cis-regulatory elements, and create the first genome-wide functionally annotated translation regulatory code.
The translation regulatory code will map targets of existing mechanisms and shed light on newly identified pathways that play a role in stress-induced translational control. The proposed project is an imperative stepping stone to understanding translational regulation by cis-regulatory elements, opening new avenues in the functional genomics research of translational control.
Max ERC Funding
1 587 500 €
Duration
Start date: 2016-03-01, End date: 2021-02-28
Project acronym TSGPs-of-CFSs
Project Role of Tumour Suppressor Gene Products of Common Fragile Sites in Human Diseases
Researcher (PI) Rami Aqeilan
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Consolidator Grant (CoG), LS4, ERC-2015-CoG
Summary Common fragile sites (CFSs) are large chromosomal regions identified by conventional cytogenetics as sequences prone to breakage in cells subjected to replication stress. The interest in CFSs stems from their key role in DNA damage, resulting in chromosomal rearrangements. The instability of CFSs was correlated with genome instability in precancerous lesions and during tumour progression. Two opposing views dominate the discussion regarding the role of CFSs. One school of thought suggested that genomic instability during cancer progression causes collateral damage to genes residing within CFSs, such as WWOX and FHIT. These genes are proposed to be unselected ‘‘passenger’’ mutations. The counter argument is that deletions and other genomic alterations in CFSs occur early in cancer development. Cancer cells with deletions in genes that span CFSs are then selectively expanded due to loss of tumour suppressor functions such as protection of genome stability, coordination of cell cycle or apoptosis.
Recent observations from my lab clearly suggest that gene products from CFSs play driver roles in cancer transformation. Moreover, we have evidence for the involvement of DNA damage and Wwox in pancreatic β-cells in the context of diabetes. Here, I propose to investigate the role of tumour suppressor gene products (TSGPs) of CFSs in human diseases. Three approaches will be taken to tackle this question. First, molecular functions of TSGPs of CFSs will be determined using state-of-the-art genetic tools in vitro. Second, novel transgenic mouse tools will be used to study CFSs and their associated TSGs in preneoplastic lesions and tumours in vivo, with confirmatory studies in human material. Third, we will examine the potential involvement of CFSs and their TSGPs in type-2 diabetes (T2D).
The expected outcome is a detailed molecular understanding of CFSs and their associated TSGPs in genomic instability as well as their roles in cancer and metabolic diseases.
Summary
Common fragile sites (CFSs) are large chromosomal regions identified by conventional cytogenetics as sequences prone to breakage in cells subjected to replication stress. The interest in CFSs stems from their key role in DNA damage, resulting in chromosomal rearrangements. The instability of CFSs was correlated with genome instability in precancerous lesions and during tumour progression. Two opposing views dominate the discussion regarding the role of CFSs. One school of thought suggested that genomic instability during cancer progression causes collateral damage to genes residing within CFSs, such as WWOX and FHIT. These genes are proposed to be unselected ‘‘passenger’’ mutations. The counter argument is that deletions and other genomic alterations in CFSs occur early in cancer development. Cancer cells with deletions in genes that span CFSs are then selectively expanded due to loss of tumour suppressor functions such as protection of genome stability, coordination of cell cycle or apoptosis.
Recent observations from my lab clearly suggest that gene products from CFSs play driver roles in cancer transformation. Moreover, we have evidence for the involvement of DNA damage and Wwox in pancreatic β-cells in the context of diabetes. Here, I propose to investigate the role of tumour suppressor gene products (TSGPs) of CFSs in human diseases. Three approaches will be taken to tackle this question. First, molecular functions of TSGPs of CFSs will be determined using state-of-the-art genetic tools in vitro. Second, novel transgenic mouse tools will be used to study CFSs and their associated TSGs in preneoplastic lesions and tumours in vivo, with confirmatory studies in human material. Third, we will examine the potential involvement of CFSs and their TSGPs in type-2 diabetes (T2D).
The expected outcome is a detailed molecular understanding of CFSs and their associated TSGPs in genomic instability as well as their roles in cancer and metabolic diseases.
Max ERC Funding
2 000 000 €
Duration
Start date: 2016-05-01, End date: 2021-04-30
Project acronym Ubl-Code
Project Revealing the ubiquitin and ubiquitin-like modification landscape in health and disease
Researcher (PI) Yifat Haya Merbl
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE LTD
Call Details Starting Grant (StG), LS2, ERC-2015-STG
Summary Post-translational modifications (PTMs) of proteins are a major tool that the cell uses to monitor events and initiate appropriate responses. While a protein is defined by its backbone of amino acid sequence, its function is often determined by PTMs, which specify stability, activity, or cellular localization. Among PTMs, ubiquitin and ubiquitin-like (Ubl) modifications were shown to regulate a variety of fundamental cellular processes such as cell division and differentiation. Aberrations in these pathways have been implicated in the pathogenesis of cancer. Over the past decade high-throughput genomic and transcriptional analyses have profoundly broadened our understanding of the processes underlying cancer development and progression. Yet, proteomic analyses and the PTM landscape in cancer, remained relatively unexplored.
Our goal is to decipher molecular mechanisms of Ubl regulation in cancer. We will utilize the PTM profiling technology that I developed and further develop it to allow for subsequent MS analysis. Together with cutting-edge genomic, imaging and proteomic technologies, we will analyze novel aspects of PTM regulation at the level of the enzymatic machinery, the substrates and the downstream cellular network. We will rely on ample in-vitro and in-vivo characterization of Ubl conjugation to:a. Elucidate the regulatory principles of substrate specificity and recognition. b. Understand signalling dynamics in the ubiquitin system. c. Reveal how aberrations in these pathways may lead to diseases such as cancer. Identifying both the Ubl modifying enzymes and the modified substrates will form the basis for deciphering the molecular pathways in which they operate in the cell and the principles of their dynamic regulation. Revealing the PTM regulatory code presents a unique opportunity for the development of novel therapeutics. More broadly, our approaches may provide a new paradigm for addressing other complex biological questions involving PTM regulation.
Summary
Post-translational modifications (PTMs) of proteins are a major tool that the cell uses to monitor events and initiate appropriate responses. While a protein is defined by its backbone of amino acid sequence, its function is often determined by PTMs, which specify stability, activity, or cellular localization. Among PTMs, ubiquitin and ubiquitin-like (Ubl) modifications were shown to regulate a variety of fundamental cellular processes such as cell division and differentiation. Aberrations in these pathways have been implicated in the pathogenesis of cancer. Over the past decade high-throughput genomic and transcriptional analyses have profoundly broadened our understanding of the processes underlying cancer development and progression. Yet, proteomic analyses and the PTM landscape in cancer, remained relatively unexplored.
Our goal is to decipher molecular mechanisms of Ubl regulation in cancer. We will utilize the PTM profiling technology that I developed and further develop it to allow for subsequent MS analysis. Together with cutting-edge genomic, imaging and proteomic technologies, we will analyze novel aspects of PTM regulation at the level of the enzymatic machinery, the substrates and the downstream cellular network. We will rely on ample in-vitro and in-vivo characterization of Ubl conjugation to:a. Elucidate the regulatory principles of substrate specificity and recognition. b. Understand signalling dynamics in the ubiquitin system. c. Reveal how aberrations in these pathways may lead to diseases such as cancer. Identifying both the Ubl modifying enzymes and the modified substrates will form the basis for deciphering the molecular pathways in which they operate in the cell and the principles of their dynamic regulation. Revealing the PTM regulatory code presents a unique opportunity for the development of novel therapeutics. More broadly, our approaches may provide a new paradigm for addressing other complex biological questions involving PTM regulation.
Max ERC Funding
1 500 000 €
Duration
Start date: 2016-05-01, End date: 2021-04-30
Project acronym VASNICHE
Project The vascular stem cell niche and the neurovascular unit
Researcher (PI) Eliahu Keshet
Host Institution (HI) THE HEBREW UNIVERSITY OF JERUSALEM
Call Details Advanced Grant (AdG), LS4, ERC-2012-ADG_20120314
Summary Recent evidence suggests that VEGF and the vasculature play multiple roles in organ homeostasis, functions extending far beyond their traditional roles in tissue perfusion. The proposed study represents a vascular-centred approach to the neurovascular unit thriving to gain further insights on the many ways by which blood vessels may affect proper brain functioning. Major focus is on the vascular stem cell niche, i.e. the contention that blood vessels are a key component of adult stem cell niches, including a niche securing proper function of neuronal stem cells (NSCs). Further insights on the niche are also critical for contemplated implementation of stem-cell based therapy. In this multidisciplinary study combining the fields of vascular biology, neurobiology, stem cell biology, and aging research, we harness unique transgenic methodologies to conditionally manipulate (via VEGF) the vasculature within the stem cell niches. We provide a first compelling proof that blood vessels at the niche indeed control stem cells properties and behaviour, evidenced by showing that mere expansion of the niche vasculature and independently of VEGF) increases dramatically adult hippocampal neurogenesis, a process known to be associated with improved cognitive performance. We will determine what aspects of stem cell biology are controlled by juxtaposed, directly contacting blood vessels and will identify signalling systems mediating the vascular/stem cell cross-talk.
Adult neurogenesis is known to rapidly decline with age and ways to sustain the process are highly desired. We hypothesize and, in fact, provide initial evidence that expanding and 'rejuvenating' the niche vasculature can override the natural age-dependent decline of adult neurogenesis. Proposed experiments will extend this exciting finding and thrive to uncover the underlying mechanisms.
Summary
Recent evidence suggests that VEGF and the vasculature play multiple roles in organ homeostasis, functions extending far beyond their traditional roles in tissue perfusion. The proposed study represents a vascular-centred approach to the neurovascular unit thriving to gain further insights on the many ways by which blood vessels may affect proper brain functioning. Major focus is on the vascular stem cell niche, i.e. the contention that blood vessels are a key component of adult stem cell niches, including a niche securing proper function of neuronal stem cells (NSCs). Further insights on the niche are also critical for contemplated implementation of stem-cell based therapy. In this multidisciplinary study combining the fields of vascular biology, neurobiology, stem cell biology, and aging research, we harness unique transgenic methodologies to conditionally manipulate (via VEGF) the vasculature within the stem cell niches. We provide a first compelling proof that blood vessels at the niche indeed control stem cells properties and behaviour, evidenced by showing that mere expansion of the niche vasculature and independently of VEGF) increases dramatically adult hippocampal neurogenesis, a process known to be associated with improved cognitive performance. We will determine what aspects of stem cell biology are controlled by juxtaposed, directly contacting blood vessels and will identify signalling systems mediating the vascular/stem cell cross-talk.
Adult neurogenesis is known to rapidly decline with age and ways to sustain the process are highly desired. We hypothesize and, in fact, provide initial evidence that expanding and 'rejuvenating' the niche vasculature can override the natural age-dependent decline of adult neurogenesis. Proposed experiments will extend this exciting finding and thrive to uncover the underlying mechanisms.
Max ERC Funding
2 499 980 €
Duration
Start date: 2013-03-01, End date: 2018-02-28
Project acronym Virocellsphere
Project Host-virus chemical arms race during algal bloom in the ocean at a single cell resolution
Researcher (PI) Asaf Vardi
Host Institution (HI) WEIZMANN INSTITUTE OF SCIENCE
Call Details Consolidator Grant (CoG), LS8, ERC-2015-CoG
Summary Phytoplankton blooms are ephemeral events of exceptionally high primary productivity that regulate the flux of carbon across marine food webs. The cosmopolitan coccolithophore Emiliania huxleyi (Haptophyta) is a unicellular eukaryotic alga responsible for the largest oceanic algal blooms covering thousands of square kilometers. These annual blooms are frequently terminated by a specific large dsDNA E. huxleyi virus (EhV).
Despite the huge ecological importance of host-virus interactions, the ability to assess their ecological impact is limited to current approaches, which focus mainly on quantification of viral abundance and diversity. On the molecular basis, a major challenge in the current understanding of host-virus interactions in the marine environment is the ability to decode the wealth of “omics” data and translate it into cellular mechanisms that mediate host susceptibility and resistance to viral infection.
In the current proposal we intend to provide novel functional insights into molecular mechanisms that regulate host-virus interactions at the single-cell level by unravelling phenotypic heterogeneity within infected populations. By using physiological markers and single-cell transcriptomics, we propose to discern between host subpopulations and define their different “metabolic states”, in order to map them into different modes of susceptibility and resistance. By using advanced metabolomic approaches, we also aim to define the infochemical microenvironment generated during viral infection and examine how it can shape host phenotypic plasticity. Mapping the transcriptomic and metabolic footprints of viral infection will provide a meaningful tool to assess the dynamics of active viral infection during natural E. huxleyi blooms. Our novel approaches will pave the way for unprecedented quantification of the “viral shunt” that drives nutrient fluxes in marine food webs, from a single-cell level to a population and eventually ecosystem levels.
Summary
Phytoplankton blooms are ephemeral events of exceptionally high primary productivity that regulate the flux of carbon across marine food webs. The cosmopolitan coccolithophore Emiliania huxleyi (Haptophyta) is a unicellular eukaryotic alga responsible for the largest oceanic algal blooms covering thousands of square kilometers. These annual blooms are frequently terminated by a specific large dsDNA E. huxleyi virus (EhV).
Despite the huge ecological importance of host-virus interactions, the ability to assess their ecological impact is limited to current approaches, which focus mainly on quantification of viral abundance and diversity. On the molecular basis, a major challenge in the current understanding of host-virus interactions in the marine environment is the ability to decode the wealth of “omics” data and translate it into cellular mechanisms that mediate host susceptibility and resistance to viral infection.
In the current proposal we intend to provide novel functional insights into molecular mechanisms that regulate host-virus interactions at the single-cell level by unravelling phenotypic heterogeneity within infected populations. By using physiological markers and single-cell transcriptomics, we propose to discern between host subpopulations and define their different “metabolic states”, in order to map them into different modes of susceptibility and resistance. By using advanced metabolomic approaches, we also aim to define the infochemical microenvironment generated during viral infection and examine how it can shape host phenotypic plasticity. Mapping the transcriptomic and metabolic footprints of viral infection will provide a meaningful tool to assess the dynamics of active viral infection during natural E. huxleyi blooms. Our novel approaches will pave the way for unprecedented quantification of the “viral shunt” that drives nutrient fluxes in marine food webs, from a single-cell level to a population and eventually ecosystem levels.
Max ERC Funding
2 749 901 €
Duration
Start date: 2016-11-01, End date: 2021-10-31
Project acronym ZPR
Project The Pancreas Regulome: From causality to prediction of non-coding mutations in human pancreatic diseases
Researcher (PI) José Carlos Ribeiro Bessa
Host Institution (HI) INSTITUTO DE BIOLOGIA MOLECULAR E CELULAR-IBMC
Call Details Starting Grant (StG), LS2, ERC-2015-STG
Summary Several human pancreatic diseases have been characterized, being the diabetes the most common. Like others, this genetic disease is related to disrupted non-coding cis-regulatory elements (CREs) that culminate in altered gene expression. Although Genome Wide Association Studies support this hypothesis, it’s still unclear how mutations on CREs contribute to disease. The translation from the “non-coding code” to phenotype is an exciting and unexplored field that we will approach in this project with the help of the zebrafish as a suitable animal model. We aim to uncover the implications of the disruption of pancreas CREs and how they contribute to diabetes in vivo. For this we will study transcriptional regulation of genes in zebrafish. The similarities between zebrafish and mammal pancreas and the evolutionary conservation of pancreas transcription factors (TF) make it an excellent model to approach and study this disease. In this project we will characterize the zebrafish insulin producing beta-cell regulome, by determining the active CREs in this cell type and their bound TFs. Then we will compare this information with a similar dataset recently available for human beta-cells, to define functional orthologs in these species. Selected CREs will be tested by in vivo gene reporter assays in zebrafish, focusing on those functionally equivalent to human CREs where risk alleles have been associated with diabetes or those regulating genes involved in diabetes. Later these CREs will be mutated in the zebrafish genome to validate their contribution to diabetes. Finally we will translate this to predict new human disease-associated CREs by focusing on the regulatory landscape of diabetes-associated genes, without the need of having countless patients to uncover them. With this project we will create a model system that will allow the identification of new diabetes-associated CREs, which might have a great impact in clinical management of this epidemic disease.
Summary
Several human pancreatic diseases have been characterized, being the diabetes the most common. Like others, this genetic disease is related to disrupted non-coding cis-regulatory elements (CREs) that culminate in altered gene expression. Although Genome Wide Association Studies support this hypothesis, it’s still unclear how mutations on CREs contribute to disease. The translation from the “non-coding code” to phenotype is an exciting and unexplored field that we will approach in this project with the help of the zebrafish as a suitable animal model. We aim to uncover the implications of the disruption of pancreas CREs and how they contribute to diabetes in vivo. For this we will study transcriptional regulation of genes in zebrafish. The similarities between zebrafish and mammal pancreas and the evolutionary conservation of pancreas transcription factors (TF) make it an excellent model to approach and study this disease. In this project we will characterize the zebrafish insulin producing beta-cell regulome, by determining the active CREs in this cell type and their bound TFs. Then we will compare this information with a similar dataset recently available for human beta-cells, to define functional orthologs in these species. Selected CREs will be tested by in vivo gene reporter assays in zebrafish, focusing on those functionally equivalent to human CREs where risk alleles have been associated with diabetes or those regulating genes involved in diabetes. Later these CREs will be mutated in the zebrafish genome to validate their contribution to diabetes. Finally we will translate this to predict new human disease-associated CREs by focusing on the regulatory landscape of diabetes-associated genes, without the need of having countless patients to uncover them. With this project we will create a model system that will allow the identification of new diabetes-associated CREs, which might have a great impact in clinical management of this epidemic disease.
Max ERC Funding
1 497 520 €
Duration
Start date: 2016-06-01, End date: 2021-05-31