MIT Computational Biology Group - Funding

We have received generous support from:
  • the National Institutes of Health (NIH)
  • the National Human Genome Research Institute (NHGRI),
  • the National Institute of Aging (NIA)
  • the National Institute of Mental Health (NIMH)
  • the National Institute of Neurological Disorders and Stroke (NINDS)
  • the National Institute of General Medical Sciences (NIGMS)
  • the National Institute on Drug Abuse (NIDA)
  • the National Cancer Institute (NCI)
  • the National Science Foundation (NSF) (DBI program, DEB program)
  • the Sloan Foundation
  • the Cure Alzheimer's Fund (CureAlz)
  • the Human Frontiers Science Program
  • EMBO
  • Novo Nordisk
  • Takeda
  • Pfizer
  • the Shering Foundation
  • the Paris Kanellakis fellowship
  • Portugal
  • MIT
  • the Broad Institute.

    Current Support - AD / Schizophrenia / Cancer / Metabolism / GWAS / Immune

    Single-cell multi-region dissection of AD-pathogen interactions for HSV-1 and CMV
    NIH R01 AG081017 (PI: Manolis Kellis, Rudolph Emile Tanzi)
    February 01, 2023-January 31, 2028

    Alzheimer's disease (AD) is a 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 has advanced the understanding of the disease in recent years, studies have failed to find definitive mechanisms that account for disease progression. The influence of pathogens on AD potentially mediates an environmental impact on the established genetic contributions to AD. Here, we directly dissect the contribution of pathogen-related effects down to the cell-type-specific molecular basis by systematic profiling, computational integration, and experimental validation of the transcriptional signatures across individuals, brain regions, and cell types. In Aim 1, we use scRNA-seq in human, mouse, and human iPSC brain organoid samples of AD that are infected with Herpes Simplex Virus 1 or Cytomegalovirus (HSV-1/CMV) to generate millions of single- cell (sc) level maps with the end goal of a transcriptional atlas. In Aim 2, we analyze the resulting datasets and underlying molecular mechanisms, enabling us to discover and converge genes, pathways, cell types, and brain regions to functional and causal mechanisms that drive pathogen-related alterations. In Aim 3, we use our well-established iPSC model 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 pathogen-related influences on AD pathology and will help deepen our understanding of the disease in general as we develop more personalized therapeutic approaches to treating AD.



    Mapping the vulnerable locus coeruleus pathways in aging and AD
    NIH U01 AG077227 (PIs: Kwanghun Chung, Li-Huei Tsai)
    August 15, 2022-July 31, 2027

    Alzheimer's disease is a devastating dementia with no known cure. While research has advanced our knowledge of the genetics and molecular biology of AD, it is not yet known why some areas of the brain are affected, while others are spared. Additionally, the sensitivity of circuits and synaptic connections in disease progression are not known. We will examine the connectivity of the locus coeruleus (LC), which projects to most areas of the brain, and is one of the regions to show pathology earliest in AD. We will characterize the populations of these neurons based on their connectivity and sensitivity to degeneration with aging and in AD model mice, both from a global connectome level and with single-cell approaches for molecular signatures. Additionally, we with look in more detail at the entorhinal cortex, one of the recipients of LC connections with preferential cell loss, and characterize the specific populations and input/output relationships in response to aging and LC pathology.



    Single-Cell Dissection of Ensembles and Cell Types Mediating Opioid Action in the Rodent Brain
    NIH R01 DA054584 (PI: Myriam Heiman, Veronica A Alvarez, Manolis Kellis)
    June 01, 2021-February 28, 2027

    Substance use disorder (SUD) is a debilitating condition characterized by compulsive use of a substance, in spite of the subjective recognition of the drawbacks associated with its use. Chronic use of the substance leads to the development of withdrawal and dependence, hindering intentional controls over its use. Among all substances, opioids are among the ones that pose the greatest negative impact on our society. Animal and human studies have implicated several brain regions involved in opioid use disorder (OUD), in particular various components of the dopamine system. Major dopamine-containing neurons are clustered in regions of the midbrain called the substantia nigra pars compacta (SNc) and ventral tegmental area (VTA). Classically, their activities correlate with valence, e.g., promoting approach behavior to rewarding stimuli. Upstream of them, two major circuits are involved: one including the nucleus accumbens projecting onto the VTA, and the other including the dorsal striatum projecting onto the SNc. The scope of this study is thus to conduct in-depth transcriptomics across the cells of these two circuits to dissect genes, pathways, and cell types mediating opioid action in the mouse brain. We will use these data to predict driver genes, regulatory regions, pathways, and cell types involved in the emergence of opioid addiction behaviors. Finally, to test the causal role of these predicted drivers, we will use opioid self-administration, a classical paradigm used in the field of addiction. By combining a rich set of behavioral protocols with state-of-the-art transcriptomics, epigenomics, and computational data analysis, we aim to obtain multi-modal data with an unprecedented level of single-cell resolution for identifying genes, pathways, and cell types affected in mouse models of OUD.



    VCID: Single-cell multi-region transcriptional and epigenomic dissection of VCID
    NIH R01 NS129032 (PI: Kellis, Heiman)
    2022-2027

    Alzheimer's disease (AD) and AD-related dementias (ADRDs) are major drivers of mortality, morbidity, and health care costs for patients and their loved ones, due to the aging population, lack of predictive diagnosis, and lack of effective treatments or prevention. Vascular contributions to cognitive impairment and dementia (VCIDs) are key contributors to AD and ADRDs and manifest through diverse cerebrovascular lesions, including atherosclerosis, microinfarcts, and small vessel strokes. VCIDs include cerebral small vessel disease (CSVD), cerebral amyloid angiopathy (CAA), and a monogenic familial form of CSVD (CADASIL, Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy). Understanding the mechanisms and drivers of VCID will enable new biomarkers and therapeutics, similar to the success of addressing cardiovascular disease and hypertension in heart disease. To understand the cellular mechanisms underlying VCID across brain regions, cell types, pathology, and molecular pathways, we perform high- resolution profiling of epigenomic and transcriptional alterations in post-mortem CNS samples from both sporadic and genetic VCID patients. Aim 1: We profile single-nucleus RNA- sequencing (snRNA-seq) and DNA accessibility (snATAC-seq) to create a transcriptional and epigenomic atlas of VCID across diagnoses, brain regions, cell types, sexes, and individuals. Aim 2: We create an atlas of SVD-associated changes in genes, modules, pathways, and cell-cell interactions. Aim 3: We predict candidate driver genes, regulators, and pathways using causality analyses across temporal and genetic models, and we validate our results experimentally using imaging studies. The successful execution of our studies will delineate clinically relevant VCID biomarkers and therapeutics across sporadic and genetic VCID, enabling us to dissect their common and distinct molecular circuits, across four affected CNS region and all major cell types within them, and capturing an unprecedented level of complexity and enabling rich computational comparisons. The datasets generated and the computational analyses will provide invaluable insights for addressing the pressing medical need of VCIDs, their temporal, region-specific, and cell-type-specific changes, which can help guide new therapeutics.



    scOpioidsHIV: Single Cell Transcriptomic and Epigenomic Dissection of Opioid and Cocaine Responses in HIV (MIT-SCORCH)
    NIH U01 DA053631 (PI: Kellis, Heiman, Co-Is: Gabuzda, Madras, Ruzicka, Yu)
    2021-2026

    Substance use disorder (SUD) is a common and debilitating condition characterized by compulsive use of an addictive substance, inability to control the use of this substance, and the emergence of withdrawal symptoms in the absence of the substance. Despite great advances in our understanding of changes that occur in various brain regions in the context of addiction/SUD, there is a limited understanding of the diversity of cell types and gene expression profiles of cells within these human brain regions, and how exposures to addictive substances such as opioids and cocaine influence the molecular properties and functions of these cells. Over 50% of HIV- infected patients experience neurological disorders collectively termed HIV-Associated Neurocognitive Disorders (HAND), which ranges from the asymptomatic neurocognitive impairment to severely disabling dementia. The use of addictive substances by HIV-infected individuals has been linked to diminished immune function, increased neuroinflammation and neuronal injury, and exacerbation of HAND. Here, we seek to directly dissect the common and distinct molecular bases of SUD/HIV infection/HAND effects on distinct cell types by systematic profiling, dissection, computational integration, and experimental validation of their transcriptional, epigenomic, and genetic signatures across individuals, brain regions, and cell types. Aim 1: We use genetic, epigenomic, and transcriptional profiles, generating a total of ~28 million genome-wide maps at the single-cell (sc) level using 2,800 samples with 10,000 cells per sample; these span 7 brain regions (prefrontal cortex, nucleus accumbens, ventral tegmental area, dorsal striatum, insula, amygdala, hippocampus), two assays (scRNA, scATAC), four phenotypic groups (SUD+/HIV+, SUD+/HIV-, SUD-/HIV+, SUD-/HIV-), and a total of 200 subjects (50 in each phenotypic group). Aim 2: We integrate these datasets to predict driver genes, regulatory regions, variants, and pathways, and the cell types and brain regions where they act. Aim 3: We validate high-priority findings at the molecular level, and functionally validate the highest priority targets' ability to modulate addictive behaviors in mouse models of in vivo cocaine self-administration. 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.



    ALS/FTLD-TDP43mod: Identification of TDP-43 modifiers through single-cell transcriptional and epigenomic dissection of ALS and FTLD-MND
    NIH R01 NS127187 (PI: Donnelly, Kellis, Belzil, Heiman)
    2021-2026

    Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are two fatal neurodegenerative conditions with no current treatment to prevent, decelerate or stop neuronal death in patients. ALS and FTLD are clinically distinct but show an overlap in postmortem brain pathology and genetic factors: nuclear clearance and cytoplasmic accumulation of TDP-43 in affected central nervous system (CNS) regions is observed in 98% of ALS and 50% of FTLD patients. While initial symptoms lead to the diagnosis of either ALS or FTLD, up to 50% of ALS patients eventually develop symptoms of FTLD, with ~15% of patients ultimately receiving both diagnoses (FTLD with motor neuron disease, FTLD/MND). Mutations in the gene encoding TDP-43 (TARDBP) lead to rare cases of ALS, while TDP-43 pathology is observed in patients carrying more prevalent mutations, such as a pathological C9orf72 hexanucleotide repeat expansion (C9orf72+)- the most common genetic cause of ALS and FTLD identified thus far. TDP-43 therefore appears to be a pivotal and convergent factor in the pathogenesis of both ALS and FTLD. Despite this, however, the reasons for selective vulnerability of motor neurons, the mechanisms responsible for TDP-43 mislocalization, and the impact on neuronal health of nuclear TDP-43 exclusion and aberrant liquid-liquid phase separation underlying cytoplasmic demixing remain unknown. To address this challenge, in Aim 1, we systematically profile the transcriptional and epigenomic alterations of ALS and FTLD/MND patients at single-cell resolution using post-mortem CNS samples. In Aim 2, we integrate the resulting datasets to study the link between genetic, epigenomic, transcriptional, and cellular signatures of ALS and FTLD/MND. We associate these links with available clinical information, elucidate the genes and biological pathways altered in each, and predict new therapeutic targets. In Aim 3, we validate the molecular and cellular effects of these targets by assessing their impact on neuronal viability and TDP-43 functions/aggregation using high-throughput directed perturbation experiments. We study both cell-autonomous and non-cell-autonomous effects of these perturbations in human dura fibroblast-derived iPSC neurons and astroglia. In Aim 4, we perform neuropathological analyses of TDP-43 modifiers in ALS and FTLD/MND postmortem tissues, and endeavor to rescue in vivo pathology and phenotypes in a mouse model. The resulting datasets, analyses, and dura-derived iPSCs will provide an invaluable resource to understand the mechanisms of TDP-43 pathology in ALS and FTLD/MND, and may reveal putative therapeutic targets able to mitigate TDP-43 pathology through genetic manipulation.

    SexAD: Single-cell epigenomic and trancriptional dissection of sex-specific differences in Alzheimer's Disease
    NIH R01 AG074003 (PI: Kellis, Tsai)
    2021-2026

    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.



    Single-Cell Transcriptional and Epigenomic Dissection to Identify Therapeutic Targets for ALS and FTD
    NIH R01 AG067151 (PI: Manolis Kellis, Veronique Belzil)
    March 01, 2021-November 03, 2025

    Frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS) are devastating and fatal neurodegenerative diseases that strike middle-aged adults just as they reach full familial, financial and career potential. Initially thought to be quite distinct, FTLD and ALS are now recognized to share many clinical, pathological, and genetic signatures, but the mechanistic basis of their shared and distinct circuitry remains unknown at the molecular level. Genome-wide association studies (GWAS) have uncovered multiple common weak-effect variants, but the vast majority are non-coding, making it difficult to identify their target genes and the cell types where they act. To address this challenge, in Aim 1, we systematically profile the transcriptional and epigenomic alterations of FTLD and ALS patients at single-cell resolution using post-mortem brain samples. In Aim 2, we integrate the resulting datasets to study the link between genetic, epigenomic, transcriptional, and cellular signatures of FTLD and ALS, and to study the common and distinct genes and pathways altered in each, to predict new therapeutic targets. In Aim 3, we validate the molecular and cellular effects of these targets using high-throughput directed perturbation experiments and both cell-autonomous and non-autonomous phenotypes guided by our predicted pathways, and we disseminate all our results to the community. The resulting datasets, analyses, and validated targets will provide an invaluable resource to understand the mechanisms of action of FTLD and ALS, and the common and unique circuitry towards new therapeutic targets.



    Single-cell multi-region transcriptional and epigenomic dissection of VCID.
    NIH RF1 NS129032 (PIs: Manolis Kellis, Myriam Heiman)
    September 15, 2022-August 31, 2025

    Alzheimer's disease (AD) and AD-related dementias (ADRDs) are major drivers of mortality, morbidity, and health care costs for patients and their loved ones, due to the aging population, lack of predictive diagnosis, and lack of effective treatments or prevention. Vascular contributions to cognitive impairment and dementia (VCIDs) are key contributors to AD and ADRDs and manifest through diverse cerebrovascular lesions, including atherosclerosis, microinfarcts, and small vessel strokes. VCIDs include cerebral small vessel disease (CSVD), cerebral amyloid angiopathy (CAA), and a monogenic familial form of CSVD (CADASIL, Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy). Understanding the mechanisms and drivers of VCID will enable new biomarkers and therapeutics, similar to the success of addressing cardiovascular disease and hypertension in heart disease. To understand the cellular mechanisms underlying VCID across brain regions, cell types, pathology, and molecular pathways, we perform high- resolution profiling of epigenomic and transcriptional alterations in post-mortem CNS samples from both sporadic and genetic VCID patients. Aim 1: We profile single-nucleus RNA-sequencing (snRNA-seq) and DNA accessibility (snATAC-seq) to create a transcriptional and epigenomic atlas of VCID across diagnoses, brain regions, cell types, sexes, and individuals. Aim 2: We create an atlas of SVD-associated changes in genes, modules, pathways, and cell-cell interactions. Aim 3: We predict candidate driver genes, regulators, and pathways using causality analyses across temporal and genetic models, and we validate our results experimentally using imaging studies. The successful execution of our studies will delineate clinically-relevant VCID biomarkers and therapeutics across sporadic and genetic VCID, enabling us to dissect their common and distinct molecular circuits, across four affected CNS region and all major cell types within them, and capturing an unprecedented level of complexity and enabling rich computational comparisons. The datasets generated and the computational analyses will provide invaluable insights for addressing the pressing medical need of VCIDs, their temporal, region-specific, and cell-type-specific changes, which can help guide new therapeutics.



    Construction of an integrated immune-vascular Brain-chip as a platform for the study, drug screening, and treatments of Alzheimer's Disease
    NIH UG3 NS115064 (PI: Tsai, Blanchard, Kellis)
    2019-2024

    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.



    AD-endosomes: Dissection of Endosomal Trafficking Mechanisms in Alzheimer's Disease
    NIH RF1 AG062377 (PIs: Li-Huei Tsai, Manolis Kellis)
    9/30/2018-6/30/2023

    Recent findings in the genetics underlying Alzheimer's disease (AD), the leading cause of late- onset dementia, suggest that AD pathophysiology is driven primarily by four biological processes: cholesterol metabolism, immune response, endosomal trafficking, and cytoskeletal systems. Among these, endosomal trafficking has received relatively less attention. Underscoring its potential importance to AD development is that a substantial number of variants associated with familial AD and sporadic or late-onset AD have direct or indirect functional connections to endosomal trafficking. However, there is currently no clear picture of the molecular and cellular players and to what extent their participation in endosomal trafficking contribute to AD pathophysiology. In this application, we propose to leverage our computational and experimental expertise to build upon our recent unexpected findings of the roles of AD risk variants in endosome trafficking to provide a detailed picture at the molecular, cellular, and gene-clusters network levels for how endosomal trafficking impacts AD pathophysiology. We integrate highly innovative in-house bioinformatics platform, with single-cell transcriptomics of human postmortem AD samples, and the powerful genetics of yeast and human induced stem cells (iPSCs) to identify, validate, and illuminate the biological consequences of AD risk endosomal genes. Our multimodal strategy will shed light on how disruptions in endosomal trafficking could contribute to AD pathogenesis, opening up new avenues for novel therapeutic targeting toward the search for effective treatments.



    AD-Psychosis: Elucidating the molecular mechanisms of neuropsychiatric symptoms in Alzheimer's disease
    NIH R01 AG062335 (PIs: Manolis Kellis, Li-Huei Tsai, Robert Sweet, Julia Kofler)
    Sep 30, 2018-May 31, 2023

    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



    Alana Down Syndrome Center
    Alana Foundation (PIs: Tsai, Amon, Boyden, Kellis)
    2019-2023

    The Alana Down Syndrome Center aims to deepen knowledge about Down syndrome and to improve health, autonomy and inclusion of people with this genetic condition. The Alana Center is multi-disciplinary, spanning labs and programs across MIT, and engages the expertise of scientists and engineers in a research effort to increase understanding of the biology and neuroscience of Down syndrome. The greater mission is to produce research and technology to give people with disabilities the possibility of developing greater social and practical skills, in order to enhance their participation in the educational system, in the workforce, and in community life.



    Past Federal Support

    AD-dissection: Epigenomic, transcriptional and cellular dissection of Alzheimer's variants
    NIH R01 AG058002 (PIs: Manolis Kellis, Li-Huei Tsai, Rudolf Jaenisch, Bradley Hyman)
    June 15, 2018-May 31, 2022

    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



    Cancer-exosomes: Blood-Based Markers of Response to Immunotherapy in Melanoma
    DOD support (PI: Boland, Kellis)
    2018-2022

    Despite decades of cancer research, sustained investment, and many new medical advances, cancer remains one of the two leading causes of death in the United States with 40 percent of men and women diagnosed with cancer at some point during their lifetimes. One of the most promising recent therapeutic avenues has been cancer immunotherapy: boosting the immune system or training it against tumor antigens. However, only a fraction of patients receive durable clinical benefits from immunotherapy. To remedy these limitations, there is a pressing need to identify factors predictive of clinical response to help make patient-specific treatment choices, understand the mechanisms of both de novo and acquired immunotherapy resistance, and nominate new therapeutic targets for combination therapy. We believe analysis of cell-free DNA (cfDNA) and circulating microvesicles (exosomes) in parallel with existing tumoral data will allow us to identify signatures reflective of response/non-response to checkpoint blocking antibodies, and track signals arising from both the tumor and the tumor microenvironment in patients with metastatic melanoma. We propose multi-level analysis of peripheral blood-derived (PB-derived) cfDNA methylation patterns and exosomal RNA expression in parallel with separately funded analysis of tumors and associated immune cells from patients undergoing checkpoint blockade therapy. We hypothesize that blood-based analysis will allow us to interrogate multiple tumor sites simultaneously, offering a more broad-based analysis of the tumor and immune landscape than tumor analysis alone. Additionally, tissue-of-origin analysis by cfDNA methylation during the course of treatment will allow us to gain a comprehensive understanding of changes in the tumor microenvironment during checkpoint blockade immunotherapy.



    ADRD: Single-cell transcriptional and epigenomic dissection of Alzheimer's disease and related dementias
    NIH U01 NS110453 (PIs: Manolis Kellis, Li-Huei Tsai)
    Sept 30, 2018-Aug 31, 2021

    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.



    AD-complexity: Cell type specific epigenetic analysis to understand complex mechanisms underlying Alzheimer's disease phenotypes
    NIH RF1 AG054012 (PI: Li-Huei Tsai, Manolis Kellis)
    07/01/2016-06/30/2021

    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.



    GENCODE: Comprehensive genome annotation for human and mouse
    NIH U41 HG007234 (PI: Flicek, Harrow, Gerstein, Kellis, et al)
    April 1, 2013-May 31, 2021

    The objective of the GENCODE consortium is to create a foundational reference genome annotation, in which all gene features in the human and mouse genomes are identified and classified with high accuracy based on biological evidence, and then to release these annotations for the benefit of biomedical research and genome interpretation. GENCODE aims for a better understanding of a `normal' human genome; using genome sequences of the most commonly used mouse strains will facilitate the most effective use of these key models for large-scale knockout analysis and disease-specific research. To produce regular annotation releases of high accuracy, GENCODE will continue to follow its well-established and conservative research design, supplemented by targeted investigations into the value of new technologies, new data and new sources of evidence. GENCODE focuses on protein-coding and non-coding loci, including their alternatively spliced isoforms and pseudogenes. Over the course of this proposal GENCODE will follow major directions in genomics, including graph- based genome representations, long-read transcriptome sequencing, connecting genes and the associated regulatory regions that affect their transcription, and identifying genes that are not present on the current reference assembly. The GENCODE consortium has four fundamental components: (1) a comprehensive gene annotation pipeline leveraging manual annotation; (2) an integrated approach to pseudogene identification and classification; (3) a set of computational methods to evaluate and enhance gene annotation; and (4) complementary experimental pipelines for validation and functional annotation. More specifically, in the next four years GENCODE aims to (1) extend the human and mouse GENCODE gene sets to as near completion as possible given current experimental technology; (2) deploy population-based genome annotation to ensure that any transcript isoform expressed in an individual human will be present in the reference annotation set; (3) extend the gene annotation to include core regulatory regions and tissue-specific enhancers from selected datasets; (4) to distribute GENCODE annotations and engage with community annotation efforts. Current popular distribution channels for GENCODE data including the GENCODE web site, the Ensembl and UCSC Genome Browsers, will be maintained. Finally, new mechanisms for prioritizing genes for manual annotation with community input will be established, with the long-term aim of establishing GENCODE as the standard annotation set for research and clinical genomics efforts.



    Schizo: Network-based prediction and validation of causal schizophrenia genes and variants
    NIH R01 MH109978(PIs: Manolis Kellis, Kevin Eggan)
    May 1st, 2016 - February 28, 2021

    The recent increase in GWAS discovery power for psychiatric disorders has led to the recognition of an undisputed genetic basis for schizophrenia (SZ). However, the mechanistic basis of the vast majority of these loci remains uncharacterized, hindering the ability to translate genetic findings into novel drug targets and develop new treatments for SZ patients. In this proposal, we overcome these challenges and seek to identify and characterize novel SZ driver genes and causal variants by combining computational and experimental methods, integrating systems-level information to prioritize individual genes and loci, and validating their gene- regulatory and cellular effects in 10 neuronal and 3 glial cell tyes derived from iPS cells. Aim 1: We infer gene co-expression networks and modules using multiple brain regions and developmental stages, and use them to predict schizophrenia driver genes based on their clustering in common networks/modules, and their linking to schizophrenia-associated loci using activity correlation, chromatin conformation and eQTLs. Aim 2: We search for schizophrenia-enriched modules of enhancer regions, discovered by clustering patterns of H3K27ac activity across brain regions, developmental stages, and individuals, using an iterative probabilistic framework for joint prediction of causal driver genes, variants, and regulators. Aim 3: We experimentally validate the gene- regulatory and neuronal/glial cellular phenotypes of predicted schizophrenia driver genes and variants in neuronal and glial cell lines based on targeted sequencing of heterozygous loci overlapping 800 putative driver genes and 10,000 putative causal variants, and systematic profiling of neuronal and glial phenotypes upon knockdown and knockout of 200 candidate genes and bidirectional CRISPR-Cas9 editing of 50 candidate causal variants. If successful, this ambitious proposal has the potential to reveal dozens of new target genes and variants associated with Schizophrenia, and open up new avenues for therapeutic development that may alleviate the personal and societal burden of schizophrenia in our lifetimes.



    ENDODE-DAC: ENCODE data analysis center
    NIH U24 HG009446 (PI: Zhiping Weng, Mark Gerstein, Manolis Kellis, et al)
    Feb 1, 2017-Jan 31, 2021

    The goal of the Encyclopedia of DNA Elements (ENCODE) project is to catalog all functional elements in the human genome through the integration and analysis of high-throughput data. We propose to continue the ENCODE Data Analysis Center (EDAC, DAC) which will provide support and leadership in analyzing and integrating data from the ENCODE project as well as work closely with other ENCODE groups including the Data Coordination Center. Our proposed DAC team (Zhiping Weng, Mark Gerstein, Manolis Kellis, Roderic Guigo, Rafael Irizarry, X. Shirley Liu, Anshul Kundaje, and William Noble) has expertise across a wide range of fields including transcriptional regulation, epigenetics, evolution, genomics and proteomics, regulatory RNA, biophysics, and computational biology, where they are the leaders in machine learning, statistical genetics, networks, and gene annotation. These investigators also have a history of successfully working collaboratively in large consortia, particularly with other ENCODE groups. Their publication records demonstrate their synergistic approach to producing high-impact science and useful resources that benefit the broader biomedical communities. The proposed DAC will pursue the following four aims: Aim 1. Analyze and integrate data and metadata from a broad range of functional genomics projects; Aim 2. Serve as an informatics resource by supporting the activities of the ENCODE Analysis Working Group; Aim 3. Create high-quality Encyclopedias of DNA elements in the human and mouse genomes; Aim 4. Assess quality and utility of the ENCODE data and provide feedback to NHGRI and the Consortium.



    Metabolism: Multi-omics integration of obesity and diabetes progression
    Novo Nordisk (PI: Kellis, Lynch)
    2019-2021

    Obesity is a multifactorial disease characterized by high energy intake and low energy expenditure. This preventable metabolic disorder is a major risk factor for non-communicable diseases such as cardiovascular diseases (CVD) and type-2 diabetes (T2D). These are considered major public health problems, as CVD is the leading cause of death and T2D affects 1 in 11 adults worldwide. Interestingly, obesity is rarely caused by isolated events but heavily depends on concerted anomalies in multi-dimensional processes. To understand how this interplay takes place we introduce a network-based framework to identify dynamic modules of pathogenesis. Here, we use tissue-specific and tissue-na�ve networks containing information about co-expression, functional associations, protein-protein-interaction and transcriptional regulation. By applying this to multi-omic data from human and murine models of obesity, we aim at uncovering organ-organ and immune cell-cell relationships during disease progress and environmental interventions. We further categorize these dynamic networks by integrating genes and regulatory elements associated with obesity, T2D and other metabolic traits. Finally, we use natural language processing to derive hierarchical maps with network-drivers by incorporating experimentally validated associations from Medline literature (~24 million articles). This network-based approach allows us to infer molecular variations across several organs and repurpose drug-target identification.



    GWAS-dissection: Interpreting non-coding variants using epigenomics, regulatory models, and validation experiments
    NIH R01 HG008155 (PI: Manolis Kellis)
    August 1, 2015 - July 31, 2020

    Despite the successes of genome-wide association studies (GWAS), important challenges remain that still limit their impact on human biology and medicine, especially for non-coding variants which remain poorly understood. In this proposal, we exploit recent advances (many pioneered by our group) to overcome these challenges and gain a systematic understanding of the role of non-coding variants in human disease and complex traits. First, we develop new statistical methods that utilize high-resolution regulatory annotations to predict disease-relevant tissues, chromatin states, and regulatory motifs, and to prioritize non-coding variants more likely to have regulatory effects within regions of genetic association using epigenomic state information, comparative genomic information, and regulatory motif analysis (Aim 1). Second, we develop a new Bayesian methods for linking regulatory regions to their upstream regulators and downstream target genes by integrating genetic information across all associated regions in the context of regulatory networks that link regulators and regulatory regions using their correlated activity, regulatory motifs, and expression quantitative trait locus (eQTL) information (Aim 2). Third, we validate our methods and predictions using massively-parallel enhancer assays to test the effect of large number of regulatory variants in isolation; using genome editing technologies to test the effects of regulatory variants in their endogenous context; and using cellular phenotypes and animal models to test the physiological effects of regulatory variants at the cellular and organismal levels (Aim 3), and use the results to refine our computational methods and models. Even though our experimental validations are only performed for a small number of traits and cell types that are amenable to such studies, our methods are general and will be applied to all genetic studies available through ongoing collaborations and public catalogs.



    PsychENCODE: Single cell transcriptional and epigenomic dissection of schizophrenia and bipolar disorder
    NIH U01 MH119509 (PI: Kellis)
    June 1, 2019-May 31, 2020

    Schizophrenia and bipolar disorder, referred to together as the "major psychoses," are both devastating illnesses that strike young adults just as they are approaching their full potential, often resulting in a lifetime of chronic and severe disability. While distinct clinical entities, postmortem brain studies show high overlap and common genetic factors and familial co-segregation suggest strong overlap at the molecular level and potentially shared mechanisms. Here, we seek to directly dissect their common and distinct molecular basis by systematic profiling, dissection, computational integration, and experimental validation of their transcriptional, epigenomic, and genetic signatures across individuals, brain regions, and cell types. We use genetic, epigenomic, and transcriptional profiles, generating a total of ~1.2 million genome-wide maps at the single-cell (sc) level using scRNA-seq and scATAC-seq across 576 post-mortem brain samples from the McLean hospital psychiatric and normal 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 mentlal disorders, and how they vary across age, sex, and traits. We validate our predictions using a new high- throughput experimental dissection pipeline based on programmable CRISPR-based genomic, epigenomic, and transcriptomic dissection. 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.



    GWAS-Smoking: Genetic association meta-analyses of smoking and drinking for the sequencing age
    NIH R01 DA037904 (PI: Scott Vrieze)
    May 1, 2015 - April 30, 2020

    Alcohol and tobacco use and addiction are leading causes of morbidity and mortality in the USA and worldwide. These behaviors do not occur in isolation. Individuals who drink alcohol excessively are much more likely to smoke, and those who smoke are more likely to drink, with family studies consistently finding that genetic predispositions contribute to these relationships. Inherited genetic variation plays a substantial role in these behaviors with genome-wide association meta-analyses detecting >10 loci associated with heaviness of smoking, smoking initiation and cessation, alcohol use, or alcohol dependence. Such studies have discovered small individual effects of common genetic variants on these complex behaviors, with no variants clearly affecting both smoking and drinking. Much larger samples are now available to discover additional common and rare loci associated with smoking and drinking behavior to provide insight into the biological bases of these behaviors. To do this, we have formed a consortium of many large participating studies with existing study samples. We will finely map the genome with improved imputation, rare variant genotyping arrays, and whole genome sequencing to provide the genomic resolution necessary to identify rare and common variants that contribute to smoking and alcohol use, as well as disentangle competing genetic mechanisms. We will develop novel methods to test for shared genetic etiology, or pleiotropy, to directly test whether any particular locus influences both smoking and drinking, and contributes to the correlation between these behaviors. Finally, we will functionally annotate these dense genomic maps to characterize the nature of discovered loci. The results of this project will stimulate a wide range of follow-up experiments to characterize discovered loci.



    RareVariants-GSAP: Powering whole genome sequence-based genetic discovery for common human diseases
    NIH U01 HG009088 (PI: Xihong Lin)
    5/2/2016-3/31/2020

    The coming NHGRI Centers for Common Disease Genomics (CCDG) and Centers for Mendelian Genomics (CMG) plan to generate whole genome sequencing (WGS) data on over 200,000 individuals. WGS will provide comprehensive and complete genetic data across coding and non-coding variation, presenting an unprecedented opportunity for discovery in the genetic analysis of human diseases. However, a lack of powerful analytic tools that fully realize the potential of these data has emerged as a bottleneck for effectively translating rich information contained in these massive WGS data into meaningful insights about human diseases. There is a pressing need to develop powerful and robust analytic methods for WGS that can accelerate genetic discoveries. To meet this need, we have assembled an interdisciplinary team of computational biologists, geneticists, and statisticians. Building on our extensive track record in sequencing studies, statistical genetics, functional analysis and computational biology, we will power the next round of genetic discoveries by (1) building a massive WGS control sample and developing the methods for incorporating these controls in studies of complex and Mendelian diseases; (2) creating more powerful statistical methods for rare variant analysis through the incorporation of functional and regulatory information and advanced statistical tools; (3) establishing methods to analyze multiple phenotypes to boost the power for association and understand how different phenotypes relate genetically. These methods will enhance our ability to identify novel associations across a wide range of genetic architectures, from Mendelian diseases driven by a strong acting allele to complex polygenic traits. Novel associations promise to lay the foundation for gaining new insight into the biological mechanisms driving disease and be the bedrock for precision prevention and medicine strategies. We will collaborate with the investigators of the Genome Sequencing Program, and will share the developed data resources, tools and methods with the community through user-friendly open source software and educational modules.



    Cancer-immune: Mechanistic dissection of tumor-immune interfaces by longitudinal systems profiling in metastatic melanoma
    Takeda Oncology (PI: Kellis, Boland)
    2018-2020

    Cancer immunotherapy has been the most promising therapeutic avenue in cancer for years, but only succeeds in 50% of patients. Failure of immune checkpoint therapy is in part driven by gene-regulatory alterations in both the tumor and immune cells in the tumor microenvironment, and specifically by epigenomic and transcriptional alterations that affect key immune-related pathways and the tumor-immune interface. Here, we will generate epigenomic, transcriptional, and immunological profiles from a longitudinal cohort of metastatic melanoma samples before and after checkpoint blockade immunotherapy. We use these multi-omics profiles to identify regulatory regions, genes, and transcriptional regulators predictive of differential response to blockade immunotherapy. Our approach will generate novel mechanistic insights into how tumors escape such treatments and provide new therapeutic targets to potentially increase blockade immunotherapy efficiency. In Aim 1, we carry out systems-level epigenomic and transcriptional profiling and modeling of metastatic melanoma immunotherapy response. We leverage a retrospective and prospective biobank of samples from melanoma patients undergoing checkpoint blockade immunotherapy at the Massachusetts General Hospital (MGH), for systems-level epigenomic and transcriptional profiling and mechanistic modeling of blockade immunotherapy success. In Aim 2, we carry out epigenomic, transcriptional, immune, and exosomal profiling to understand the tumor-immune interface. In Aim 3, we carry out systems-level modelling and computational integration. We anticipate that our study will not only validate many of the mechanisms that have been previously implicated in checkpoint blockade immunotherapy success, but also discover new testable hypotheses amenable to follow-up experimental dissection using functional genomics tools.



    ROSMAP-reserve: Epidemiologic study of neural reserve and neurobiology of aging
    NIH R01 AG017917 (PI: David Bennett)
    March 1, 2015-March 31, 2019

    The prevention of cognitive decline and dementia is a major public health priority. The Memory and Aging Project (MAP) has made considerable progress elucidating the complex relationships between risk factors, common neuropathologies and resilience markers that increase or decrease the rate of cognitive decline, and risk of dementia. In nearly 200 peer-reviewed manuscripts, we reported genetic, medical, experiential, and psychological factors associated with increased or decreased rates of cognitive decline and dementia risk. We reported that Alzheimer's, vascular, and Lewy body pathologies (AD, CVD, LBD) explain about 40% of the variation of cognitive decline, and that resilience markers (e.g., cortical presynaptic proteins and locus coeruleus neurons) were associated with a slower rate of decline accounting for 8% of the variation, and that many risk factors with little direct relation o pathologies or resilience markers accounted for >10% of the variation of decline, controlling for pathologies. Additional factors related to cognitive decline await discovery. The overall goal of the planned continuation of MAP is to discover additional proteins associated with the slope of cognitive decline, after accounting for the effects of common pathologies. We use the term "residual cognitive decline" to describe this innovative primary study outcome. Methods to interrogate the genome, and therefore, the proteome, have improved markedly over the past decade, making the discovery of proteins that underlie cognitive decline both timely and feasible. We plan to continue collecting clinical and post-mortem data on MAP participants. Aim 1, Discovery and Verification Phase, will combine "omics-wide" association studies with innovative integrative pathway analyses on genomic, epigenomic, and transcriptomic data from human brain from 500 MAP participants to discover proteins associated with residual cognitive decline; it will select 200 proteins for quantitation in human brain to verify that protein level i associated with residual cognitive decline. Aim 2, Validation Phase, will quantify the 200 proteins selected and verified in Aim 1 with a new sample of 350 brains from MAP participants that will accrue by the end the funding period, followed by a joint statistical analysis to increas power (n=850). Aim 3 will link the validated proteins to the wealth of available risk factor data t discover novel biologic pathways linking risk factors to cognitive decline and dementia. Our plan is supported by extensive and compelling new preliminary data that demonstrate its high likelihood of success and demonstrate that the study is innovative, high yield and low risk. The continuation of MAP will discover additional proteins associated with cognitive decline and novel biologic pathways linking risk factors to cognitive decline. These proteins will represent either unknown pathologies that increase rates of cognitive decline or resilience markers that decrease rates of cognitive decline. Both sets of proteins will offer new therapeutic targets for the prevention and treatment of cognitive decline and dementia. Thus, the study has the potential to have a high and sustained impact on aging and dementia research



    CureAlz-CIRCUITS: Consortium to Infer Regulatory Circuits and to Uncover Innovative Therapeutic Strategies
    CureAlz (PI: Kellis, Tsai, Tanzi, Pfenning, Bertram, Hide, Jaenisch, Ecker, Hyman, Bennett)
    2017-2019

    Alzheimer's Disease (AD) is a devastating neurodegenerative disorder, afflicting 1 in 3 dying seniors and costing $236 billion annually in the US alone. Its prevalence is increasing rapidly in an aging population, and there is currently no cure. Recent genetic studies provide new hope for therapeutic avenues, but translating genetic results into therapeutics has been remarkably difficult, due primarily to the fact that most genetic mutations do not alter protein function directly, but instead affect the expression of nearby genes in subtle ways. We propose to form the CureAD-CIRCUITS (Consortium to Infer Regulatory Circuits and to Uncover Innovative Therapeutic Strategies) consortium to leverage the combined expertise and resources of our laboratories in collecting and interpreting multi-omic data, uniquely positioning us to accelerate discovery of novel drug targets for AD.This collaboration will dramatically transform our ability to leverage large-scale genetic and genomic information for therapeutic development and provide a powerful new data driven model for therapeutic target discovery. At a time when costs of pharmaceutical development have skyrocketed, leading to very costly failures at later-stage clinical trials, we have the potential to identify new targets for therapeutic intervention that were previously not considered, and to increase the success rate of pharmaceutical development. This ambitious pilot collaboration will result in some of the most comprehensive epigenomic and functional dissection of disease genes, variants, cell types, and regulators, and transform the process of therapeutic discovery, and possibly the larger industry. The pipelines we create for functional analysis of disease variant dissection will easily generalize across diverse clinical indications and additional areas of strategic importance.



    ROSMAP-pathways: Pathway discovery, validation and compound identification for Alzheimer's disease
    NIH U01 AG046152 (PI: Philip De Jager)
    Sept 20, 2013-August 31, 2018

    The overall goal of the proposed study is the discovery and preclinical validation of novel targets associated with molecular processes that lead to cognitive decline, the primary outcome of most AD trials. The recent NIA Alzheimer's Disease Research Summit 2012: Path to Treatment and Prevention that led to the RFA to which we are responding, made a number of recommendations which motivated the strategy adopted in the proposed study. Our proposal brings together an exceptionally strong and unique multi-disciplinary team with the relevant skills needed to achieve our overall goal. In Aim 1, we take a systems biology approach to mine a truly unique set of deep clinical, paraclinical, pathologic, genomic, epigenomic, and transcriptomic data assembled from frozen dorsolateral prefrontal cortex brain tissue of 1000 subjects from two cohort studies of aging and dementia, the Religious Order Study and the Memory and Aging Project, in conjunction with other publicly available functional datasets. These unique data provide an excellent substrate for the identification and nomination of molecular targets for drug discovery. Target discovery is coupled with a flexible translational strategy that first validates targets by a targeted proteomic study of brain tissue from the same region and subjects in Aim 2. In Aim 3, an RNA interference (RNAi) and overexpression functional validation study in cultured human neurons and astrocytes is executed in parallel in Aim 3. Finally, these data come together in Aim 4 which performs high throughput small molecule screens on neurons and astrocytes derived from induced pluripotent stem cells (iPSC) on the most promising targets. Our proposal is ambitious but realistic: it reflects the deployment of cutting-edge approaches with a practical mindset in which redundancies have been carefully considered to mitigate risk and ensure the delivery of data, network models and lead compounds. The proposed study will discover and validate novel targets associated with molecular processes that lead to cognitive decline, and it will demonstrate the druggability of one or more targets, setting the stage for clinical trials with new and novel approaches to the prevention and treatment of AD.



    Roadmap-comparative: Comparative analysis and regulatory architecture of epigenomics datasets
    NIH 1 R01 GM113708 (PI: Manolis Kellis)
    April 1st, 2015 - March 31, 2018

    The NIH Roadmap Epigenomics and ENCODE projects have generated a collection of 3000+ epigenomics datasets, including histone modification, DNA methylation, gene expression, and DNaseI hypersensitivity profiled across 190 cell and tissue types. In order to maximize its impact on gene regulation, cellular differentiation, and human health, novel computational analyses are needed. To address this challenge, we will develop new methods for epigenomic analysis, building on our extensive experience interpreting epigenomic information, and our preliminary studies building chromatin states, activity clusters, and regulatory motif maps for the Roadmap Epigenomics and ENCODE datasets. In Aim 1, we will characterize epigenomic differences and changes during lineage differentiation by developing new tools for systematic comparison of groups of epigenomes that directly exploit the complexity of epigenomic datasets; we will also develop methods for clustering epigenomes into developmental lineages based on automatically-learned diverse epigenomic features that distinguish them; and methods that learn the unidirectional epigenomic changes that pluripotent cells undergo during lineage commitment to gain more insights into differentiation and automatically learn to classify lineages and differentiation trajectories. In Am 2, we will seek to characterize higher-order chromatin architecture and chromatin conformation to enable systematic interpretation of cis-regulatory modules: we will develop a novel statistical approach for enhancer-enhancer and enhancer-gene linking to reveal interacting regions and their target genes based on their coordinated activity patterns across cell and tissue types; we will train a supervised learning method for predicting both constitutive and tissue-specific chromatin conformation information based on chromatin state information, individual chromatin marks, genomic distance, activity, regulatory motif information, and DNA sequence; and we will use these higher-order interaction maps to predict gene expression levels based on the combined action of multiple regulatory regions and to define the cis-regulatory architecture of each gene in the human genome. The resulting resources will be invaluable for studies of gene regulation, by revealing the set of regulatory elements that are linked to each gene, and for the interpretation of genetic studies, by revealing the set of regulatory elements which jointly act to regulate each target gene and the potential target genes of non-coding variants associated with human disease.



    eGTEx: Epigenomic variation atlas across human tissues and individuals in GTEx
    NIH U01 HG007610 (PI: Manolis Kellis)
    June 2, 2014 - March 31, 2017

    The Genotype-Tissue Expression project (GTEx) is expanding the availability of primary tissue samples for studying the impact of genetic variation on gene expression across individuals and tissues. However, inter-individual variation in gene expression level can often result from the combined effects of multiple variants, each of which may affect the activity of different regulatory regions in complex ways, and also integrate complex post-transcriptional events. Thus, to directly assess the effect of sequence variants on regulatory elements, we propose to systematic profile epigenetic modifications on stored GTEx biospecimens to enhance the analysis of gene expression variation already available and planned by the GTEx consortium. Aim 1. We will use whole-genome bisulfite sequencing (WGBS) to profile DNA methylation levels for all 28M CpGs in the genome of 20 individuals in 8 GTEx tissues. We will use these to identify variable methylated regions (VMRs) across cell types and across individuals, to evaluate the effects of eQTL SNPs on DNA methylation levels of neighboring CpGs, and to recognize allele-specific methylation (ASM) regions and their tissue specificity. Aim 2. We will systematically profile DNA methylation levels across 250 individuals in the same 8 tissues using hybrid-selection bisulfite sequencing (HSBS) for a subset of 2M regions selected within enhancer-associated, eQTL-associated, and inter-individual variable regions. We will use these to predict methylation quantitative trait loci (meQTLs) in each tissue, relate these to GTEx expression QTLs (eQTLs) for the same tissues to provide mechanistic hypotheses for inter- individual gene expression variation, and to discover meQTLs for genes where no single eQTL is significant. Aim 3. We will use chromatin immunoprecipitation sequencing (ChIP-Seq) to profile enhancer- and promoter-associated histone modification H3K27ac in 6 GTEx brain samples across 100 individuals, to recognize genotype-associated enhancer regions and enhQTLs. We will relate these to eQTLs in the same tissues and meQTLs discovered using HSBS on enhancer-proximal and eQTL proximal CpGs to generate mechanistic models for regulatory changes associated with genotype variation. We will work closely with the GTEx Analysis Consortium to integrate GTEx eQTLs with our predicted meQTLs, ASM regions, and enhQTLs to relate regulatory region variation to gene expression variation and to genome-wide association studies to understand the role of sequence variation in tissue-specific gene regulation, gene expression, and human disease.



    ENCODE-Chromatin: Expanding the catalog of chromatin regulatory elements in the human genome
    NIH U54 HG006991 (PI: Bradley Bernstein)
    Sept 21, 2012 - Jul 31, 2016

    The human genome is regulated by hundreds of thousands of functional DNA elements that play pivotal roles in development and disease, yet remain poorly understood. Comprehensive identification and characterization of functional DNA elements is thus an essential goal with major implications for human health. Genomic DNA is organized into chromatin, a higher-order structure composed of DNA, RNA and proteins. Chromatin mapping technologies have emerged as a powerful means for identifying functional DNA elements, which are associated with characteristic chromatin structures. Mapping has enabled the systematic annotation of diverse types of functional elements, including promoters, enhancers and silencers, and new discoveries related to chromatin organization and genome regulation in health and disease. The proposed ENCODE data production center aims to vastly expand the catalog of functional elements in the human genome through production mapping of Protein, DNA and RNA constituents of chromatin. High-throughput pipelines will be used to map histone modifications, chromatin regulatory proteins and non-coding RNA binding interactions. These pipelines will be applied to designated ENCODE cell lines, embryonic stem (ES) cells and derivatives, induced pluripotent stem (iPS) cells and phenotypically diverse human tissues. Key histone modifications will also be mapped in representative cell and tissue types from multiple individuals, in order to address the extent and significance of inter-individual variation in chromatin landscapes and their relationships to genetic background. These multi-dimensional datasets will be integrated through innovative computational algorithms to identify sequences, motifs, variants and regulatory interactions that dictate chromatin state and functional element activity. Thus, the proposed project will dramatically increase the number, resolution and precision of functional DNA elements in the ENCODE catalog, and explicitly define causal sequences and physical interactions that mediate chromatin states and regulation in the human genome. All data will be rapidly released and made freely available to the scientific community



    ENCODE-DAC: EDAC: ENCODE Data Analysis Center
    NIH U41 HG007000 (PI: Zhiping Weng)
    Sept 21, 2012 - August 1, 2015

    The objective of the Encyclopedia of DNA Elements (ENCODE) Project is to provide a complete inventory of all functional elements in the human genome using high-throughput experiments as well as computational methods. This proposal aims to create the ENCODE Data Analysis Center (EDAC, or the DAC), consisting of a multi-disciplinary group of leading scientists who will respond to directions from the Analysis Working Group (AWG) of ENCODE and thus integrate data generated by all groups in the ENCODE Consortium in an unbiased manner. These analyses will substantially augment the value of the ENCODE data by integrating diverse data types. The DAC members are leaders in their respective fields of bioinformatics, computational machine learning, algorithm development, and statistical theory and application to genomic data (Zhiping Weng, Manolis Kellis, Mark Gerstein, Mark Daly, Roderic Guigo, Shirley Liu, Rafael Irizarry, and William Noble). They have a strong track record of delivering collaborative analysis in the context of the ENCODE and modENCODE Projects, in which this group of researchers was responsible for the much of the analyses and the majority of the figures and tables in the ENCODE and modENCODE papers. The proposed DAC will pursue goals summarized as the following seven aims: Aim 1. To work with the AWG to define and prioritize integrative analyses of ENCODE data; Aim 2.To provide shared computational guidelines and infrastructure for data processing, common analysis tasks, and data exchange; Aim 3. To facilitate and carry out data integration for element-specific analyses; Aim 4.To facilitate and carry out exploratory data analyses across elements; Aim 5.To facilitate and carry out comparative analyses across human, mouse, fly, and worm; Aim 6.To facilitate integration with the genome-wide association studies community and disease datasets; and Aim 7.To facilitate writing Consortium papers and assist evaluating ENCODE data. RELEVANCE: The Encyclopedia of DNA Elements (ENCODE) Project is a coordinated effort to apply high-throughput, cost-efficient approaches to generate a comprehensive catalog of functional elements in the human genome. This proposal establishes a data analysis center to support, facilitate, and enhance integrative analyses of the ENCODE Consortium, with the ultimate goal of facilitating the scientific and medical communities in interpreting this human genome and using it to understand human biology and improve human health.



    Phylo: Assembling the Tree of Life: Can Phylogenomcis Resolve Deep Phylogeny
    NSF DEB 0936234 - (PIs: Alm, Kellis)
    09/1/09-8/31/13

    Resolving the topology and relative timing of the deepest branches remains one of the most significant challenges in Assembling the Tree of Life. Knowledge of this deep phylogeny, however, is critical for understanding foundational questions in biology such as how the complex networks that underlie modern life arose, and how they have been shaped by co-evolution with the Earth. This research project will develop new algorithms to infer a Tree of Life that is based on the thousands of complete genome sequences currently available rather than a single gene or small set of genes. Accurately identifying and accounting for horizontal gene transfer, a process in which genes are exchanged between unrelated species, potentially obscuring phylogenetic relationships, is a key challenge that will be addressed. Calibrating the timing of the deepest branches on the Tree of Life with the geological record will be done in collaboration with experts in early Earth history and geochronology. This project will result in a better understanding of the origin and history of life on Earth, and new algorithms for inferring the evolutionary history of genes and species across billions of years of evolutionary time. The project will bring together expertise from diverse fields of study, and expose graduate, undergraduate, and postdoctoral students to interdisciplinary research. In addition, museum partnerships and podcasts will enable dissemination of research results to the general public.



    Motifs: Regulatory motif discovery in the human genome using comparative genomics
    NIH R01 HG004037 (PI: Manolis Kellis)
    10/01/07-09/31/121 (Sum)
    To realize the promise of the human genome project, we need not only the parts list of all the genes, but also a comprehensive understanding of how they function together. Along with genes, our genome contains all the signals necessary for controlling gene expression in response to environmental and developmental stimuli. These regulatory processes are governed by short sequence motifs, responsible for modulating gene usage at every level. Despite their prevalence, regulatory motifs have been particularly challenging to identify, due to their short length and the varying distances at which they can act. Given their extraordinary importance, their systematic understanding still remains one of the major challenges of modern biology. In the proposed work, we use comparative genomics of multiple mammals to systematically identify and characterize regulatory motifs in the human genome based on their evolutionary conservation. We have pioneered a new powerful approach for de novo motif discovery by using genome-wide conservation, and successfully applied it in four yeast genomes, twelve fly genomes, and human promoters and 3'-UTRs. Here we expand this methodology to undertake motif discovery across the entire human genome: (1) we develop methods that use dozens of mammalian species for motif discovery and characterization; (2) we identify significant motif combinations and grammars and reveal their functional roles; and (3) we discover functional regions of motif clustering and study motif role in specifying enhancer function. The proposed work is timely, given that NHGRI's sequencing efforts now encompass more than 30 mammalian genomes, specifically for understanding the human. Moreover, large-scale systematic experimentation is providing the functional information necessary to inform and validate our findings. By revealing the underlying sequence patterns that govern gene usage, we complement these ongoing efforts and provide access to the concrete building blocks of human gene regulation. This will enable researchers world-wide to link new genes in pathways by their co-regulation, elucidate the role of non- coding SNPs in regulatory diseases, and lead to new tests and therapeutics for modern medicine. A global map of regulatory motifs constitutes a necessary knowledge infrastructure towards a comprehensive understanding of regulation, development, and disease.


    NSF-CAREER: Comparative Genomics and Biological Signal Discovery in the Human Genome
    NSF DBI 0644282 (PI: Manolis Kellis)
    10/01/07-09/31/121 (Sum)
    Massachusetts Institute of Technology researcher Manolis Kellis is awarded a Faculty Early Career Development award to develop comparative genomics methods that can maximally use mammalian sequences for biological signal discovery and to systematically interpret the human genome. In Aim 1 comparative methods for ortholog identification will be developed that can scale to dozens of complete mammals, can account for the complex phylogenies relating them, gene duplication and loss, and varying rates of divergence across gene families, and across species. In a second aim a classification framework for gene identification will be constructed. Beyond the simple use of comparative genomics to recognize conserved regions, evolutionary signatures will be defined specific to protein-coding regions, based on patterns of insertion and deletion, codon mutational biases, and motif abundance in proximity of exons. Finally, phylogenetically-informed tools for motif discovery and enhancer identification will be developed. The study of rates of motif gain and loss will allow them to derive parameters for every lineage based on the information content of each motif. These parameters will then be used to discover motifs at the genome scale, tolerating motif movement and loss, based on a likelihood ratio of motif conservation over a branch length within a given window of tolerated movement. Further research will develop methods for identifying functional motif combinations, and use these to identify regions of motif clustering, and relate them with experimentally identified enhancer elements and other regions of regulatory importance. Functional information of expression, regulator binding, and chromatin structure from human tissues and cell lines, across the entire genome and in selected ENCODE regions, will provide training data for our methods, and validate datasets for the predictions. Two key model organisms will be the yeast Saccharomyces cerevisiae, and the fruit fly Drosophila melanogaster, which provide unique models for the computational and experimental efforts. Both benefit from large phylogenetic trees with many completely sequenced relatives, compact genomes, and extensive experimental information, which can inform the power of the methods. The ability to work with multiple model organisms will allows the study of each aspect of human biology at the appropriate scale of complexity, from yeast, to fly, mouse, and human, by virtue of the early shared ancestry of our common biology. The education plan involves developing new courses and curricula and a computational biology textbook. The tools produced by the project will be distributed freely. Already strong efforts to include underrepresented groups will continue along outreach activities, such as lectures at museums or working with high school teachers in the Boston area


    ENCODE-Chromatin: High-throughput sequencing of chromatin regulatory elements
    NIH U54 HG004570 (PIs: Bernstein, Kellis)
    09/30/07-09/30/110.3 (Sum)

    A key goal in characterizing the human genome is to acquire a complete catalog of chromatin regulatory elements and sequence determinants of chromatin state. In this application, we propose to undertake a large-scale project to map the genomewide locations of >35 histone modifications and related proteins in >20 human cell types by coupling chromatin IP with massive-throughput sequencing on the Solexa platform. Our preliminary data demonstrate that this approach is highly accurate and a major advance over existing technologies in terms of cell requirements, genome coverage, throughput and cost-effectiveness. A sequencing pipeline will be applied to generate more than 500 genomewide datasets of chromatin structure by sequencing >1000 samples. A computational pipeline will then convert the sequencing reads into high resolution, genomewide maps that can be visualized in genome browsers and used for downstream analysis. Comparative genomic analysis and motif-finding tools will be applied to classify genomic sites based on associated chromatin structures and to identify underlying sequence determinants. Cell and molecular biology methods will be used to validate inferred functions for a representative subset of discovered elements. Chromatin regulation is extensively implicated in many aspects of human development and disease. In particular, cancer cells may universally exhibit aberrant chromatin states. The proposed systematic identification and characterization of chromatin regulatory elements in the human genome will offer unprecedented insight into the structure and function of chromatin, and provide an invaluable resource for investigators in chromatin, genomics, cancer and many other fields of research. All data collected in the context of the proposed project will be made available pre-publication to the greater scientific community once reliability has been confirmed.



    GENCODE: Integrated human genome annotation: generation of a reference gene set
    NIH U54 HG004555-01 (PIs: Hubbard, Kellis, Gerstein, Reymond, Guigo, Haussler, Brent, Valencia)
    09/30/07-09/30/110.3 (Sum)
    The specific aim of this proposal is to annotate all the evidence-based gene features at high accuracy on the human genome reference sequence. This includes identifying all the protein-coding loci with associated alternative variants, non-coding loci which have transcript evidence available in the public nucleotide database (NCBI/EMBL/DDBJ) and pseudogenes. To achieve this goal we will integrate computational approaches, including recent comparative methods, expert manual annotation, able to integrate literature information, and targeted experimental approaches. Based on the exhaustive experimental and computation investigation of our initial GENCODE annotation of the ENCODE regions we are confident that we can deliver a gene set with high specificity and sensitivity that will provide critical information to other biologists and other ENCODE groups. As part of this process we will label all apparent gene loci clearly, classifying them according to their likely current functional status, so users are informed where regions that appear gene like are most likely pseudogenes or where transcript evidence is most likely artefactual. There are a number of motivated groups working in the area of defining protein coding genes for the human genome. This proposal includes most such groups and coordinates with other key groups. Critically, all the groups bring extensive experience of data integration and evaluation, leading to the resolution of annotation discrepancies by multiple approaches. This gives us confidence that through this integrated project we will be able to eliminate many of the remaining uncertainties about the precise location of genes and their component exons and transcript structure in the human genome. Genome-wide, highly accurate transcript definition will be of enormous value to the myriad of researchers working on the human genome. It will both have large cost savings worldwide due to increased specificity of reagent design and provide a more complete view of human genes, in particular those associated with disease. From this foundation, more accurate descriptions of the genetic causes of disease can be discovered.


    Roadmap-integration: Integrative analysis of genomic and epigenomic datasets in multiple cell types
    NIH RC1 HG005334 (PI: Manolis Kellis)
    09/30/09-09/29/111 (Acad)
    This application addresses broad Challenge Area (08) Genomics, and specific Challenge Topic, 08-OD-101, Computational approaches for epigenomic analysis. While the primary DNA sequence of the human genome is ultimately responsible for the encoding and functioning of each cell, a plethora of chromatin and DNA modifications have been described in recent years that can modulate the interpretation of this primary sequence. These epigenetic modifications lead to the diversity of function across different human cell types, and play key roles in the establishment and maintenance of cellular identity during development, and also in health and disease. The human ENCODE project, the NIH Epigenome Roadmap, and several other large-scale experimental efforts are currently underway to map dozens of histone and DNA modifications across multiple human cell types and disease states, generating a diversity of rich epigenomic datasets. This creates a pressing need for the development of rigorous computational methods for the systematic integrative analysis of epigenomic datasets, and for understanding their relationship to other genomic datasets, including gene expression, disease association, and phenotypic profiling. In this proposal, we will develop and apply graphical probabilistic models for describing chromatin modifications, based on multivariate hidden Markov models. We will use these models to discover the set of underlying chromatin state, based on recurrent combinations of epigenetic marks across the entire genome (Aim 1). We will validate and functionally characterize these states based on their enrichments and positional biases with respect to existing functional elements, as well as large-scale gene expression and disease association datasets (Aim 2). Lastly, we will extend these methods to study dynamics of chromatin state across both healthy and disease cell types, and study how these correlate with functional differences between the observed cell types (Aim 3). We will work closely with the scientists involved in data production and facilitate communication and data integration across them, and also with data analysis and coordination centers already established to facilitate sharing of methods and results across the ENCODE and Epigenome Roadmap consortia, and with the larger community. Overall, the proposed integrative analysis of large-scale genomic and epigenomic datasets will provide a unified view of current and planned epigenomic datasets, towards a systematic understanding of gene and genome regulation in health and disease. While the primary DNA sequence of the human genome is ultimately responsible for the encoding and functioning of each cell, a plethora of chromatin and DNA modifications have been described in recent years that can modulate the interpretation of this primary sequence, leading to the diversity of function across different human cell types. This project will create a computational framework and resource to integrate large-scale genomic and epigenomic datasets, to understand their functional role in health and disease, and to understand their dynamics across different cell lines and disease states. The knowledge gained can play key roles in understanding the establishment and maintenance of cellular identity during healthy development, and how dysregulation of these processes can lead to the onset of disease.


    modENCODE-DAC: A Data Analysis Center for integration of fly and worm modENCODE datasets
    NIH RC2 HG005639 (PIs: Kellis, Gerstein, Park, Liu, Gunsalus, Brenner, Bickel, Lai, MacAlpine, Henikoff)
    09/30/09-09/29/111 (Acad)
    The aims of the ENCODE (Encyclopedia of DNA Elements) and modENCODE (model organism ENCODE) projects are to apply high-throughput, cost-efficient approaches to generate a catalog of functional elements in the human, worm, and fly genomes, which will serve as the basis for biomedical research advances. By their smaller genome size, powerful genetics, and ease of experimentation, D. melanogaster and C. elegans can help guide the study of functional elements in the human genome, reveal new insights into global gene regulation and embryo development, and enable experimental studies of gene function and regulation which are not accessible in mammalian systems. This proposal aims to enhance the value of these datasets by creating a Data Analysis Center (DAC) to support, facilitate, and enhance integrative analyses of the modENCODE consortium in fly and worm, to achieve a high-resolution annotation of all their functional elements, and to reveal new insights into the biology and gene regulation of animal genomes including the human. We foresee four central roles for the DAC, and have organized our aims around them. Aim 1: We will provide common computational guidelines for data processing in fly and worm, a common computational infrastructure and pipeline for common analysis and statistical tasks. Aim 2: We will facilitate and carry out element-specific integrative analyses to identify diverse classes of functional elements based on combinations of relevant datasets coming from multiple groups. This includes (a) enhancers, promoters, insulators, and other regions of regulatory importance, (b) protein-coding and non-coding genes, (c) regulatory networks of transcription factor and microRNA targeting, and (d) sequence features predictive of diverse classes of functional elements. Aim 3: We will carry out exploratory data analyses across different data types to discover potentially novel correlations and insights relating diverse classes of elements. In particular we will apply dimensionality reduction techniques to coordinate-based genome-wide genomic and epigenomic datasets, we will apply clustering and bi-clustering methods to identify functionally related sets of genes and modules, and we will analyze structural and dynamic properties of discovered networks. Aim 4: We will carry out comparative analyses across the two model organisms, and also with yeast and human. We will provide an ortholog resource between the species, compare regulatory relationships and dynamics for orthologous cell lines and developmental points, and carry over biological knowledge across model organisms and human. To achieve these four aims, we will work closely with members of the consortium, the modENCODE Analysis Working Group (AWG), consisting of all Principal Investigators and analysis groups, and the Data Coordination Center (DCC), responsible for all data sharing within the consortium and with the larger worm and fly communities. PUBLIC HEALTH RELEVANCE: The aims of the ENCODE (Encyclopedia of DNA Elements) and modENCODE (model organism ENCODE) projects are to apply high-throughput, cost-efficient approaches to generate a catalog of functional elements in the human, worm, and fly genomes, which will serve as the basis for biomedical research advances. By their smaller genome size, powerful genetics, and ease of experimentation, D. melanogaster and C. elegans can help guide the study of functional elements in the human genome, reveal new insights into global gene regulation and embryo development, and enable experimental studies of gene function and regulation which are not accessible in mammalian systems. This proposal aims to enhance the value of these datasets by creating a Data Analysis Center (DAC) to support, facilitate, and enhance integrative analyses of the modENCODE consortium in fly and worm, to achieve a high-resolution annotation of all their functional elements, and to reveal new insights into the biology and gene regulation of animal genomes including the human.


    Chrom-RNA: Genome-wide chromatin modification targeting by endogenous small RNAs
    NIH RC1 CA147187 (PIs: Ronald Hart, Manolis Kellis)
    09/30/09-09/29/110.3 (Acad)
    This application addresses broad Challenge Area (08) Genomics, and specific Challenge Topic 08-CA-104: Regulatory functions of small RNAs. Our goal is to identify small RNAs (smRNAs) as regulators of histone modifying enzymes, mediating interactions with promoters and/or non-coding RNAs present in promoter proximal regions and mediating long-term effects on gene expression. This is a novel model that would tie together genome-encoded small RNAs, non-coding RNAs, and the regulation of histone modifications, leading to long-term modulation of gene expression. Epigenetic modifications are inheritable alterations to the genome in the form of enzymatic manipulations of key histone residues, or the methylation of specific cytosines in the DNA sequence. Carcinogenesis is likely to include aberrant use of this pathway to cause long-term changes in gene expression in transformed cells. In fact, epigenetic dysregulation is a hallmark of numerous cancers, and compounds which broadly affect histone modifications are under intense scrutiny as cancer therapies. While this strategy may be effective, we believe that it will be possible to therapeutically target epigenetic modulation of selected genes by exploiting the existing RNAi machinery, as demonstrated for several smRNAs and genes. This project aims to understand the relationship between small, non-coding RNA sequences and epigenetic histone modifications, and explore the mechanisms by which epigenome-modifying complexes may be recruited by small RNAs to their target DNA sequences. Comparison of smRNAs identified in human embryonic stem cells by deep sequencing with genomic alignment sites and previously published epigenomic data of key histone modifications has identified striking association between these two previously independent pathways. In this proposal we will test the hypothesis that smRNAs mediate the placement of epigenetic marks. Specifically, we will (1) identify and characterize classes of smRNA sequences associated with specific chromatin modifications in human ES Cells during neural differentiation; (2) bioinformatically predict as yet unobserved smRNA-mediated epigenetic effects; and (3) experimentally confirm the functional interaction between small non-coding RNAs and epigenetic modifications in hESC. Epigenetic marking is one of the next great questions in biology and medicine. We propose that endogenous smRNAs are the key link between histone modification mechanisms and individual genomic loci. If smRNAs do in fact direct epigenetic modifications, this may represent a novel mechanism by which cells are capable of transcriptional regulation. This will provide both additional therapeutic targets and enhanced understanding of disease etiologies for regions of altered histone modification during cancers. Public health relevance: Epigenetic dysregulation is a hallmark of several human disorders and numerous cancers. Histone modifications represent one mechanism of epigenetic regulation that directly affects the transcriptional availability of adjacent genomic elements. This project aims to identify the relationship between small, non-coding RNA sequences and epigenetic histone modifications, and explore the mechanisms by which epigenome modifying complexes may be recruited by small RNAs to their target genomic regions. The association of small RNAs with specific histone modifications provides understanding of disease etiologies and suggests mechanisms to transcriptionally regulate target genes in patients.


    modENCODE-TFs: A Cis-Regulatory Map of the Drosphila Genome
    NIH U01 HG004264 (PIs: White, Kellis, Ren, Posakony, Bellen)
    03/01/07-03/28/110.3 (Sum)

    Drosophila presents an ideal model organism for the study of human cis-regulation. Its genome shares the structure and major features of the human genome; all major families of transcription factors, both the basal machinery and site-specific factors; the overall regulatory structure of developmental genes such as the Hox clusters; many of the "master regulatory" transcription factor proteins such as PAX6/eyeless and Distalless that control organ or tissue identity. In many cases, human proteins function in Drosophila just as well as Drosophila proteins. In addition to the shared biology, the fly also offers powerful experimental tools not yet available for mammalian species. Its small genome size, at 130 Mb, is amenable to genome-wide systematic experimentation at a resolution not yet achievable in the human. Additionally, a total of 12 fully sequenced species provide one of the richest comparative genomic datasets, powerful enough to identify individual functional binding sites of regulators across the entire genome. Finally, the wealth of high throughput experimental techniques enables us to validate functional predictions in vivo. In this proposal, we exploit these unique features to produce a comprehensive cis-regulatory map of the Drosophila genome, using a combined experimental and computational approach. We will identify fly promoter regions, short and long range enhancers, insulator and repressor regions, and their defining characteristics based on cis-regulatory motifs and grammars, chromatin state, and transcription factor binding. This project serves as a pilot for the full-scale mapping of functional regulatory elements in the human genome. The methods and strategies developed will be crucial for informing the human genome. In addition, the body of knowledge gained will be invaluable in the understanding of many human diseases and disorders due to regulatory malfunction of gene regulation. Drosophila is an ideal model system for this project, for its compact genome size, experimental resources, and comparative genomics power, while retaining all the major characteristics of the human genome for the study of gene regulation.



    Past Foundation and Industry Support and Chair Appointments

    We are also supported by the following fondations, faculty appointments, and industry-supported projects
    Pfizer Inc
    2017-2019.
    Translating Genetics into Therapeutic Hypotheses Addressing Immune Traits through Genetic Mechanistic Dissection

    Alfred P. Sloan Foundation Award
    2008-2011, discretionary funds.

    Karl Van Tassel Career Development Chair
    2007-2010, discretionary funds.

    Distinguished Alumnus (1964) Career Development Chair
    2005-2007, Discretionary funds.

    Fellowship support

    Numerous students and postdocs in the group have benefited from fellowship support.
    NSF Postdoctoral Fellowship
    Jason Ernst, 2009-2012 - Full support. Title: Beyond the human genome: integrative analysis of epigenomic datasets.
    Loyal Goff, 2009-2012 - Full support. Title: Small Noncoding RNA Molecules Direct Epigenetic Regulation in Human Embryonic Stem Cells.

    NIH Postdoctoral Fellowship
    Chris Bristow, 2009-2012 - Full support. Title: Analysis and integration of expression patterns in embryonic regulatory networks

    NSF Computing Innovation Fellowship
    Sushmita Roy, 2009-2011 - Full support. Title: Evolution of regulatory networks.

    NSF Graduate Fellowship
    Angela Yen, 2011-2014 - Partial tuition and stipend support.
    Ben Holmes, 2011-2014 - Partial tuition and stipend support.
    Rachel Sealfon, 2010-2013 - Partial tuition and stipend support.
    Jessica Wu, 2008-2011 - Partial tuition and stipend support.
    Pouya Kheradpour, 2005-2008 - Partial tuition and stipend support.

    National Defense Science and Engineering (NDSEG) Graduate Fellowship
    Rachel Sealfon, 2010-2013 - Full tuition and support.

    Siebel Graduate Fellow
    Angela Yen, 2010-2011 - Full tuition and support.

    Miltos Kambourides Graduate Fellowship
    Jessica Yi-Chieh Wu, 2011-2012 - Full tuition and support.

    William H. Chao Fellowship
    Michael F. Lin, Spring 2006, Full support.
    Pouya Kheradpour, Fall 2006 and Spring 2007, Partial tuition and stipend support.

    Human Frontiers Science Program
    Alexander Stark, Full support, Nov 2006-Nov 2009.

    Erwin Schroedinger fellowship
    Stefan Washietl, Long noncoding RNAs in eukaryotic genomes (J 2966-B12), 2010-2012.

    Swiss National Science Foundation Fellowship
    Daniel Marbach, 2009-2010.

    EMBO postdoctoral fellowship
    Alexander Stark, Full support, Jan-Feb 2006.

    EMBO short-term graduate fellowship
    Wouter Meulman, Full support, Summer 2010.

    Schering fellowship
    Alexander Stark, Full support, March-Oct 2006.

    Paris Kanellakis Fellowship
    Georgios Papachristoudis, Fall 2007 and Spring 2008.

    NIH Training Grant
    Matt Rasmussen, 2004-2007, partial support.

    Portugal Fellowship
    Rogerio Candeias, 2007-2010, full support.


    Startup funds

    We have also received generous discretionary funds from the following institutions:
    EECS Department
    Funding for 4 student years and additional discretionary funds.

    Broad Institute of MIT and Harvard
    Discretionary funds.

    Computer Science and Artificial Intelligence Lab
    Discretionary funds.