Li-Huei Tsai - US grants
Affiliations: | Massachusetts Institute of Technology, Cambridge, MA, United States |
Area:
mechanisms of Alzheimer's, neuronal migration and positioningWebsite:
https://tsailaboratory.mit.edu/li-huei-tsai/We are testing a new system for linking grants to scientists.
The funding information displayed below comes from the NIH Research Portfolio Online Reporting Tools and the NSF Award Database.The grant data on this page is limited to grants awarded in the United States and is thus partial. It can nonetheless be used to understand how funding patterns influence mentorship networks and vice-versa, which has deep implications on how research is done.
You can help! If you notice any innacuracies, please sign in and mark grants as correct or incorrect matches.
High-probability grants
According to our matching algorithm, Li-Huei Tsai is the likely recipient of the following grants.Years | Recipients | Code | Title / Keywords | Matching score |
---|---|---|---|---|
1998 — 2006 | Tsai, Li-Huei | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Signal Transduction Pathway Mediating Cdk5 Function @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): The mammalian cerebral cortex is an orderly laminated structure containing six distinct layers of neurons. This structure is constructed during cortical development involving programmed cell migration and positioning. Disruption of brain cytoarchitecture can result in severe consequences including epilepsy, mental retardation and early lethality. A small protein ser/thr kinase Cdk5 in conjunction with its regulatory partners, p35 and p39, plays an indispensable role in these processes. We and others showed that in the absence of Cdk5, neuronal migration is impaired resulting in cell positioning defects in the cerebral cortex, hippocampus, cerebellum and other hind brain structures. We have evidence to suggest that Cdk5 regulates actin and microtubule dynamics as well as cadherin mediated cell adhesion. These functions of Cdk5 may contribute to its role in neuronal positioning. Recently, we have obtained experimental evidence to suggest interactions of cdk5 with other known pathways regulating neuronal migration. Haplo-insufficiency of Lis 1 is responsible for type I lissencephaly, a disastrous neurological disorder in humans featuring abnormal neuronal positioning. We and others showed that mammalian Lis 1 interacts with cytoplasmic dynein and modulates dynein activity. Nudel is a novel protein that interacts with both Lis 1 and dynein. Interestingly, Nudel is a physiological substrate of cdk5 as phosphorylation of Nudel is diminished in brain extract lacking cdk5 kinase activity. The integrin receptors are implicated in migration of many different cell types. We found that cdk5 down regulates focal adhesions. Furthermore, the focal adhesion kinase (FAK) is phosphorylated on serine732 by cdk5. Phosphorylation of S732 is developmentally regulated and abolished in the p35/p39 compound mutant brain extract indicating that FAK is an endogenous substrate of cdk5. We hypothesize that phosphorylation of Nudel and FAK by cdk5 plays a regulatory role in neuronal migration and positioning and propose experiments to decipher the biological consequences of these phosphorylation events. Finally, we will make use of an in vitro system established in my laboratory to study and compare cdk5- and Lis1-dependent migration defects in living brain slices. These experiments should bring novel insight into the mechanism by which neuronal migration and positioning is regulated during cortical development. |
1 |
2001 — 2015 | Tsai, Li-Huei | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
@ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): Cyclin-dependent kinase 5 (Cdk5) is a multifaceted protein serine/threonine kinase with recently described roles in the pathogenesis of a number of neurological disorders, the best-characterized being Alzheimer's disease (AD). Aberrant Cdk5 activity has also been implicated in other neurological disorders, including Neiman Pick's Type C disease and ischemic brain injury. Two related neuron-specific proteins, p35 and 39, have been identified that activate Cdk5 upon direct binding. In the past decade, Cdk5 activity has been demonstrated to regulate many events during brain development including neuronal migration, axon, and dendrite development. Emerging evidence indicates pivotal roles for Cdk5 in synaptic plasticity, behavior, and cognition. Paradoxically, dysregulation of Cdk5 activity due to conversion of p35 to p25 by calpain is implicated in neurotoxicity and neurodegenerative diseases. The inducible p25 transgenic mice (CK-p25 Tg) exhibit hallmarks of AD including profound neuronal loss, reactive astrogliosis, tau pathology, and elevated beta- amyloid (Ab) peptides. In the past grant period, we made the unexpected finding that, while chronic p25 overexpression causes neurodegeneration and impaired synaptic plasticity, acute p25 overexpression in the CK-p25 mice enhances synaptic plasticity and facilitates learning and memory. This led us to the hypothesis that p25 has a physiological role in cognition and, when dysregulated, promotes neuropathological conditions. To understand the impact of Cdk5 dysregulation in neural disease, it is essential to elucidate the basic physiological functions of Cdk5 in the neuron. In this application, we will test the hypothesis that Cdk5 plays a key role in synaptic function and cognition, and that dysregulation of Cdk5 is detrimental to the nervous system. Using a multidisciplinary approach involving techniques of biochemistry, electrophysiology, and behavior, as well as taking advantage of a novel knock-in p35 mutant mouse, we will dissect the physiological and pathological roles of Cdk5 in synaptic plasticity and neurodegeneration, respectively. Completion of this project will lead to a greater understand the pathogenesis of Alzheimer's-like neurodegenerative diseases, and will elucidate promising targets for therapeutic intervention. |
1 |
2004 — 2006 | Tsai, Li-Huei | R03Activity Code Description: To provide research support specifically limited in time and amount for studies in categorical program areas. Small grants provide flexibility for initiating studies which are generally for preliminary short-term projects and are non-renewable. |
Fine-Tuning of Lis1-Nudel Interactions @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant) The long-term goal of the research supported by the paternal grant is to analyze developmental regulation of neuronal migration. The serine-threonine kinase cdk5, a focus of interest of the parental laboratory, is one of the key genes in this process. Ablation of cdk5, or its regulatory activators, causes disruption of mouse brain structure, thus identification of cdk5 substrates may provide us with molecules with crucial activities during brain development. Cdk5 interacts with several signaling networks involved in neuronal migration. Among several substrates, cdk5 phosphorylates Nudel, a LIS1/dynein interacting protein. Human brain organization is also disrupted in case of neuronal migration disorders, as lissencephaly (i.e. "smooth brain"), which may be the result of mutations in LIS1. The Reiner laboratory research is focused on the functional analysis of genes involved in lissencephaly. The premise that underlies the collaboration between Dr. Tsai's and Dr. Reiner's laboratory is that combining of knowledge and expertise from both laboratories can enhance significantly the research in each individual laboratory. The Reiner laboratory has gained expertise in understanding protein domains through computational and experimental analysis. The Tsai laboratory has integrated and methods of in vivo analysis to neuronal development. The specific objectives of the current grant are: 1. Fine mapping of the interaction between Nudel and LIS1: the Nudel interphase. 2. Fine mapping of the interaction between Nudel and LIS1: the LIS1 interphase. 3. Determination of the in vivo functions of LIS1 phosphorylation mutants that affect its interaction with Nudel, and Nudel mutants with aberrant interactions with LIS1, in migrating neurons in the cortex. The combination of skills from both laboratories will allow elucidating the molecular interphase of NudeI-LIS1 interactions and their importance during brain development. This research will be done primarily in The Weizmann Institute, Israel, in collaboration with Li-Huei Tsai as an extension of NIH grant # ROINS037007. [unreadable] |
1 |
2006 | Tsai, Li-Huei | R13Activity Code Description: To support recipient sponsored and directed international, national or regional meetings, conferences and workshops. |
2006 Gordon Research Conference On Molecular and Cellular Neurobiology @ Gordon Research Conferences [unreadable] DESCRIPTION (provided by applicant): Under the sponsorship of the Gordon Research Conferences, Drs. Li-Huei Tsai (Harvard Medical School) and Louis Reichardt (UCSF) are organizing a conference on Molecular and Cellular Neurobiology, to be held at Hong Kong University of Science & Technology, Hong Kong, China on June 11-16, 2006. Objectives The proposed GRC is organized with the following particular objectives: 1. To bring together a group of neuroscientists working at the forefront of molecular and cellular neurobiology and allow them to discuss in depth all aspects of the most recent advances in the field and to stimulate new directions of neuroscience research. 2. To promote interactions among neuroscientists from many nations over different continents. Particular emphasis will be placed between USA and Asian countries. The conference will provide a unique opportunity for scientists from North America, Europe and Asia to communicate recent scientific findings and to explore new areas of international collaborations. Furthermore, the conference will provide an opportunity for active interactions among scientists in relation to advancing the frontiers of molecular and cellular neurobiology in Asia and the Pacific region. Most of the Asian participants, especially those from China, would not have a chance to participate in this conference had it been held in the US, due to financial difficulties. Since 9/11, scientists from China also have difficulties obtaining entry visas to the US. The previous two GRC meetings in Hong Kong clearly served as a major platform for the Chinese trained graduate students and postdoctoral fellows to be exposed to world class neurobiologists. In keeping with the traditions of the prestigious GRC, all discussions in the week-long program will be informal and off-the-record to encourage open communication about frontier neuroscience research and developments. This "no disclosure" condition facilitates dissemination of information and ideas among the world's scientific community in a way that cannot be achieved through the usual channels of communication. [unreadable] [unreadable] [unreadable] |
0.901 |
2009 — 2010 | Tsai, Li-Huei | RC1Activity Code Description: NIH Challenge Grants in Health and Science Research |
Hdac1 Activating Compounds as Therapeutics For Neurodegenerative Disorders @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): This application addresses Broad Challenge Area (15) Translation Science and Specific Challenge Topic: 15-NS-103 Demonstration of "proof-of-concept" for a new therapeutic approach in a neurological disease. Alzheimer's disease (AD) is an irreversible neurological disorder that progressively attenuates the cognitive abilities of those afflicted, ultimately leading to death. AD is the most common of all neurodegenerative disorders, with an estimated 25 million victims worldwide. As life expectancies continue to rise, AD is becoming increasingly common, and it is estimated that the number of those afflicted with AD will increase to 114 million by the year 2050, and cost over $700 billion a year if nothing is done to curb the disease. Despite intensive studies, the pathogenesis of this illness remains to be elucidated and effective therapies still await discovery. Recent findings suggest that treatment of amyloid accumulation may be insufficient for treating Alzheimer's disease, and raise the possibility that scientific and corporate research efforts have been too narrowly focused on the amyloid aspect of Alzheimer's disease. Alternative therapeutic strategies may ultimately prove to be key. In particular, targeting fundamental mechanisms of cell survival and maintenance that are disrupted in Alzheimer's disease may lead to cures of other neurodegenerative disease as well, as these downstream events are often reported in multiple age-dependent neurodegenerative disorders. Importantly, we have recently shown that deregulation of HDAC1 may be critically involved in CNS pathology that may be relevant to stroke/ischemia and Alzheimer's disease. Using an inducible p25/Cdk5 neurodegeneration mouse model, we have observed that the catalytic activity of the histone deacetylase (HDAC1), a key regulator of epigenetic modifications, was inhibited by p25/Cdk5. Loss-of-function of HDAC1 in neurons through multiple means resulted in the accumulation of DNA damage, cell cycle activity, and neuronal death. Conversely, overexpression of HDAC1 resulted in a rescue against p25-induced DNA damage and neuronal death. These results suggest a role for HDAC1 in the maintenance of DNA integrity and cell cycle suppression in adult neurons. Furthermore, overexpression of HDAC1 resulted in significant rescue against DNA damage and neurodegeneration in a rodent stroke model, demonstrating therapeutic potential for HDAC1 gain-of-function. As p25 accumulation, cell cycle reentry, and DNA damage appear to be features shared in multiple neurodegenerative conditions, it is anticipated that HDAC1 gain-of-function may be a valid therapeutic strategy against Alzheimer's disease, stroke, and other disorders. We propose here to develop a "proof-of-concept" of this strategy by identifying small molecules through a high-throughput screen that can increase the deacetylase activity of HDAC1. Preliminary results suggest the feasibility of these efforts and the ability of such small-molecule probes to prevent neurotoxicity. Given that the proposed strategy is significantly different from any of the previous therapeutic strategies that are undergoing development or have been abandoned, there is tremendous opportunity for these studies to have a high impact on human health. PUBLIC HEALTH RELEVANCE: The overall goal of this project is to discover and characterize selective chemical activators of the HDAC1 that can be tested for the ability to prevent neurotoxicity. Findings from this application may offer novel therapeutic strategy against Alzheimer's disease, stroke, and other neurological disorders. |
1 |
2010 — 2014 | Tsai, Li-Huei | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Chemical Genomic Approaches to Neurobiology of Disc1 @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): Schizophrenia and bipolar disorder are chronic debilitating disorders that affect 1-3% of the population. The etiology of these disorders is completely unknown. Disrupted in Schizophrenia-1 (DISC1) was identified as the gene that was disrupted on chromosome 1 in a Scottish family with a high concordance of major psychiatric disorders and a balanced translocation between chromosomes 1 and 11. Karyotyping has been done on five generations of this family and 18 of the 29 members with this translocation have schizophrenia, recurrent major depression, or bipolar disorder. This type of near Mendelian segregation for psychiatric disorders indicates an integral role for DISC1 in maintaining mental health and that disruption of DISC1 functions plays a role in the etiology of psychiatric disorders. Indeed, DISC1 mouse models exhibit behavioral abnormalities reminiscent of human disease phenotypes such as hyperlocomotion, increased depressive-like behavior, decreased sociability and working memory. Several of these mouse models also display brain pathology indicative of impairment in brain development. Recently, we reported that DISC1 regulates the proliferation of neural progenitors during embryonic brain development and adult neurogenesis. Furthermore, we discovered that DISC1 regulated neural progenitor proliferation by positively regulating the Wnt signaling pathway by acting as an inhibitor of GSK32, a pleiotropic protein serine/threonine kinase that triggers the degradation of 2-catenin. In the dentate gyrus, DISC1 loss of function causes reduced proliferation of adult neural progenitors, which is accompanied by behavioral consequences including hyperactivity and increased depressive-like behavior. The adult neurogenesis defects and abnormal behavior were reversed upon treatment with a GSK3 chemical inhibitor. Our results indicate that DISC1 plays an important role in controlling GSK32/2-catenin activity, which in turn impacts neurogenesis and psychiatric-related behaviors. In this application, we propose to decipher how DISC1 mutant variants may alter the role of DISC1 in Wnt signaling and neurogenesis. We plan to take a multidisciplinary approach by performing studies with mouse cell and in vivo models, as well as including experiments using human cells to analyze Wnt signaling and neural progenitor development. Furthermore, we will use chemical-genetic approaches to identify and characterize small-molecule probes for conditional and selective modulation of the DISC1/GSK32-Wnt/2-catenin pathway. These studies will ultimately enable a better understanding of the relationship between human genetic variation in DISC1 and the effects on neurogenesis, as well as to lead to a better understanding of the causes and treatment of neuropsychiatric disease. |
1 |
2011 — 2015 | Tsai, Li-Huei | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
The Epigenetics of Alzheimer's Disease @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): Alzheimer's disease (AD) is an age-related neurodegenerative disorder associated with severe memory impairments for which, currently, there is no cure. Although the role of beta-amyloid (A[unreadable]) in the disease is strongly supported by genetic evidence, the mechanism between A[unreadable] and neurodegeneration/memory impairments is far from clear. In combating AD, it is imperative that we expand our approach beyond the current focus upon amyloid pathology. Research into novel therapeutic approaches to combat the symptoms of AD has revealed beneficial effects of increased chromatin remodeling and gene expression. We have shown that small molecule inhibitors of histone deacetylases (HDACs) restore learning ability in the CK-p25 mouse model of AD even after severe neuronal loss has occurred. The class I histone deacetylase, HDAC2, has been shown to participate in the regulation of hippocampal-dependent learning and memory. HDAC2 binds to the regulatory elements of genes implicated in synapse formation and synaptic plasticity, and is upregulated in both the CK-p25 and the 5XFAD mouse models of AD. These findings have led to the idea that, during neurodegeneration, an altered epigenetic landscape, mediated by HDAC2 up-regulation, may repress the expression of gene products necessary for maintaining synaptic plasticity and memory functions. Thus, inhibition of HDAC2, even after the onset of neurodegeneration, can improve the function of surviving neurons. In the current application, we will test the hypothesis that a novel disease mechanism, involving HDAC2 mediated alteration of the epigenetic landscape, underlies the cognitive impairment and synaptic dysfunction of Alzheimer's disease. PUBLIC HEALTH RELEVANCE: Alzheimer's disease (AD) is an age-related neurodegenerative disorder associated with severe memory impairments for which, currently, there is no cure. We have shown that small molecule inhibitors of histone deacetylases (HDACs) restore learning ability in the CK-p25 mouse model of AD even after severe neuronal loss has occurred. The current application will test the hypothesis that a novel disease mechanism, involving epigenetics, underlies the cognitive impairment and synaptic dysfunction of Alzheimer's disease. |
1 |
2012 | Haggarty, Stephen J Tsai, Li-Huei |
RF1Activity Code Description: To support a discrete, specific, circumscribed project to be performed by the named investigator(s) in an area representing specific interest and competencies based on the mission of the agency, using standard peer review criteria. This is the multi-year funded equivalent of the R01 but can be used also for multi-year funding of other research project grants such as R03, R21 as appropriate. |
Epigenomic Characterization of Alzheimer's Disease Neurons From Ipscs @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): Alzheimer's disease (AD) is the most common neurodegenerative disorder associated with dementia and, while other major causes of death have declined in the past ten years, the incidence of AD has risen alarmingly. In a mouse model of AD-like neurodegeneration, the CK-p25 mouse, we showed that alterations in chromatin remodeling, via the inhibition of histone deacetylase (HDAC) activity, ameliorated synaptic and cognitive impairments. Moreover, we found that the enhancement of cognitive function specifically required inhibition of the class I histone deacetylase, HDAC2, which plays a critical role in cognition by binding to the regulatory elements of genes implicated in synaptic plasticity and memory formation. Our preliminary data show that HDAC2 expression is induced upon neurotoxic stimulation, including A?42 and H2O2 treatments. Importantly, in postmortem human AD brains and in AD mouse models, HDAC2 shows a marked upregulation, which is accompanied by increased association of HDAC2 with memory genes and the drastic reduction in the expression of these genes. We propose to examine the mechanism underlying the HDAC2-mediated inhibition of plasticity genes in human induced pluripotent stem cell (iPSC)-derived neurons derived from control individuals as well as both familial and sporadic AD patients. We will examine the expression of HDAC2 mRNA and protein in these iPSC-derived neurons, and will PCR-sequence each sporadic AD line for the APOE genotype. We will then expose these neurons to neurotoxic stimuli, and will examine the epigenetic alterations in the control, familial, and sporadic AD neurons by conducting chromatin immunoprecipitation with antibodies against HDAC2, followed by Illumina second-generation sequencing. We will also examine changes in the transcriptome of these iPSC-derived neurons, with and without neurotoxic stimuli, by conducting RNA sequencing. The proposed studies will test the hypothesis that neurons derived from familial or sporadic AD patients are more susceptible to neurotoxic insults than those from controls, and that this heightened sensitivity results in an increased suppression of neural plasticity genes by HDAC2 upregulation. Importantly, we will also examine the effects of shRNA-mediated HDAC2 knockdown, as well as treatment with several HDAC inhibitor compounds, both known and novel, upon the HDAC2-mediated alterations in learning and memory gene expression in control, familial, and sporadic AD neurons with and without neurotoxic stimulation. The experiments outlined in this application will test the role of epigenetic alteration as a novel mechanism underlying AD-associated cognitive decline. PUBLIC HEALTH RELEVANCE: We have previously shown that class I histone deacetylase, HDAC2, responds to neurotoxicity by binding to the promoters, and suppressing the expression, of genes critical to normal learning and memory. The current proposal will use human neurons derived from patient induced pluripotent stem cells to test the hypothesis that neurons from Alzheimer's disease patients are more susceptible to neurotoxicity-induced chromatin remodeling via HDAC2. |
1 |
2014 | Tsai, Li-Huei | RF1Activity Code Description: To support a discrete, specific, circumscribed project to be performed by the named investigator(s) in an area representing specific interest and competencies based on the mission of the agency, using standard peer review criteria. This is the multi-year funded equivalent of the R01 but can be used also for multi-year funding of other research project grants such as R03, R21 as appropriate. |
Alzheimer's Disease Risk Genes in Human Microglia and Neurons Derived From Ipscs @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): The looming specter of 13.8 million Americans with Alzheimer's disease (AD) by the year 2050 motivates us to expand our biomedical research paradigm outside of the typical cell line to rodent to human trials model. The disappointing performance of all AD drugs that have come to Phase III clinical trials to date also forces us to think more creatively about how to study the mechanisms that underlie neurodegeneration. The advent of human induced pluripotent stem cells (iPSCs) allows us to create disease models from patients with sporadic AD as well as from those with defined familial mutations. In addition, we can use cutting-edge genome editing techniques, such as the Crispr/Cas system, to introduce disease-associated mutations into the genome of otherwise healthy human derived pluripotent cells. In the past five years, neuroscientists have made incredible advances in the creation of different brain cell types, such as neurons, astrocytes, and oligodendrocytes, from human-derived pluripotent cells. What is lacking, however, is a human cellular model of neuroinflammation, a critical component of all neurodegenerative disorders, including AD. In the current application, we describe the creation of human microglia, the brain's immune cell, from patient-derived and control pluripotent cells. We propose a comprehensive set of experiments that will determine the role that known and novel AD-associated genes play in these cells using cutting-edge genome editing techniques combined with high-throughput functional assays, transcriptomic profiling, and high-resolution proteomics. |
1 |
2014 | Tsai, Li-Huei Tye, Kay Maxine |
RF1Activity Code Description: To support a discrete, specific, circumscribed project to be performed by the named investigator(s) in an area representing specific interest and competencies based on the mission of the agency, using standard peer review criteria. This is the multi-year funded equivalent of the R01 but can be used also for multi-year funding of other research project grants such as R03, R21 as appropriate. |
Examination of Neural Circuits Underlying Mood Disorders in Alzheimer?S Disease @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): Recent research suggests that the presence of neuropsychiatric symptoms is a risk factor for progression from mild cognitive impairment (MCI) to Alzheimer's disease (AD), and an individual's susceptibility to distress significantly increases their risk of AD. These findings imply that poor resilience to behavioral stress is not simply a characteristic of dementia, but may reflect mechanisms involved in disease etiology. The increased activation of stress-related brain circuits, such as that between the basolateral amygdala (BLA) and the hippocampus, may underlie aspects of hippocampus pathology and exacerbate memory impairment in AD. However, the mechanistic link between behavioral stress, amygdala output, and hippocampal dysfunction in the normal and diseased brain remains unclear. Our preliminary results utilize optogenetic and pharmacogenetic techniques to show that activation of specific BLA afferents to the hippocampus mimics the effects of behavioral stress upon both cellular pathology and cognitive function. Importantly, silencing these pathways prevents cognitive impairment following repeated behavioral stress. Moreover, chronic inactivation of this circuit appears to ameliorate AD-like phenotypes in a mouse model of familial AD. Therefore, the BLA- hippocampus circuit, so heavily implicated in the impact of stress upon hippocampal function, should be closely evaluated, in a manner only achievable via the use of cell- and circuit-specific optogenetic techniques, for its contribution to cognitive dysfunction and cellular pathology in AD. Our preliminary data also show that the BLA- hippocampal stress circuit is not comprised of a solitary pathway, but that the ventral and dorsal components of this circuit may play differential roles in the modulation of anxiety and the impact of behavioral stress upon cognitive function. This application will test the hypothesis that the activation of BLA input pathways to the HPC as a result of behavioral stress leads to the exacerbation of AD pathology, and will determine the relative contribution of the ventral and dorsal components of this pathway. These studies will examine how the targeted silencing of specific brain circuits can slow disease progress and ameliorate cognitive impairment, and may provide rationale for the application of deep brain stimulation techniques in the treatment of AD. |
1 |
2014 — 2017 | Desimone, Robert (co-PI) [⬀] Tye, Kay (co-PI) [⬀] Wickersham, Ian [⬀] Tsai, Li-Huei |
N/AActivity Code Description: No activity code was retrieved: click on the grant title for more information |
Brain Eager: Cell-Type-Specific Optogenetics in Wild-Type Animals @ Massachusetts Institute of Technology This project consists of engineering a system for producing selective expression of light-inducible molecules in targeted neuron population in non-genetically modified animals of any species. The result will be a set of reagents that will be made freely available to the scientific community through nonprofit repositories and service centers. This new set of tools will enable the study of neural circuitry with greater resolution, power, and throughput than is currently possible, allowing major advances to be made in understanding the organization of the complex neural systems underlying perception, cognition, and behavior. This increased understanding could also result in improved artificial intelligence and machine learning. Finally, the future direct application of the technology in human patients holds promise for potentially treating conditions such as Parkinson's disease and epilepsy, by allowing the selective activation or inactivation of distinct components of the compromised neural circuitry that is associated with these disorders. |
1 |
2016 | De Jager, Philip L Regev, Aviv Tsai, Li-Huei |
RF1Activity Code Description: To support a discrete, specific, circumscribed project to be performed by the named investigator(s) in an area representing specific interest and competencies based on the mission of the agency, using standard peer review criteria. This is the multi-year funded equivalent of the R01 but can be used also for multi-year funding of other research project grants such as R03, R21 as appropriate. |
Demystifying Microglia in Aging and Alzheimer's Disease @ Massachusetts Institute of Technology Inflammation has become a well-recognized component of most neurodegenerative disorders, including Alzheimer's disease (AD). Until recently, we did not have the tools to reliably separate resident brain microglia from peripheral myeloid cells to delineate their functions in the brain. In this proposal, we will characterize different myeloid cell populations, such as resident brain microglia and peripheral monocytes, using elegant single-cell next-generation RNA sequencing (scRNA-Seq). We will determine the response of these neural immune components to physiological brain activity and neurodegeneration. Using novel models of mice with conditional targeting, we will manipulate AD risk genes specifically in brain microglia, and characterize the consequences, at both the single-cell and whole-animal level, in wildtype mice and mouse models of AD-like pathology. Importantly, we will compare these gene expression profiles to those obtained from over 500 well- studied healthy individuals and AD patients to assess the relevance of different monocyte phenotypes to the onset and progression of human AD. |
1 |
2016 | Kellis, Manolis (co-PI) [⬀] Tsai, Li-Huei |
RF1Activity Code Description: To support a discrete, specific, circumscribed project to be performed by the named investigator(s) in an area representing specific interest and competencies based on the mission of the agency, using standard peer review criteria. This is the multi-year funded equivalent of the R01 but can be used also for multi-year funding of other research project grants such as R03, R21 as appropriate. |
@ Massachusetts Institute of Technology Alzheimer's disease (AD) is an age-related neurodegenerative disorder associated with severe memory impairments for which, currently, there is no cure. Studies in human patients and mouse models reveal disease profiles that involve the downregulation of genes involved in neural functions, such as synaptic transmission, and the upregulation of immune response and inflammatory genes. Although there are a number of neural cell types implicated in AD risk and etiology, including neurons and different types of glia, the majority of studies thus far have utilized total tissue. While these have offered great insight into AD, the establishment of techniques for analyzing cell type-specific mechanisms underlying AD etiology is critically important. In the current proposal, we will utilize a combination of molecular techniques to label and isolate populations of specific cell types, including glia and neurons, and to generate cell type-specific epigenetic and transcriptomic maps from mouse models of neurodegeneration, postmortem human brain, and human adult stem cell-derived cultures. Using integrative bioinformatic analysis, we will determine the pathways disrupted during early and late neurodegeneration. We believe these studies will provide invaluable information about the gene expression and cellular programs that mechanistically underlie AD in the human brain, and will identify novel targets for therapeutic strategies. |
1 |
2016 — 2021 | Tsai, Li-Huei | R37Activity Code Description: To provide long-term grant support to investigators whose research competence and productivity are distinctly superior and who are highly likely to continue to perform in an outstanding manner. Investigators may not apply for a MERIT award. Program staff and/or members of the cognizant National Advisory Council/Board will identify candidates for the MERIT award during the course of review of competing research grant applications prepared and submitted in accordance with regular PHS requirements. |
@ Massachusetts Institute of Technology Cyclin-dependent kinase 5 (Cdk5) is a fascinating and enigmatic enzyme necessary for neuronal migration, synapse development, and synaptic homeostasis. To date, a large body of literature also supports the role of Cdk5 in numerous phenotypes associated with neurodegenerative disorders, including Alzheimer's disease (AD). Cdk5 is not catalytically active unless it is associated with a regulatory activator, such as p35 or p39. We and others showed that p35 can be cleaved by calpain under neurotoxic conditions, which leads to the generation of the p25 peptide. Various transgenic p25 mouse models exhibit neurodegeneration phenotypes such as Tau hyperphosphorylation, increased amyloid beta (A?), neuroinflammation, synaptic loss, neuronal loss, and memory impairments, demonstrating that the activity of the p25/Cdk5 kinase can be neurotoxic. In our last grant period, we created a mouse model harboring a calpain-resistant version of p35 (the ?p35KI mouse), and found that blocking p25 production abolished AD phenotypes in vivo. In this application, using the ?p35KI mouse model, we aim to determine the role of p25 generation in Tau-mediated neurodegeneration, as well as its role in neuroinflammation mediated by microglia. Furthermore, using genome editing in human induced pluripotent stem cells (iPSCs), we will determine whether p25 generation mediates AD-related pathology, including amyloid and Tau pathology, synaptic deficits, DNA damage, epigenetic dysregulation, endosome defects, and neuronal survival in human neurons. The overall hypothesis to be tested in this application is that p25-mediated Cdk5 dysregulation plays key roles in AD-like neurodegeneration. |
1 |
2017 — 2021 | Tsai, Li-Huei | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
@ Massachusetts Institute of Technology DNA damage perturbs genomic stability and has been linked to age-associated cognitive decline, as well as to early stages of various neurodegenerative disorders including Alzheimer?s disease (AD), amyotrophic lateral sclerosis, and frontotemporal dementia (FTD). However, our mechanistic understanding of how DNA damage contributes to neuronal vulnerability and deterioration remains an unresolved, yet extremely important question. A major confounding factor is that the sources of damage that are most pertinent to neurodegeneration remain unknown and the precise mechanisms that connect genomic instability to neurodegeneration are poorly understood. In addition, it is unclear whether the deterioration of brain function results solely from a random accumulation of DNA damage throughout the genome, or whether there are ?hotspots? of damage that mediate this process. The goal of our research is to better understand the mechanisms underlying genomic instability in neurodegeneration and identify novel therapeutic targets to dampen this early pathological hallmark of neuronal vulnerability. We hypothesize that genomic instability is a major underlying mechanism of cognitive decline and neuronal vulnerability in AD and FTD. Towards testing this hypothesis, our specific aims are: 1) to identify genomic loci that are vulnerable to the accumulation of DNA damage, particularly DNA double strand breaks (DSBs) in mouse and human induced pluripotent cell (iPSC)-derived models of AD and FTD, 2) to determine the precise defects in DSB signaling/repair in mouse and human iPSC-derived models of AD and FTD, and 3) to identify modifiers that reduce DNA damage susceptibility in iPSC-derived neural cells from patients with familial AD and FTD using a novel high-throughput screening strategy. Our preliminary findings suggest that excessive DNA DSBs are an early pathological hallmark of neurodegeneration that can be modeled in both mouse and human systems. Obtaining increased mechanistic insight into the failure to respond to and/or repair DNA DSBs in the context of neurodegenerative mutations will broaden our understanding of how genomic instability contributes to decline in brain health and cognition, and provide novel avenues for early therapeutic intervention in neurodegeneration. |
1 |
2017 — 2021 | Hyman, Bradley T. (co-PI) [⬀] Jaenisch, Rudolf (co-PI) [⬀] Kellis, Manolis [⬀] Tsai, Li-Huei |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Epigenomic, Transcriptional and Cellular Dissection of Alzheimer's Variants @ Massachusetts Institute of Technology Abstract The search for effective treatments for Alzheimer's disease (AD), the leading cause of late-onset dementia, has proven challenging. While recent successes in identifying more than a dozen new genes contributing to late-onset or sporadic AD (sAD) have generated considerable excitement in AD research, it is clear from large population studies including GWAS and whole-exome sequencing projects that many single nucleotide polymorphisms (SNPs) contributing to elevated sAD risk reside in non-coding intragenic or regulatory regions. The biological significance of these noncoding SNPs with respect to sAD pathogenesis is not clear. In the current application, we propose a scalable discovery platform for discerning which AD risk SNPs are associated with functional enhancers in specific neural cell types derived from human induced stem cells (hIPSCs). These hIPSCs, created from fibroblasts of sAD patients with a wealth of phenotypes that clearly lead to AD heterogeneity, will enable us to obtain a high-resolution map of AD risk SNPs associated with enhancers and their putative target genes in varied cell types. We will utilize CRISPR/Cas9/dCas9 technologies to directly determine the cell biological consequences of these AD risk genomic variants via 2D and 3D cytosystems. Our comprehensive strategy will identify novel genetic elements and unexpected regulatory pathways contributing to AD pathogenesis and progression that will lead to new therapeutic avenues. |
1 |
2018 | Jaffe, Jacob David Tsai, Li-Huei |
U54Activity Code Description: To support any part of the full range of research and development from very basic to clinical; may involve ancillary supportive activities such as protracted patient care necessary to the primary research or R&D effort. The spectrum of activities comprises a multidisciplinary attack on a specific disease entity or biomedical problem area. These differ from program project in that they are usually developed in response to an announcement of the programmatic needs of an Institute or Division and subsequently receive continuous attention from its staff. Centers may also serve as regional or national resources for special research purposes, with funding component staff helping to identify appropriate priority needs. |
@ Broad Institute, Inc. ABSTRACT The overarching goal of our LINCS project (U54HG008097) is to test the hypothesis that modulation of phosphorylation-mediated signaling events in response to perturbations can establish new cellular states by altering their phosphoproteomic and epigenetic landscapes. To achieve this goal, we propose performing mass spectrometry (MS)- based proteomic assays that specifically target quantitative readouts of phosphosignaling and chromatin modifications in cells on > 15,000 perturbational conditions. These perturbations will focus on modulation of signaling cascades and epigenetic marks by small molecules and genetic manipulations. We will study several different cellular model systems, including comprehensive studies of neuronal lineage differentiation starting from stem cells. We have established a center with the necessary infrastructure, pipelines, data management, and analytics required to perform the largest set of related experiments with MS proteomic read outs to date. We also employ next- generation MS acquisition technologies to establish a permanently minable MS data resource that will be accessible to the public. We contribute the resulting data and tools to the Library of Integrated Network-based Cellular Signatures (LINCS) program for the purpose of making connections among disparate perturbations through phosphoproteomic and chromatin modification signatures in concert with other data types to be contributed to LINCS by other centers. The resulting analyses help identify novel therapeutic opportunities and synergies, as dysregulation of phosphosignaling and epigenetic systems are two of the most common molecular etiologies identified in a growing number of genetic, developmental, and environmental diseases. In this supplement request we will study a unique model of Alzheimer's etiology derived from cell lines obtained from Down syndrome patients. We will test how cells with triplication of chromosome 21 respond differentially to kinase inhibitor stimuli from their otherwise isogenic disomic counterparts. We will measure aspects of signaling and epigenetics using proteomic analyses (P100 and GCP), and transcriptional responses using RNA-Seq and L1000 assays for maximum compatibility with the body of existing LINCS data. |
0.946 |
2020 | Blanchard, Joel William Kellis, Manolis (co-PI) [⬀] Tsai, Li-Huei |
UG3Activity Code Description: As part of a bi-phasic approach to funding exploratory and/or developmental research, the UG3 provides support for the first phase of the award. This activity code is used in lieu of the UH2 activity code when larger budgets and/or project periods are required to establish feasibility for the project. |
@ Massachusetts Institute of Technology Abstract Alzheimer's disease (AD) is a debilitating brain disorder, with staggering human and financial cost. While genetic studies are increasingly identifying polymorphisms that correlate with AD, there still is no clear picture of the molecular and cellular players and the extent to which each contributes to AD. The genetic and molecular complexity of AD and the lack of technology for experimentally unraveling it in human tissues create a bottleneck constricting the discovery of therapeutics and their successful translation into the clinic. Using human iPSCs we recently developed an in vitro blood-brain barrier (iBBB) and deployed it to discover mechanisms causing genetic predisposition to cerebral amyloid angiopathy (CAA). Identical to clinical studies, we found that APOE4, the strongest genetic risk factor for CAA and AD significantly increased amyloid deposition in our iBBB. The tractability of our engineered tissues then enabled dissection of the cellular causes of the disease. We found expression of APOE4 in pericytes alone was sufficient to increase cerebral vascular amyloid accumulation. Pinpointing the causal cells mediating CAA risk then enabled molecular and biochemical studies that established the underlying mechanism and revealed new therapeutic opportunities for mitigating genetic risk of CAA and potentially AD. Here, we will build upon our success, using the iBBB as a scaffold; we will incorporate neurons, oligodendrocytes, and microglia to generate a micro-integrated brain on a chip (miBrain-chip). In UG3 Aim1.1 we will establish miBrain-chips that represent healthy and diseased states of the human brain through iterative rounds of optimization that incorporate state-of-the-art biopolymers and engineering expertise from Robert Langer's lab at MIT. UG3 Aim1.2 will integrate and validate genetically encoded modulators and reporters of neuronal activity enabling the miBrain-chip to investigate how neuronal activity is influenced, and in turn, influences AD pathogenesis. UG3 Aim2 will model the pathological progression of AD in miBrain-chips across cohort of male and female sAD iPSC lines for which we have matched brains samples, clinical history, and genomic sequences. We will build computational models describing the transcriptional, cellular-dynamics and histological transformations that lead up to the end-states of post-mortem AD brains. These longitudinal pathological maps from genetically diverse healthy and sAD individuals will yield mechanistic insight into AD development and create a platform for discovery and efficacy screening of therapeutics. We hypothesize that the mechanisms underlying AD are significantly influenced by genetic variability. In UH3 we will establish the mechanisms underlying APOE4 pathogenesis (UH3 Aim1) and then ascertain the efficacy, toxicity, and therapeutic window of a panel of preclinical and clinical AD drugs using isogenic APOE3 and APOE4 miBrain-chips (UH3 Aim2). Our multimodal strategy will shed light on how genetic variation influences AD pathogenesis and therapeutic response, opening up new avenues for expeditious drug discovery and translation of effective therapeutics to the clinic. |
1 |
2020 | Kellis, Manolis [⬀] Tsai, Li-Huei |
U01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Single-Cell Transcriptional and Epigenomic Dissection of Alzheimer's Disease and Related Dementias @ Massachusetts Institute of Technology Dementia is a major public health problem with substantial personal, social, and financial burden, affecting more than 47 million people worldwide, with no cure to date. The major types of dementia include Alzheimer?s disease (AD), Lewy Body dementia (LBD), and frontotemporal dementia (FTD), which show distinct and overlapping pathological, neurological, and cellular signatures, but their detailed molecular signatures remain uncharacterized. Here, we systematically profile the molecular signatures of AD, LBD, FTD, and healthy aging, at the single-cell level, across traits, individuals, brain regions, cell types, age, sex, and disease severity. We use genetic, epigenomic, and transcriptional profiles, generating a total of ~1.5 million genome-wide maps at the single-cell (sc) level using scRNA-seq and scATAC-seq across 768 post-mortem brain samples from the Religious Order Study and Memory and Aging Project (ROS MAP) cohorts. We analyze the resulting datasets in the context of genetic variation from whole-genome sequencing, and phenotypic variation from rich longitudinal profiling and cognitive evaluations, enabling us to discover genes, control regions, pathways, cell types, and brain regions playing causal roles in AD and ADRD, and how they vary across age, sex, and traits. The resulting datasets will help guide the search for new therapeutics, by providing detailed therapeutic targets, and the specific conditions where they are predicted to act. |
1 |
2020 — 2021 | Kellis, Manolis [⬀] Tsai, Li-Huei |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Elucidating the Molecular Mechanisms of Neuropsychiatric Symptoms in Alzheimer's Disease @ Massachusetts Institute of Technology Abstract Alzheimer's disease (AD) is the most common cause of dementia, and the most common neurodegenerative disease worldwide, affecting 1 in 8 individuals over 65 years old in the US. Within AD, approximately 40-60% individuals are affected by psychotic symptoms (AD+P), which are associated with more rapid cognitive decline, greater disability, mortality and caregiver burden, resulting in a disproportionately large disease burden. Recent studies indicate a genetic basis for AD+P risk, but the molecular basis of AD+P remains largely uncharacterized, hindering the search for appropriate treatments and novel therapeutics. In this proposal, we seek to systematically dissect the mechanistic basis of AD+P by systematic generation, integration, and experimental dissection of transcriptional and epigenomic phenotypes across two brain regions and four cell types. (1) We profile single-cell RNA-seq and cell-type specific H3K27ac ChIP-seq across 192 post-mortem brain samples, each in two regions across AD patients with psychosis, AD patients with no psychosis, schizophrenia patients with no AD, and control individuals. (2) We integrate the resulting datasets with genetic information and GWAS data to predict driver genes, regions, variants, and pathways underlying AD+P using state-of-the-art machine learning methods for causality, mediation analysis, and genetic Bayesian fine- mapping. (3) We use our computational predictions to guide a systematic dissection of the molecular underpinnings of AD+P using a modular and programmable CRISPR-Cas9 methodology in iPSC lines to modulate regulatory elements, genes and alleles, and measure the resulting molecular and cellular phenotypes in cell-autonomous and non-autonomous phenotypes. If successful, this ambitious proposal has the potential to provide the first mechanistic insights on the development of psychotic symptoms in AD+P and/or P-AD, reveal functional risk variants and target genes for therapeutic intervention that will likely influence clinical management in order to alleviate the personal and societal burden associated with these disorders. |
1 |
2020 — 2021 | Goate, Alison M (co-PI) [⬀] Ray, William J Tsai, Li-Huei |
U01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Development of Pu.1 Inhibitory Modulators as Novel Therapeutics For Alzheimer's Disease @ Massachusetts Institute of Technology Project Summary/Abstract Alzheimer's disease (AD) is a fatal neurodegenerative disease with a global prevalence close to 50 million people, which is expected to double by 2040. Finding an effective treatment for AD has proven difficult, as evidenced by numerous high profile Phase 3 clinical trial failures, most of which directly target the reduction of ß-amyloid. Thus, it is becoming increasingly urgent to develop new pharmacological strategies to combat AD. Drugs are twice as likely to successfully negotiate the drug development pipeline and obtain FDA approval when their targets are supported from human genetic studies of disease. Human genetic studies have revealed a critical role for microglia involvement in Alzheimer?s disease progression, and it has recently been discovered that the transcription factor PU.1 is a driver of the pro-neurodegenerative phenotype adopted by microglia during aging and disease. This proposal therefore aims to develop novel, newly-discovered PU.1 Inhibitory Modulators (PIMs) for preclinical development, with the long term goal of clinically testing the hypothesis that reducing PU.1 activity in microglia will safely delay the age of AD onset (AAO) in at-risk populations. The studies in this proposal leverage the interdisciplinary structure of the Neurodegeneration Consortium, a unique collaboration between basic science researchers and industry drug development veterans operating under a collaborative agreement to push forward novel therapeutics aimed at treating Alzheimer?s diseaes and other neurodegenerative diseases. Under Specific Aim 1, the in vivo safety and efficacy of PIMs will be determined in mouse models of AD. Under Specific Aim 2, parallel target engagement studies will be performed to identify the target of PIMs, and the identified targets will be used to develop assays to determine the efficacy of PIMs in AD and in ex vivo models. Under Specific Aim 3, selected PIMs will be optimized using PK/PD and ADMET screening to develop lead tool compounds into candidate compounds suitable for future Phase I studies. The combined biology, chemistry, and pharmacology expertise in the Neurodegeneration Consortium, spanning The University of Texas MD Anderson Cancer Center, the Massachussets Institute of Technology, and the Mt. Sinai School of Medicine, make this group of researchers ideally suited to execute the proposed aims. |
1 |
2021 | Tsai, Li-Huei | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
@ Massachusetts Institute of Technology Alzheimer's disease (AD) is an incurable brain disease, distinguished by the progressive accumulation of toxic amyloid and tau protein aggregates that are partly due to impaired waste clearance by the glymphatic and meningeal lymphatic systems. We have recently shown that noninvasive Gamma ENtrainment Using Sensory stimuli (GENUS) to induce neural oscillations in the gamma frequency range (30-90 Hz) could ameliorate pathology in various AD mouse models. Mice subjected to GENUS regime exhibited positive effects on microglia, astrocytes and the brain vasculature as well as reduced accumulation of amyloid and hyperphosphorylated tau in respective amyloid and tauopathy mouse models. However, the impact of GENUS on the glymphatic/lymphatic systems in the clearance of amyloid and tau accumulation is not clear. We will use amyloid and tauopathy mouse models to determine whether and identify the mechanisms by which GENUS enhances paravascular fluid movement and thereby promotes meningeal lymphatic drainage and glymphatic clearance of brain toxic metabolites including those associated with amyloid and tau. |
1 |
2021 | Kellis, Manolis [⬀] Tsai, Li-Huei |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
@ Massachusetts Institute of Technology Abstract Alzheimer?s disease (AD) is a devastating neurodegenerative disorder that leads to dramatic effects on the affected individuals and their families. While the characterization of the genetic contribution to AD and underlying molecular mechanisms have advanced the understanding of the disease in recent years, studies show that sex differences account for much of the observed differences in risk, progression, and severity across individuals. Here, we directly dissect the contribution of sex-specific variation down to the region- specific and cell-type-specific molecular basis by systematic profiling, computational integration, and experimental validation of the transcriptional, epigenomic, and genetic signatures across individuals, brain regions, and cell types. In Aim 1, we use genetic, epigenomic, and transcriptional profiles, generating millions of single-cell (sc) level maps using scRNA-seq and scATAC-seq across human and mouse samples of varying ages and genetic risk status. In Aim 2, we analyze the resulting datasets in the context of known AD genetic risk variation and underlying molecular mechanisms, enabling us to discover and converge variants, regulatory regions, genes, pathways, cell types, and brain regions to functional, causal mechanisms that drive sex-related differences. In Aim 3, we use our well-established mouse and iPSC models to test our predicted mechanisms with both high-throughput and cell-type specific assays. The resulting datasets, computational predictions, and experimentally-supported mechanisms will shed light on the sex-related differences of AD and will help deepen our understanding the disease in general as we develop more personalized therapeutic approaches in treating AD. |
1 |